Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
2.
Brain ; 146(12): 5235-5248, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37503746

ABSTRACT

The extracellular matrix (ECM) has an important role in the development and maintenance of skeletal muscle, and several muscle diseases are associated with the dysfunction of ECM elements. MAMDC2 is a putative ECM protein and its role in cell proliferation has been investigated in certain cancer types. However, its participation in skeletal muscle physiology has not been previously studied. We describe 17 individuals with an autosomal dominant muscular dystrophy belonging to two unrelated families in which different heterozygous truncating variants in the last exon of MAMDC2 co-segregate correctly with the disease. The radiological aspect of muscle involvement resembles that of COL6 myopathies with fat replacement at the peripheral rim of vastii muscles. In this cohort, a subfascial and peri-tendinous pattern is observed in upper and lower limb muscles. Here we show that MAMDC2 is expressed in adult skeletal muscle and differentiating muscle cells, where it appears to localize to the sarcoplasm and myonuclei. In addition, we show it is secreted by myoblasts and differentiating myotubes into to the extracellular compartment. The last exon encodes a disordered region with a polar residue compositional bias loss of which likely induces a toxic effect of the mutant protein. The precise mechanisms by which the altered MAMDC2 proteins cause disease remains to be determined. MAMDC2 is a skeletal muscle disease-associated protein. Its role in muscle development and ECM-muscle communication remains to be fully elucidated. Screening of the last exon of MAMDC2 should be considered in patients presenting with autosomal dominant muscular dystrophy, particularly in those with a subfascial radiological pattern of muscle involvement.


Subject(s)
Muscular Dystrophies , Adult , Humans , Muscular Dystrophies/genetics , Muscle, Skeletal/metabolism , Extracellular Matrix Proteins
3.
Hum Mol Genet ; 32(7): 1127-1136, 2023 03 20.
Article in English | MEDLINE | ID: mdl-36322148

ABSTRACT

Nemaline myopathy 8 (NEM8) is typically a severe autosomal recessive disorder associated with variants in the kelch-like family member 40 gene (KLHL40). Common features include fetal akinesia, fractures, contractures, dysphagia, respiratory failure and neonatal death. Here, we describe a 26-year-old man with relatively mild NEM8. He presented with hypotonia and bilateral femur fractures at birth, later developing bilateral Achilles' contractures, scoliosis, and elbow and knee contractures. He had walking difficulties throughout childhood and became wheelchair bound from age 13 after prolonged immobilization. Muscle magnetic resonance imaging at age 13 indicated prominent fat replacement in his pelvic girdle, posterior compartments of thighs and vastus intermedius. Muscle biopsy revealed nemaline bodies and intranuclear rods. RNA sequencing and western blotting of patient skeletal muscle indicated significant reduction in KLHL40 mRNA and protein, respectively. Using gene panel screening, exome sequencing and RNA sequencing, we identified compound heterozygous variants in KLHL40; a truncating 10.9 kb deletion in trans with a likely pathogenic variant (c.*152G > T) in the 3' untranslated region (UTR). Computational tools SpliceAI and Introme predicted the c.*152G > T variant created a cryptic donor splice site. RNA-seq and in vitro analyses indicated that the c.*152G > T variant induces multiple de novo splicing events that likely provoke nonsense mediated decay of KLHL40 mRNA explaining the loss of mRNA expression and protein abundance in the patient. Analysis of 3' UTR variants in ClinVar suggests variants that introduce aberrant 3' UTR splicing may be underrecognized in Mendelian disease. We encourage consideration of this mechanism during variant curation.


Subject(s)
Contracture , Myopathies, Nemaline , Male , Infant, Newborn , Humans , Child , Adolescent , Adult , Myopathies, Nemaline/genetics , 3' Untranslated Regions/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , RNA Splice Sites/genetics , RNA, Messenger , Contracture/genetics , Mutation
4.
Curr Opin Neurol ; 35(5): 651-657, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35942668

ABSTRACT

PURPOSE OF REVIEW: This review summarizes recent advances in our understanding of the genetics of rhabdomyolysis. RECENT FINDINGS: Rhabdomyolysis is the acute breakdown of myofibres resulting in systemic changes that can be life-threatening. Environmental triggers, including trauma, exercise, toxins and infections, and/or gene defects can precipitate rhabdomyolysis. A schema (aptly titled RHABDO) has been suggested for evaluating whether a patient with rhabdomyolysis is likely to harbour an underlying genetic defect. It is becoming increasingly recognized that defects in muscular dystrophy and myopathy genes can trigger rhabdomyolysis, even as the sole or presenting feature. Variants in genes not previously associated with human disease have been identified recently as causative of rhabdomyolysis, MLIP , MYH1 and OBSCN . Our understanding of the pathomechanisms contributing to rhabdomyolysis have also improved with an increased awareness of the role of mitochondrial dysfunction in LPIN1 , FDX2 , ISCU and TANGO2 -mediated disease. SUMMARY: An accurate genetic diagnosis is important for optimal clinical management of the patient, avoiding associated triggers and genetic counselling and cascade screening. Despite recent advances in our understanding of the genetics contributing to rhabdomyolysis, many patients remain without an accurate genetic diagnosis, suggesting there are many more causative genes, variants and disease mechanisms to uncover.


Subject(s)
Muscular Diseases , Muscular Dystrophies , Rhabdomyolysis , Exercise , Humans , Muscular Diseases/genetics , Muscular Dystrophies/complications , Phosphatidate Phosphatase/genetics , Rhabdomyolysis/complications , Rhabdomyolysis/diagnosis , Rhabdomyolysis/genetics
5.
Article in English | MEDLINE | ID: mdl-34728497

ABSTRACT

BACKGROUND AND OBJECTIVES: To study the clinical and laboratory features of antineurofascin-155 (NF155)-positive autoimmune nodopathy (AN). METHODS: Patients with anti-NF155 antibodies detected on routine immunologic testing were included. Clinical characteristics, treatment response, and functional scales (modified Rankin Scale [mRS] and Inflammatory Rasch-built Overall Disability Scale [I-RODS]) were retrospectively collected at baseline and at the follow-up. Autoantibody and neurofilament light (NfL) chain levels were analyzed at baseline and at the follow-up. RESULTS: Forty NF155+ patients with AN were included. Mean age at onset was 42.4 years. Patients presented with a progressive (75%), sensory motor (87.5%), and symmetric distal-predominant weakness in upper (97.2%) and lower extremities (94.5%), with tremor and ataxia (75%). Patients received a median of 3 (2-4) different treatments in 46 months of median follow-up. Response to IV immunoglobulin (86.8%) or steroids (72.2%) was poor in most patients, whereas 77.3% responded to rituximab. HLA-DRB1*15 was detected in 91.3% of patients. IgG4 anti-NF155 antibodies were predominant in all patients; anti-NF155 titers correlated with mRS within the same patient (r = 0.41, p = 0.004). Serum NfL (sNfL) levels were higher in anti-NF155+ AN than in healthy controls (36.47 vs 7.56 pg/mL, p < 0.001) and correlated with anti-NF155 titers (r = 0.43, p = 0.001), with I-RODS at baseline (r = -0.88, p < 0.001) and with maximum I-RODS achieved (r = -0.58, p = 0.01). Anti-NF155 titers and sNfL levels decreased in all rituximab-treated patients. DISCUSSION: Anti-NF155 AN presents a distinct clinical profile and good response to rituximab. Autoantibody titers and sNfL are useful to monitor disease status in these patients. The use of untagged-NF155 plasmids minimizes the detection of false anti-NF155+ cases. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that anti-NF155 antibodies associate with a specific phenotype and response to rituximab.


Subject(s)
Autoantibodies/blood , Autoimmune Diseases of the Nervous System , Cell Adhesion Molecules/immunology , Immunologic Factors/pharmacology , Nerve Growth Factors/immunology , Ranvier's Nodes/immunology , Rituximab/pharmacology , Adult , Aged , Autoimmune Diseases of the Nervous System/blood , Autoimmune Diseases of the Nervous System/drug therapy , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/physiopathology , Female , Humans , Male , Middle Aged , Retrospective Studies , Young Adult
6.
Brain ; 145(11): 3985-3998, 2022 11 21.
Article in English | MEDLINE | ID: mdl-34957489

ABSTRACT

Rhabdomyolysis is the acute breakdown of skeletal myofibres in response to an initiating factor, most commonly toxins and over exertion. A variety of genetic disorders predispose to rhabdomyolysis through different pathogenic mechanisms, particularly in patients with recurrent episodes. However, most cases remain without a genetic diagnosis. Here we present six patients who presented with severe and recurrent rhabdomyolysis, usually with onset in the teenage years; other features included a history of myalgia and muscle cramps. We identified 10 bi-allelic loss-of-function variants in the gene encoding obscurin (OBSCN) predisposing individuals to recurrent rhabdomyolysis. We show reduced expression of OBSCN and loss of obscurin protein in patient muscle. Obscurin is proposed to be involved in sarcoplasmic reticulum function and Ca2+ handling. Patient cultured myoblasts appear more susceptible to starvation as evidenced by a greater decreased in sarcoplasmic reticulum Ca2+ content compared to control myoblasts. This likely reflects a lower efficiency when pumping Ca2+ back into the sarcoplasmic reticulum and/or a decrease in Ca2+ sarcoplasmic reticulum storage ability when metabolism is diminished. OSBCN variants have previously been associated with cardiomyopathies. None of the patients presented with a cardiomyopathy and cardiac examinations were normal in all cases in which cardiac function was assessed. There was also no history of cardiomyopathy in first degree relatives, in particular in any of the carrier parents. This cohort is relatively young, thus follow-up studies and the identification of additional cases with bi-allelic null OBSCN variants will further delineate OBSCN-related disease and the clinical course of disease.


Subject(s)
Calcium , Rhabdomyolysis , Adolescent , Humans , Rhabdomyolysis/genetics , Rhabdomyolysis/diagnosis , Rhabdomyolysis/pathology , Myalgia/genetics , Sarcoplasmic Reticulum/metabolism , Loss of Heterozygosity , Protein Serine-Threonine Kinases , Rho Guanine Nucleotide Exchange Factors/genetics
8.
Clin Genet ; 100(1): 106-110, 2021 07.
Article in English | MEDLINE | ID: mdl-33818761

ABSTRACT

The pathogenic role of intronic variants is generally difficult to assess, except for those near known splice sites for which aberrant splicing is suspected, although deeper intronic variants can also alter splicing. We have identified a novel (NM_213599.2:c.1180+6T>C) ANO5 variant that causes the exclusion of exon 12. The mutation, identified in a Roma individual, has an estimated carrier rate of 1.68% among the Iberian Roma population, this being the first ANO5 pathogenic variant communicated in this ethnic group. In this study, we have also characterized the ANO5 splice forms expressed in human muscle with the detection of an alternative transcript, in which exons 8 and 9 are spliced out.


Subject(s)
Anoctamins/genetics , Introns/genetics , Muscular Dystrophies/genetics , RNA Splicing/genetics , Roma/genetics , Exons/genetics , Female , Humans , Middle Aged , Mutation/genetics , RNA Splice Sites/genetics
9.
J Med Genet ; 57(12): 835-842, 2020 12.
Article in English | MEDLINE | ID: mdl-32179706

ABSTRACT

BACKGROUND: UBA5 is the activating enzyme of UFM1 in the ufmylation post-translational modification system. Different neurological phenotypes have been associated with UBA5 pathogenic variants including epilepsy, intellectual disability, movement disorders and ataxia. METHODS AND RESULTS: We describe a large multigenerational consanguineous family presenting with a severe congenital neuropathy causing early death in infancy. Whole exome sequencing and linkage analysis identified a novel homozygous UBA5 NM_024818.3 c.31C>T (p.Arg11Trp) mutation. Protein expression assays in mouse tissue showed similar levels of UBA5 in peripheral nerves to the central nervous system. CRISPR-Cas9 edited HEK (human embrionic kidney) cells homozygous for the UBA5 p.Arg11Trp mutation showed reduced levels of UBA5 protein compared with the wild-type. The mutant p.Arg11Trp UBA5 protein shows reduced ability to activate UFM1. CONCLUSION: This report expands the phenotypical spectrum of UBA5 mutations to include fatal peripheral neuropathy.


Subject(s)
CRISPR-Cas Systems/genetics , Intellectual Disability/genetics , Nervous System Malformations/genetics , Proteins/genetics , Ubiquitin-Activating Enzymes/genetics , Ataxia/genetics , Ataxia/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Consanguinity , Epilepsy/genetics , Epilepsy/pathology , Female , Gene Expression Regulation/genetics , Genetic Linkage , HEK293 Cells , Homozygote , Humans , Infant , Intellectual Disability/pathology , Male , Movement Disorders/genetics , Movement Disorders/pathology , Mutation/genetics , Nervous System Malformations/pathology , Pedigree , Peripheral Nerves/metabolism , Peripheral Nerves/pathology
10.
Ann Clin Transl Neurol ; 7(3): 353-362, 2020 03.
Article in English | MEDLINE | ID: mdl-32153140

ABSTRACT

OBJECTIVE: To develop, test, and iterate a comprehensive neuromuscular targeted gene panel in a national referral center. METHODS: We designed two iterations of a comprehensive targeted gene panel for neuromuscular disorders. Version 1 included 336 genes, which was increased to 464 genes in Version 2. Both panels used TargetSeqTM probe-based hybridization for target enrichment followed by Ion Torrent sequencing. Targeted high-coverage sequencing and analysis was performed on 2249 neurology patients from Australia and New Zealand (1054 Version 1, 1195 Version 2) from 2012 to 2015. No selection criteria were used other than referral from a suitable medical specialist (e.g., neurologist or clinical geneticist). Patients were classified into 15 clinical categories based on the clinical diagnosis from the referring clinician. RESULTS: Six hundred and sixty-five patients received a genetic diagnosis (30%). Diagnosed patients were significantly younger that undiagnosed patients (26.4 and 32.5 years, respectively; P = 4.6326E-9). The diagnostic success varied markedly between disease categories. Pathogenic variants in 10 genes explained 38% of the disease burden. Unexpected phenotypic expansions were discovered in multiple cases. Triage of unsolved cases for research exome testing led to the discovery of six new disease genes. INTERPRETATION: A comprehensive targeted diagnostic panel was an effective method for neuromuscular disease diagnosis within the context of an Australasian referral center. Use of smaller disease-specific panels would have precluded diagnosis in many patients and increased cost. Analysis through a centralized laboratory facilitated detection of recurrent, but under-recognized pathogenic variants.


Subject(s)
Genetic Testing/methods , High-Throughput Nucleotide Sequencing/methods , Neuromuscular Diseases/diagnosis , Neuromuscular Diseases/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Australia , Child , Child, Preschool , Cohort Studies , Female , Genetic Testing/standards , High-Throughput Nucleotide Sequencing/standards , Humans , Infant , Male , Middle Aged , New Zealand , Referral and Consultation , Young Adult
11.
Ann Clin Transl Neurol ; 6(11): 2328-2333, 2019 11.
Article in English | MEDLINE | ID: mdl-31612648

ABSTRACT

CAPN3 mutations cause a limb girdle muscular dystrophy. Functional characterization of novel mutations facilitates diagnosis of future cases. We have identified a novel (c.1992 + 2T>G) CAPN3 mutation that disrupts the donor splice site of intron 17 splicing out exon 17, with mRNA levels severely reduced or undetectable. The mutation induces a strong change in the 3D structure of the mRNA which supports no-go mRNA decay as the probable mechanism for RNA degradation. The mutation was identified in two unrelated Roma individuals showing a common ancestral origin and founder effect. This is the first Roma CAPN3 mutation to be reported.


Subject(s)
Calpain/genetics , Muscle Proteins/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Adolescent , Child , Female , Founder Effect , Humans , Introns/genetics , Male , Mutation , RNA Splicing , RNA Stability/genetics , Roma/genetics
12.
Nat Commun ; 10(1): 1396, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30918256

ABSTRACT

Myoglobin, encoded by MB, is a small cytoplasmic globular hemoprotein highly expressed in cardiac myocytes and oxidative skeletal myofibers. Myoglobin binds O2, facilitates its intracellular transport and serves as a controller of nitric oxide and reactive oxygen species. Here, we identify a recurrent c.292C>T (p.His98Tyr) substitution in MB in fourteen members of six European families suffering from an autosomal dominant progressive myopathy with highly characteristic sarcoplasmic inclusions in skeletal and cardiac muscle. Myoglobinopathy manifests in adulthood with proximal and axial weakness that progresses to involve distal muscles and causes respiratory and cardiac failure. Biochemical characterization reveals that the mutant myoglobin has altered O2 binding, exhibits a faster heme dissociation rate and has a lower reduction potential compared to wild-type myoglobin. Preliminary studies show that mutant myoglobin may result in elevated superoxide levels at the cellular level. These data define a recognizable muscle disease associated with MB mutation.


Subject(s)
Inclusion Bodies/pathology , Muscle Fibers, Skeletal/pathology , Muscle Weakness/genetics , Muscular Diseases/genetics , Myocytes, Cardiac/pathology , Myoglobin/genetics , Adult , Female , Heart Failure/etiology , Heme/metabolism , Humans , Male , Middle Aged , Muscle Weakness/physiopathology , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/physiopathology , Muscular Diseases/diagnostic imaging , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Mutation , Oxygen/metabolism , Pedigree , Respiratory Insufficiency/etiology , Superoxides/metabolism , Tomography, X-Ray Computed , White People/genetics
13.
Brain Commun ; 1(1): fcz011, 2019.
Article in English | MEDLINE | ID: mdl-32954258

ABSTRACT

Variants in MCM3AP, encoding the germinal-centre associated nuclear protein, have been associated with progressive polyneuropathy with or without intellectual disability and ptosis in some cases, and with a complex phenotype with immunodeficiency, skin changes and myelodysplasia. MCM3AP encoded protein functions as an acetyltransferase that acetylates the replication protein, MCM3, and plays a key role in the regulation of DNA replication. In this study, we report a novel variant in MCM3AP (p.Ile954Thr), in a family including three affected individuals with characteristic features of Charcot-Marie-Tooth neuropathy and multiple sclerosis, an inflammatory condition of the central nervous system without known genetic cause. The affected individuals were homozygous for a missense MCM3AP variant, located at the Sac3 domain, which was predicted to affect conserved amino acid likely important for the function of the germinal-centre associated nuclear protein. Our data support further expansion of the clinical spectrum linked to MCM3AP variant and highlight that MCM3AP should be considered in patients with accompaniment of recessive motor axonal Charcot-Marie-Tooth neuropathy and multiple sclerosis.

14.
Neurology ; 91(4): e339-e348, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29950440

ABSTRACT

OBJECTIVE: To describe a large series of BIN1 patients, in which a novel founder mutation in the Roma population of southern Spain has been identified. METHODS: Patients diagnosed with centronuclear myopathy (CNM) at 5 major reference centers for neuromuscular disease in Spain (n = 53) were screened for BIN1 mutations. Clinical, histologic, radiologic, and genetic features were analyzed. RESULTS: Eighteen patients from 13 families carried the p.Arg234Cys variant; 16 of them were homozygous for it and 2 had compound heterozygous p.Arg234Cys/p.Arg145Cys mutations. Both BIN1 variants have only been identified in Roma, causing 100% of CNM in this ethnic group in our cohort. The haplotype analysis confirmed all families are related. In addition to clinical features typical of CNM, such as proximal limb weakness and ophthalmoplegia, most patients in our cohort presented with prominent axial weakness, often associated with rigid spine. Severe fat replacement of paravertebral muscles was demonstrated by muscle imaging. This phenotype seems to be specific to the p.Arg234Cys mutation, not reported in other BIN1 mutations. Extreme clinical variability was observed in the 2 compound heterozygous patients for the p.Arg234Cys/p.Arg145Cys mutations, from a congenital onset with catastrophic outcome to a late-onset disease. Screening of European Roma controls (n = 758) for the p.Arg234Cys variant identified a carrier frequency of 3.5% among the Spanish Roma. CONCLUSION: We have identified a BIN1 founder Roma mutation associated with a highly specific phenotype, which is, from the present cohort, the main cause of CNM in Spain.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Founder Effect , Mallory Bodies/pathology , Muscular Dystrophies/genetics , Mutation/genetics , Myopathies, Structural, Congenital/genetics , Nuclear Proteins/genetics , Roma/genetics , Scoliosis/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Child , Cohort Studies , Humans , Mallory Bodies/genetics , Middle Aged , Muscular Dystrophies/diagnostic imaging , Muscular Dystrophies/ethnology , Myopathies, Structural, Congenital/diagnostic imaging , Myopathies, Structural, Congenital/ethnology , Phenotype , Prospective Studies , Retrospective Studies , Roma/ethnology , Scoliosis/diagnostic imaging , Scoliosis/ethnology , Spain/ethnology , Young Adult
15.
Neuromuscul Disord ; 27(9): 873-878, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28629674

ABSTRACT

Nephropathic cystinosis is an autosomal recessive lysosomal disease in which cystine cannot exit the lysosome to complete its degradation in the cytoplasm, thus accumulating in tissues. Some patients develop a distal myopathy involving mainly hand muscles. Myopathology descriptions from only 5 patients are available in the literature. We present a comprehensive clinical, pathological and genetic description of 3 patients from 2 families with nephropathic cystinosis. Intrafamiliar variability was detected in one family in which one sibling developed a severe distal myopathy while the other sibling did not show any signs of skeletal muscle involvement. One of the patients was on treatment with Cysteamine for over 12 years but still developed the usual complications of nephropathic cystinosis in his twenties. Novel pathological findings consisting in sarcoplasmic deposits reactive for slow myosin were identified. Three previously known and one novel mutation are reported. Nephropathic cystinosis should be included in the differential diagnosis of distal myopathies in those with early renal failure. Novel clinical and pathological features are reported here contributing to the characterization of the muscle involvement in nephropathic cystinosis.


Subject(s)
Amino Acid Transport Systems, Neutral/genetics , Cystinosis , Distal Myopathies , Cardiac Myosins/genetics , Cystine/metabolism , Cystinosis/complications , Cystinosis/genetics , Cystinosis/pathology , Distal Myopathies/etiology , Distal Myopathies/genetics , Distal Myopathies/pathology , Family Health , Female , Humans , Male , Microscopy, Electron , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Mutation/genetics , Myosin Heavy Chains/genetics , Myosins/metabolism , Young Adult
16.
EMBO Mol Med ; 8(11): 1289-1309, 2016 11.
Article in English | MEDLINE | ID: mdl-27807076

ABSTRACT

Skeletal muscle regeneration by muscle satellite cells is a physiological mechanism activated upon muscle damage and regulated by Notch signaling. In a family with autosomal recessive limb-girdle muscular dystrophy, we identified a missense mutation in POGLUT1 (protein O-glucosyltransferase 1), an enzyme involved in Notch posttranslational modification and function. In vitro and in vivo experiments demonstrated that the mutation reduces O-glucosyltransferase activity on Notch and impairs muscle development. Muscles from patients revealed decreased Notch signaling, dramatic reduction in satellite cell pool and a muscle-specific α-dystroglycan hypoglycosylation not present in patients' fibroblasts. Primary myoblasts from patients showed slow proliferation, facilitated differentiation, and a decreased pool of quiescent PAX7+ cells. A robust rescue of the myogenesis was demonstrated by increasing Notch signaling. None of these alterations were found in muscles from secondary dystroglycanopathy patients. These data suggest that a key pathomechanism for this novel form of muscular dystrophy is Notch-dependent loss of satellite cells.


Subject(s)
Glucosyltransferases/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Mutation , Receptors, Notch/metabolism , Satellite Cells, Skeletal Muscle/pathology , Signal Transduction , Biopsy , Glycosylation , Glycosyltransferases/metabolism , Humans , Muscles/pathology , Sequence Analysis, DNA , Spain
17.
Neurocrit Care ; 23(1): 103-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25650014

ABSTRACT

BACKGROUND: Hyperglycemia has been found to be associated with higher risk of ICU-acquired weakness. However, the impact of hyperglycemia on the outcome of patients with respiratory failure from a primary neuromuscular condition is not known. METHODS: We reviewed 85 patients admitted to an ICU at Mayo Clinic (Rochester) with primary acute neuromuscular respiratory failure. Time of hyperglycemia (defined as >140 mg/dL and as >180 mg/dL) was calculated for each patient. Associations between hyperglycemic time, insulin administered, and outcome measures (duration of mechanical ventilation, in-hospital mortality, functional outcome at discharge, and at last follow-up) were evaluated using logistic regression analysis. RESULTS: Although longer hyperglycemic time was associated with longer mechanical ventilation time and poorer short-term outcome on univariate analyses, these associations were no longer present when the analysis was adjusted for length of ICU stay. On this adjusted analysis, there were no significant associations between hyperglycemic time and duration of mechanical ventilation, in-hospital mortality, or functional outcome at discharge or at follow-up either for the entire cohort or for relevant subgroups (diabetics, non-diabetics, patients with neuropathy). The amount of insulin administered did not influence the outcome measures. Neither hyperglycemia nor the amount of insulin during the first 7 days of ICU admission was associated with any of the outcome measures. CONCLUSIONS: In our cohort, we did not find evidence that the duration of hyperglycemia or the amount of insulin given had any major impact on the outcomes of patients with primary acute neuromuscular respiratory failure.


Subject(s)
Hyperglycemia/diagnosis , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Neuromuscular Diseases/complications , Outcome Assessment, Health Care , Respiratory Insufficiency/etiology , Aged , Critical Care , Female , Follow-Up Studies , Humans , Hyperglycemia/drug therapy , Male , Middle Aged , Time Factors
18.
Brain ; 138(Pt 4): 836-44, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25681410

ABSTRACT

Dystroglycanopathies are a heterogeneous group of diseases with a broad phenotypic spectrum ranging from severe disorders with congenital muscle weakness, eye and brain structural abnormalities and intellectual delay to adult-onset limb-girdle muscular dystrophies without mental retardation. Most frequently the disease onset is congenital or during childhood. The exception is FKRP mutations, in which adult onset is a common presentation. Here we report eight patients from five non-consanguineous families where next generation sequencing identified mutations in the GMPPB gene. Six patients presented as an adult or adolescent-onset limb-girdle muscular dystrophy, one presented with isolated episodes of rhabdomyolysis, and one as a congenital muscular dystrophy. This report expands the phenotypic spectrum of GMPPB mutations to include limb-girdle muscular dystrophies with adult onset with or without intellectual disability, or isolated rhabdomyolysis.


Subject(s)
Muscular Dystrophies, Limb-Girdle/diagnosis , Muscular Dystrophies, Limb-Girdle/genetics , Mutation/genetics , Nucleotidyltransferases/genetics , Phenotype , Adolescent , Adult , Aged , Child , Child, Preschool , Dystroglycans/genetics , Fatal Outcome , Female , Humans , Male , Middle Aged , Pedigree , Young Adult
19.
Muscle Nerve ; 51(1): 140-3, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25187204

ABSTRACT

INTRODUCTION: Mutations in the choline kinase beta (CHKB) gene are associated with a congenital muscular dystrophy with giant mitochondria at the periphery of muscle fibers. METHODS: We describe a patient of Italian origin in whom whole-exome sequencing revealed a novel homozygous nonsense mutation, c.648C>A, p.(Tyr216*), in exon 5 of CHKB. RESULTS: The patient presented with limb-girdle weakness and hypotonia from birth with mental retardation, and had sudden and transient deteriorations of muscle strength with acute intercurrent illnesses. Previously undescribed sarcolemmal overexpression of utrophin was noted in the muscle biopsy. CONCLUSIONS: Pathological features broaden the description of the entity and provide new insight in the pathogenic mechanisms. This case highlights the usefulness of next-generation sequencing in the diagnosis of rare and incompletely understood conditions.


Subject(s)
Choline Kinase/genetics , Mitochondrial Myopathies/genetics , Mutation/genetics , Adenosine Triphosphate/metabolism , Child , Electron Transport Complex IV/metabolism , Female , Humans , Mitochondrial Myopathies/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myosins/metabolism , Utrophin/metabolism
20.
Hum Mol Genet ; 23(7): 1856-68, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24234652

ABSTRACT

Congenital myasthenic syndromes (CMS) are heterogeneous disorders in which the safety margin of neuromuscular transmission is compromised by one or more specific mechanisms. Using Sanger and exome sequencing in a CMS patient, we identified two heteroallelic mutations, p.Glu1233Lys and p.Arg1277His, in LRP4 coding for the postsynaptic low-density lipoprotein receptor-related protein 4. LRP4, expressed on the surface of the postsynaptic membrane of the neuromuscular junction, is a receptor for neurally secreted agrin, and LRP4 bound by agrin activates MuSK. Activated MuSK in concert with Dok-7 stimulates rapsyn to concentrate and anchor AChR on the postsynaptic membrane and interacts with other proteins implicated in the assembly and maintenance of the neuromuscular junction. LRP4 also functions as an inhibitor of Wnt/beta-catenin signaling. The identified mutations in LRP4 are located at the edge of its 3rd beta-propeller domain and decrease binding affinity of LRP4 for both MuSK and agrin. Mutations in the LRP4 3rd beta-propeller domain were previously reported to impair Wnt signaling and cause bone diseases including Cenani-Lenz syndactyly syndrome and sclerosteosis-2. By analyzing naturally occurring and artificially introduced mutations in the LRP4 3rd beta-propeller domain, we show that the edge of the domain regulates the MuSK signaling whereas its central cavity governs Wnt signaling. We conclude that LRP4 is a new CMS disease gene and that the 3rd beta propeller domain of LRP4 mediates the two signaling pathways in a position-specific manner.


Subject(s)
Agrin/metabolism , LDL-Receptor Related Proteins/genetics , Myasthenic Syndromes, Congenital/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cholinergic/metabolism , Adolescent , Animals , Base Sequence , COS Cells , Cell Line , Chlorocebus aethiops , Cholinergic Agonists/therapeutic use , Cholinesterase Inhibitors/therapeutic use , Edrophonium/therapeutic use , Enzyme Activation/genetics , Female , HEK293 Cells , Humans , Mice , Muscle Proteins/metabolism , Mutation , Neuromuscular Junction/metabolism , Protein Structure, Tertiary , Pyridostigmine Bromide/therapeutic use , Sequence Analysis, DNA , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway/genetics , beta Catenin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...