Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 343
Filter
1.
Ann Clin Transl Neurol ; 8(9): 1809-1816, 2021 09.
Article in English | MEDLINE | ID: mdl-34323022

ABSTRACT

BACKGROUND: Single-center clinical series provide important information on genetic distribution that can guide genetic testing. However, there are few such studies on pediatric populations with inherited peripheral neuropathies (IPNs). METHODS: Thorough genetic testing was performed on IPN patients under 20 years of age from a geographically well-defined Mediterranean area (Valencian Community, Spain), annually assessed with the Charcot-Marie-Tooth disease Pediatric Scale (CMTPedS). RESULTS: From 86 families with IPNs, 99 patients (59 males) were identified, 85 with sensorimotor neuropathy or CMT (2/3 demyelinating form) and 14 with distal hereditary motor neuropathy (dHMN). Genetic diagnosis was achieved in 79.5% families, with a similar mutation detection rate in the demyelinating (88.7%) and axonal (89.5%) forms, significantly higher than in the dHMN families (27.3%). CMT1A was the most common subtype, followed by those carrying heterozygous mutations in either the GDAP1 or GJB1 genes. Mutations in 15 other genes were identified, including a new pathogenic variant in the ATP1A gene. The CMTPedS detected significant disease progression in all genetic subtypes of CMT, at a rate of 1.84 (±3.7) over 1 year (p < 0.0005, n = 62) and a 2-year rate of 3.6 (±4.4: p < 0.0005, n = 45). Significant disease worsening was also detected for CMT1A over 1 (1.7 ± 3.6, p < 0.05) and 2 years (4.2 ± 4.3, p < 0.0005). CONCLUSIONS: This study highlights the unique spectrum of IPN gene frequencies among pediatric patients in this specific geographic region, identifying the CMTPedS as a sensitive tool to detect significant disease worsening over 1 year that could help optimize the design of clinical trials.


Subject(s)
Hereditary Sensory and Motor Neuropathy , Peripheral Nervous System Diseases , Adolescent , Adult , Age of Onset , Charcot-Marie-Tooth Disease/diagnosis , Charcot-Marie-Tooth Disease/epidemiology , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Child , Child, Preschool , Female , Hereditary Sensory and Motor Neuropathy/diagnosis , Hereditary Sensory and Motor Neuropathy/epidemiology , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Longitudinal Studies , Male , Mediterranean Region/epidemiology , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/epidemiology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/physiopathology , Referral and Consultation , Spain/epidemiology , Young Adult
2.
Ann Clin Transl Neurol ; 8(6): 1330-1342, 2021 06.
Article in English | MEDLINE | ID: mdl-33943039

ABSTRACT

BACKGROUND: The expansion of GGC repeat in the 5' untranslated region of the NOTCH2NLC has been associated with various neurogenerative disorders of the central nervous system and, more recently, oculopharyngodistal myopathy. This study aimed to report patients with distal weakness with both neuropathic and myopathic features on electrophysiology and pathology who present GGC repeat expansions in the NOTCH2NLC. METHODS: Whole-exome sequencing (WES) and long-read sequencing were implemented to identify the candidate genes. In addition, the available clinical data and the pathological changes associated with peripheral nerve and muscle biopsies were reviewed and studied. RESULTS: We identified and validated GGC repeat expansions of NOTCH2NLC in three unrelated patients who presented with progressive weakness predominantly affecting distal lower limb muscles, following negative results in an initial WES. We found intranuclear inclusions with multiple proteins deposits in the nuclei of both myofibers and Schwann cells. The clinical features of these patients are compatible with the diagnosis of distal motor neuropathy and rimmed vacuolar myopathy. INTERPRETATION: These phenotypes enrich the class of features associated with NOTCH2NLC-related repeat expansion disorders (NRED), and provide further evidence that the neurological symptoms of NRED include not only brain, spinal cord, and peripheral nerves damage, but also myopathy, and that overlapping symptoms might exist.


Subject(s)
Distal Myopathies/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Adult , Distal Myopathies/pathology , Distal Myopathies/physiopathology , Female , Hereditary Sensory and Motor Neuropathy/pathology , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Intranuclear Inclusion Bodies/genetics , Male , Middle Aged , Pedigree , Phenotype , Trinucleotide Repeat Expansion/genetics , Exome Sequencing
3.
J Peripher Nerv Syst ; 25(4): 423-428, 2020 12.
Article in English | MEDLINE | ID: mdl-33107133

ABSTRACT

Minifascicular neuropathy (MN) is a rare, autosomal recessive disease with prominent structural changes of peripheral nerves. So far, it has been observed in females with a 46,XY karyotype and mutations of the Desert Hedgehog (DHH) gene, thus linking MN to gonadal dysgenesis (GD) and disorders of sex development (DSD). However, a 46,XX proband with normal female sex and gender development underwent clinical evaluations, nerve conduction studies and genetic screening for a severe motor-sensory neuropathy with a pathological phenotype that hinted at MN. Indeed, sural nerve biopsy revealed a profound disturbance of perineurium development with a thin and loose structure. High-resolution ultrasound (HRUS) also disclosed diffuse changes of nerve echotexture that visibly correlated with the pathological features. After extensive genetic testing, a novel homozygous DHH null mutation (p.Ser185*) was identified in the proband and in her sister, who was affected by a similar motor-sensory neuropathy, but was eventually found to be a 46,XY patient according to a late diagnosis of DSD with complete GD. DHH should therefore be considered as a possible cause of rare non-syndromic hereditary motor-sensory neuropathies, regardless of DSD. Furthermore, HRUS could effectively smooth the complex diagnostic workup as it demonstrated a high predictive power to detect MN, providing the same detailed correlations to the pathologic features of the nerve biopsy and Dhh-/- mice in both sisters. Hence, HRUS may assume a pivotal role in guiding molecular analysis in individuals with or without DSD.


Subject(s)
Disorder of Sex Development, 46,XY/diagnosis , Hedgehog Proteins/genetics , Hereditary Sensory and Motor Neuropathy , Consanguinity , Female , Genetic Testing , Hereditary Sensory and Motor Neuropathy/diagnosis , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Microscopy, Acoustic , Middle Aged , Siblings , Sural Nerve/pathology , Syndrome
4.
Ann Clin Transl Neurol ; 7(8): 1400-1409, 2020 08.
Article in English | MEDLINE | ID: mdl-32856791

ABSTRACT

OBJECTIVE: Hereditary Neuropathy with Liability to Pressure Palsies (HNPP) is caused by a heterozygous deletion of peripheral myelin protein-22 (PMP22) gene resulting in focal sensorimotor deficits. Our lab has identified a disruption of myelin junctions in excessively permeable myelin that impairs action potential propagation. This mechanism is expected to cause fatigue in patients with HNPP. Therefore, the objective was to characterize fatigue in patients with HNPP and determine the relationship of fatigue to nerve pathology, disability, and quality of life. METHODS: Nine females with HNPP participated in a single visit that included genotyping, nerve conduction studies, neurological exam, quantitative magnetic resonance imaging, and a physical therapy exam incorporating upper and lower extremity function and survey measures of fatigue. This visit was followed by 2 weeks of ecological momentary assessment (wrist-worn device) that captured fatigue ratings five times per day. RESULTS: Participants demonstrated mild neurological impairment (CMTNS: 5.7 ± 2.8), yet reported high fatigue levels (average fatigue intensity over 2 weeks: 5.9 out of 10). Higher fatigue levels were associated with poorer quality of life and more pain. Higher fatigue was associated with significantly greater distal nerve proton density changes on peripheral nerve MRI, which is in line with hyper-permeable myelin in HNPP. INTERPRETATION: Fatigue is common and severe among patients with HNPP whose disabilities are minimal by conventional outcome measures. Therapeutic interventions targeting fatigue have the potential to improve quality of life and may serve as a robust outcome measure to show longitudinal changes for patients with HNPP.


Subject(s)
Arthrogryposis/complications , Arthrogryposis/diagnosis , Fatigue/diagnosis , Fatigue/etiology , Hereditary Sensory and Motor Neuropathy/complications , Hereditary Sensory and Motor Neuropathy/diagnosis , Adult , Arthrogryposis/physiopathology , Ecological Momentary Assessment , Fatigue/physiopathology , Female , Genotype , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Magnetic Resonance Imaging , Neural Conduction/physiology , Neurologic Examination
5.
J Neuromuscul Dis ; 7(4): 505-510, 2020.
Article in English | MEDLINE | ID: mdl-32538861

ABSTRACT

Hereditary neuropathy with liability to pressure palsies (HNPP) is an autosomal dominant inherited disorder commonly presenting with acute-onset, non-painful focal sensory and motor mono neuropathy. In 80% of cases, the genetic defect is a 1.5 Mb deletion on chromosome 17p11.2, including PMP22. Only few cases of partial deletion and point mutations in PMP22 are involved in HNPP. We investigated a 62-years-old man with lower limb plexopathy first considered as Garland's syndrome. A month later, his 29 years old son also consulted for paresthesia on the peroneal nerve.Targeted sequencing of the PMP22 gene identified a c.370delT (p.Trp124Glyfs*31) in both affected patients.We report a new PMP22 point mutation associated with an atypical clinical phenotype of HNPP, a painful plexopathy of the lower limb worsenen by diabetes and a mere paresthesia, but a typical ENMG. This study illustrates the large spectrum of the disease, and emphasizes the importance of a complete ENMG and family history.


Subject(s)
Arthrogryposis/genetics , Arthrogryposis/physiopathology , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Myelin Proteins/genetics , Adult , Arthrogryposis/diagnosis , Hereditary Sensory and Motor Neuropathy/diagnosis , Humans , Male , Middle Aged , Pedigree , Phenotype , Point Mutation
7.
Neuromuscul Disord ; 30(3): 227-231, 2020 03.
Article in English | MEDLINE | ID: mdl-32085962

ABSTRACT

The Tropomyosin-receptor kinase fused gene (TFG) encodes TFG which is expressed in spinal motor neurons, dorsal root ganglia and cranial nerve nuclei, and plays a role in the dynamics of the endoplasmic reticulum. Two dominant missense TFG mutations have previously been reported in limited geographical areas (Far East, Iran, China) in association with hereditary motor sensory neuropathy with proximal involvement (HMSN-P) of the four limbs, or with Charcot-Marie-Tooth disease type 2 (CMT2). The 60-year-old female proband belonging to a three-generation Italian family presented with an atypical neuropathy characterized by diffuse painful cramps and prominent motor-sensory impairment of the distal upper limbs. Her sural nerve biopsy showed chronic axonal neuropathy without active degeneration or regeneration. Targeted next-generation sequencing of a panel with 98 genes associated with inherited peripheral neuropathies/neuromuscular disorders identified three candidate genes: TFG, DHTKD1 and DCTN2. In the family, the disease co-segregated with the TFG p.(Gly269Val) variant. TFG should be considered in genetic testing of patients with heterogeneous inherited neuropathy, independently of their ethnic origin.


Subject(s)
Hereditary Sensory and Motor Neuropathy , Proteins/genetics , Upper Extremity/physiopathology , Female , Hereditary Sensory and Motor Neuropathy/diagnosis , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , Hereditary Sensory and Motor Neuropathy/physiopathology , High-Throughput Nucleotide Sequencing , Humans , Middle Aged , Pedigree
8.
J Peripher Nerv Syst ; 25(1): 19-26, 2020 03.
Article in English | MEDLINE | ID: mdl-31919945

ABSTRACT

Hereditary neuropathies may be misdiagnosed with chronic inflammatory demyelinating polyradiculoneuropathy (CIDP). A correct diagnosis is crucial for avoiding unnecessary therapies and access genetic counseling. We report on nine patients (seven men, mean age 49.2 ± 16.1) diagnosed with and treated as CIDP, in whom mutations or variants of unknown significance (VUS) in genes associated with hereditary neuropathies were reported. All underwent neurological and neurophysiological examination, eight also cerebrospinal fluid (CSF) analysis. In 4/9, nerve ultrasound and/or MR-neurography were performed. All the patients complained of progressive upper or lower limbs sensory-motor symptoms, with heterogeneous disease duration (1-34 years, mean 8.6 ± 10.8). Neurophysiology showed demyelinating signs in seven patients, mixed findings with predominant axonal damage in two patients. Neuroimaging disclosed diffuse abnormalities at proximal and distal segments. Molecular screening showed PMP22 duplication in two patients, mutations in the MPZ, EGR2, and GJB1 genes were reported in each of the remaining patients. The two patients with mixed neurophysiological findings had p.Val30Met mutation in the transthyretin gene. Two patients had VUS in the MARS and HSPB1 genes. Four patients had partial response to immunomodulant therapies, and CSF and neurophysiological features suggesting an inflammatory condition concomitant with the hereditary neuropathy. Hereditary neuropathy may be misdiagnosed with CIDP. The most common pitfalls are CSF (high protein levels and oligoclonal bands), incorrect interpretation of neurophysiology, and transient benefit from therapies. Neuroimaging may be helpful in cases with atypical presentations or when severe axonal damage complicate the neurophysiological interpretation.


Subject(s)
Diagnostic Errors , Hereditary Sensory and Motor Neuropathy/diagnosis , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnosis , Adult , Aged , Female , Hereditary Sensory and Motor Neuropathy/cerebrospinal fluid , Hereditary Sensory and Motor Neuropathy/drug therapy , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Immunologic Factors/pharmacology , Magnetic Resonance Imaging , Male , Middle Aged , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/cerebrospinal fluid , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/drug therapy , Practice Guidelines as Topic , Ultrasonography
9.
Neuropediatrics ; 51(3): 173-177, 2020 06.
Article in English | MEDLINE | ID: mdl-31784971

ABSTRACT

Heterozygous deletions of the gene PMP22 are associated to hereditary neuropathy with liability to pressure palsies (HNPP), a demyelinating neuromuscular disease causing variable transitory focal muscles weakness. Deletions involving both copies of PMP22 cause more severe phenotypes, with early-onset neuropathy and impairment in motor development. We report a patient with a severe early-onset demyelinating neuropathy, caused by two different inherited deletions of PMP22, whose parents had an HNPP. The patient showed neurological signs and delay in motor development but normal intellective abilities. A motor and sensitive conduction study showed severe signs of demyelination, suggestive for Dejerine Sottas Syndrome (DSS). The patient's father had a typical HNPP caused by a heterozygous 17p11.2 deletion, encompassing PMP22. The patient's mother reported no neuropathic symptoms, but in a nerve conduction studies, parents and several relatives showed signs of sensory-motor deficit with focal slowing of conduction at common sites of entrapment. Quantitative analysis of PMP22, performed in our patient by multiplex ligation-dependent probe amplification, revealed a compound heterozygous status with the same deletion of the father and a deletion of PMP22 exon 5, after proved to be inherited from the mother. Therefore, when we face an early-onset, severe form of neuropathy, we have to consider rare forms of hereditary neuropathy caused by homozygous or compound heterozygous mutations in PMP22, even if parents are asymptomatic; an exhaustive family history and an electrodiagnostic study are essential to guide genetic tests and to make a diagnosis.


Subject(s)
Arthrogryposis , Hereditary Sensory and Motor Neuropathy , Myelin Proteins/genetics , Arthrogryposis/diagnosis , Arthrogryposis/genetics , Arthrogryposis/physiopathology , Child, Preschool , Female , Gene Deletion , Hereditary Sensory and Motor Neuropathy/diagnosis , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Neural Conduction/physiology , Pedigree
10.
J Neurol Sci ; 409: 116629, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31862515

ABSTRACT

BACKGROUND: Hereditary neuropathy with liability to pressure palsies (HNPP) is a rare neuropathy with a heterogeneous clinical profile. Painless recurrent palsies are the usual presentation, but neuropathic pain could be predominant or inaugural. Browsing the medical literature, we only found two articles reffering to this important clinical feature. Whether there are differences between patients with or without pain is unclear. The main objective of this study was to compare the clinical and electrophysiological features of these patients and to evaluate the impact on their disability. METHODS: All patients diagnosed with HNPP at the Limoges University Hospital Centre were included and separated into two groups according to the presence or absence of neuropathic pain. In each case, the clinical, genetic, electrodiagnostic, therapeutic features and the modified Rankin Scale (mRS) were evaluated. RESULTS: Out of 23 patients, 52% presented with neuropathic pain. There was no difference between groups regarding to clinical and electrophysiological features, except for the amplitude of the ulnar sensory nerve (p < 0,003). The amplitudes of sensory nerve action potentials (SNAPs) seemed to be higher in patients with pain, but were below the lower limit of normal. Patients with pain had a higher mRS than patients without pain (p < 0,007). CONCLUSION: This study supports previous published results and highlights a trend for higher sensory amplitudes in HNPP patients with pain. We found a prevalence of neuropathic pain of 52% in patients with HNPP, underlining the need to systematically assess pain in such patients in order to improve their management.


Subject(s)
Arthrogryposis/diagnosis , Arthrogryposis/physiopathology , Electrophysiological Phenomena/physiology , Hereditary Sensory and Motor Neuropathy/diagnosis , Hereditary Sensory and Motor Neuropathy/physiopathology , Pain/diagnosis , Pain/physiopathology , Adolescent , Adult , Aged , Arthrogryposis/epidemiology , Child , Child, Preschool , Cohort Studies , Female , Hereditary Sensory and Motor Neuropathy/epidemiology , Humans , Male , Middle Aged , Pain/epidemiology , Retrospective Studies , Young Adult
11.
J Neurol Neurosurg Psychiatry ; 91(1): 49-57, 2020 01.
Article in English | MEDLINE | ID: mdl-31690697

ABSTRACT

OBJECTIVE: Sensory neuropathies (SNs) are often classified as idiopathic even if immunological mechanisms can be suspected. Antibodies against the intracellular domain of the fibroblast growth factor receptor 3 (FGFR3) possibly identify a subgroup of SN affecting mostly the dorsal root ganglion (DRG). The aim of this study was to identify the frequency of anti-FGFR3 antibodies and the associated clinical pattern in a large cohort of patients with SN. METHODS: A prospective, multicentric, European and Brazilian study included adults with pure SN. Serum anti-FGRF3 antibodies were analysed by ELISA. Detailed clinical and paraclinical data were collected for each anti-FGFR3-positive patient and as control for anti-FGFR3-negative patients from the same centres ('center-matched'). RESULTS: Sixty-five patients out of 426 (15%) had anti-FGFR3 antibodies, which were the only identified autoimmune markers in 43 patients (66%). The neuropathy was non-length dependent in 89% and classified as sensory neuronopathy in 64%, non-length-dependent small fibre neuropathy in 17% and other neuropathy in 19%. Specific clinical features occurred after 5-6 years of evolution including frequent paresthesia, predominant clinical and electrophysiological involvement of the lower limbs, and a less frequent mixed large and small fibre involvement. Brazilians had a higher frequency of anti-FGFR3 antibodies than Europeans (36% vs 13%, p<0.001), and a more frequent asymmetrical distribution of symptoms (OR 169, 95% CI 3.4 to 8424). CONCLUSIONS: Anti-FGFR3 antibodies occur in a subgroup of SN probably predominantly affecting the DRG. Differences between Europeans and Brazilians could suggest involvement of genetic or environmental factors.


Subject(s)
Autoantibodies/immunology , Hereditary Sensory and Motor Neuropathy/immunology , Receptor, Fibroblast Growth Factor, Type 3/immunology , Adult , Autoantibodies/analysis , Brazil , Cohort Studies , Electrodiagnosis , Europe , Female , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Male , Middle Aged , Nerve Fibers/pathology , Prospective Studies
12.
Sci Rep ; 9(1): 19336, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31852952

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.


Subject(s)
Early Growth Response Protein 2/genetics , Genetic Predisposition to Disease , Hereditary Sensory and Motor Neuropathy/genetics , Mutation/genetics , Adolescent , Adult , Age of Onset , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , Computer Simulation , Early Growth Response Protein 2/chemistry , Female , Hereditary Sensory and Motor Neuropathy/diagnostic imaging , Hereditary Sensory and Motor Neuropathy/pathology , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neural Conduction , Pedigree , Protein Domains , Schwann Cells/metabolism , Transcription, Genetic , Transcriptional Activation/genetics , Exome Sequencing
13.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(11): 1136-1140, 2019 Nov 10.
Article in Chinese | MEDLINE | ID: mdl-31703145

ABSTRACT

Distal hereditary motor neuropathy (dHMN) is a group of clinically and genetically heterogeneous disorders characterized by progressive distal weakness and atrophy. The onset of dHMN is at mid-adulthood or early childhood, and the symptoms are mainly present in the lower limbs. Besides weakness and atrophy of distal limb muscles, some patients may develop bulbar paralysis, and some may also present with mild sensory disturbance. Decreased or absent tendon reflexes may be discovered. Electromyography may show neurogenic damages. Muscular biopsy may reveal neurogenic amyotrophy. An increasing number of genes have been associated with dHMN. Pathogenesis of dHMN may include formation of protein aggregates, impairment of autophagy pathway, RNA processing, translation synthesis, axonal transport, endoplasmic reticulum stress, calcium channel and neuroprotection. A review for recent progress made on clinical characterization and molecular genetics of dHMN is provided.


Subject(s)
Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Genetic Research , Humans
14.
J Peripher Nerv Syst ; 24(4): 348-353, 2019 12.
Article in English | MEDLINE | ID: mdl-31523922

ABSTRACT

Mutations in apoptosis-inducing factor mitochondrion-associated-1 (AIFM1) cause X-linked peripheral neuropathy (Cowchock syndrome, CMT4X); however, more recently a cerebellar presentation has been described. We describe a large Irish family with seven affected males. They presented with a variable age of onset, 18 months to 39 years of age. All developed variably present sensorineural deafness, peripheral neuropathy, cerebellar ataxia, and pyramidal involvement. In addition, three had colour vision deficiency. Scale for the assessment and rating of ataxia ranged 2 to 23/40, while Charcot-Marie-Tooth neuropathy score 2 varied between 7 and 13/36. All individuals had normal cognitive assessment. Neurophysiology demonstrated length-dependent large-fibre sensorimotor axonal neuropathy, with particular involvement of superficial radial sensory responses. Brain imaging, performed in four, revealed varying extent of cerebellar atrophy, and white matter changes in one. Optical coherence tomography was abnormal in one, who had unrelated eye pathology. Four obligate female carriers were assessed clinically, two of them neurophysiologically; all were unaffected. Whole genome sequencing demonstrated a previously reported hemizygous AIFM1 mutation. Analysis for mutations in other genes associated with colour deficiency was negative. AIFM1-associated phenotype in this family demonstrated significant variability. To our knowledge, this is the first report of AIFM1-associated colour blindness. Superficial radial nerve was particularly affected neurophysiologically, which could represent a phenotypic marker towards this specific genetic diagnosis.


Subject(s)
Apoptosis Inducing Factor/genetics , Cerebellar Ataxia , Color Vision Defects , Hearing Loss, Sensorineural , Hereditary Sensory and Motor Neuropathy , Adult , Cerebellar Ataxia/etiology , Cerebellar Ataxia/genetics , Cerebellar Ataxia/physiopathology , Color Vision Defects/etiology , Color Vision Defects/genetics , Color Vision Defects/physiopathology , Hearing Loss, Sensorineural/etiology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/physiopathology , Hereditary Sensory and Motor Neuropathy/complications , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Ireland , Male , Middle Aged , Pedigree , Severity of Illness Index
15.
Muscle Nerve ; 60(6): 739-744, 2019 12.
Article in English | MEDLINE | ID: mdl-31449671

ABSTRACT

BACKGROUND: Hereditary motor and sensory neuropathy with proximal dominant involvement (HMSN-P) is characterized by adult onset, a slowly progressive course and autosomal dominant inheritance. It remains unclear whether myopathic changes occur histopathologically. METHODS: We encountered 2 patients in a family with a heterozygous p.P285L mutation in TRK-fused gene (TFG), which is known to cause HMSN-P. The affected individuals developed proximal-dominant muscle weakness in their 40s, which slowly progressed to a motor neuron disease-like phenotype. RESULTS: Muscle biopsy showed myopathic pathology including fiber size variability, increased internal nuclei, fiber splitting, and core-like structures, associated with neurogenic changes: large groups of atrophic fibers and fiber type-grouping. Immunohistochemistry revealed sarcoplasmic aggregates of TFG, TDP-43, and p62 without congophilic material. CONCLUSIONS: The present study demonstrates myopathic changes in HMSN-P. Although the mechanisms underlying the skeletal muscle involvement remain to be elucidated, immunohistochemistry suggests that abnormal protein aggregation may be involved in the myopathic pathology.


Subject(s)
Hereditary Sensory and Motor Neuropathy/pathology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Action Potentials , DNA-Binding Proteins/metabolism , Female , Fluorescent Antibody Technique , Hereditary Sensory and Motor Neuropathy/diagnostic imaging , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle, Skeletal/diagnostic imaging , Muscular Atrophy/pathology , Neural Conduction , Pedigree , Proteins/genetics , RNA-Binding Proteins/metabolism , Sarcoplasmic Reticulum/metabolism , Siblings
17.
Hand (N Y) ; 14(3): 333-338, 2019 05.
Article in English | MEDLINE | ID: mdl-29058949

ABSTRACT

BACKGROUND: Lipomas are common benign tumors. When they develop in proximity to peripheral nerves, they can cause neurologic symptoms secondary to mass effect. Previous reports have shown symptom resolution after removal of lipomas compressing various upper extremity peripheral nerves. However, brachial plexus lipomas are relatively rare. Our multidisciplinary experience with brachial plexus lipoma resection is reviewed in the largest case series to date. METHODS: A retrospective chart review of all patients undergoing resection of brachial plexus lipomatous tumors between 2006 and 2016 was performed. Patient demographic data, diagnostic imaging, clinical presentation, operative details, surgical pathology, and clinical outcomes were reviewed. RESULTS: Twelve brachial plexus lipomatous tumors were resected in 11 patients: 10 lipomas, 1 hibernoma, and 1 atypical lipomatous tumor. The most common tumor location was supraclavicular (50%), followed by axillary (42%), and proximal medial arm (8%). The most common brachial plexus segment involved was the upper trunk (50%), followed by posterior cord (25%), lateral pectoral nerve (8%), lower trunk (8%), and proximal median nerve (8%). Most patients presented with an enlarging painless mass (58%). Of the patients who presented with neurologic symptoms, symptoms resolved in the majority (80%). CONCLUSIONS: Brachial plexus lipomas are rare causes of compression neuropathy in the upper extremity. Careful resection and knowledge of brachial plexus anatomy, which may be distorted by the tumor, are critical to achieving a successful surgical outcome with predictable symptom resolution. Finally, surveillance magnetic resonance imaging may be warranted for atypical lesions.


Subject(s)
Brachial Plexus/pathology , Lipoma/surgery , Upper Extremity/pathology , Adult , Arthrogryposis/etiology , Arthrogryposis/physiopathology , Brachial Plexus/anatomy & histology , Female , Hereditary Sensory and Motor Neuropathy/etiology , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Lipoma/diagnostic imaging , Magnetic Resonance Imaging/standards , Male , Middle Aged , Retrospective Studies , Treatment Outcome , Upper Extremity/innervation
18.
Hand (N Y) ; 14(4): 477-482, 2019 07.
Article in English | MEDLINE | ID: mdl-29582694

ABSTRACT

Background: Cubital tunnel syndrome is a common compressive neuropathy of the upper extremity. The anconeus epitrochlearis muscle is an unusual but occasional contributor. We review our experience with this anomalous muscle in elbows with cubital tunnel syndrome. Methods: We retrospectively reviewed charts of 13 patients noted to have an anconeus epitrochlearis muscle associated with cubital tunnel syndrome. Results: Ten patients had unilateral ulnar neuropathy supported by nerve conduction studies. Three had bilateral cubital tunnel syndrome symptoms with 1 of those having normal nerve conduction studies for both elbows. Eight elbows were treated with myotomy of the anconeus epitrochlearis muscle and submuscular transposition of the ulnar nerve. The other 8 elbows were treated with myotomy of the anconeus epitrochlearis muscle and in situ decompression of the ulnar nerve only. All but 1 patient had either clinical resolution or improvement of symptoms at follow-up ranging from 2 weeks to 1 year after surgery. The 1 patient who had persistent symptoms had received myotomy and in situ decompression of the ulnar nerve only. Conclusions: An anomalous anconeus epitrochlearis occasionally results in compression of the ulnar nerve but is usually an incidental finding. Its contribution to compression neuropathy can be tested intraoperatively by passively ranging the elbow while observing the change in vector and tension of its muscle fibers over the ulnar nerve. Regardless of findings, we recommend myotomy of the muscle and in situ decompression of the ulnar nerve. Submuscular transposition of the ulnar nerve may be necessary if there is subluxation.


Subject(s)
Cubital Tunnel Syndrome/surgery , Decompression, Surgical/methods , Elbow Joint/surgery , Muscle, Skeletal/surgery , Adult , Aftercare , Aged , Arthrogryposis/physiopathology , Cubital Tunnel Syndrome/diagnosis , Female , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Male , Middle Aged , Muscle, Skeletal/abnormalities , Myotomy , Neural Conduction/physiology , Retrospective Studies , Treatment Outcome , Ulnar Nerve/surgery , Ulnar Neuropathies/diagnosis , Ulnar Neuropathies/physiopathology
19.
Hum Genet ; 137(11-12): 911-919, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30460542

ABSTRACT

Mutations in the SACS gene have been initially reported in a rare autosomal recessive cerebellar ataxia syndrome featuring prominent cerebellar atrophy, spasticity and peripheral neuropathy as well as retinal abnormalities in some cases (autosomal recessive spastic ataxia of Charlevoix-Saguenay, ARSACS). In the past few years, the phenotypic spectrum has broadened, mainly owing to the availability and application of high-throughput genetic testing methods. We identified nine patients (three sib pairs, three singleton cases) with isolated, non-syndromic hereditary motor and sensory neuropathy (HMSN) who carried pathogenic SACS mutations, either in the homozygous or compound heterozygous state. None of the patients displayed spasticity or pyramidal signs. Ataxia, which was noted in only three patients, was consistent with a sensory ataxia. Nerve conduction and nerve biopsy studies showed mixed demyelinating and axonal neuropathy. Brain MRI scans were either normal or revealed isolated upper vermis atrophy of the cerebellum. Our findings confirm the broad clinical spectrum associated with SACS mutations, including pure polyneuropathy without characteristic clinical and brain imaging manifestations of ARSACS.


Subject(s)
Ataxia/genetics , Genes, Recessive/genetics , Heat-Shock Proteins/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Ataxia/physiopathology , Cerebellum/physiopathology , Female , Hereditary Sensory and Motor Neuropathy/physiopathology , Homozygote , Humans , Magnetic Resonance Imaging , Male , Mutation , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...