Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 661
Filter
1.
J Med Case Rep ; 18(1): 420, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39252049

ABSTRACT

BACKGROUND: Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes and nemaline myopathy are two rare genetic conditions. We report the first case reported in world literature with coexistence of both these rare disorders. CASE PRESENTATION: A 11-year-old previously healthy Sri Lankan male child, product of a nonconsanguineous marriage with normal development presented with acute onset short lasting recurring episodes of right-sided eye deviation with impaired consciousness. In between episodes he regained consciousness. Family history revealed a similar presentation in the mother at 36 years of age. Examination was significant for short stature and proximal upper and lower limb weakness. His plasma and cerebrospinal fluid lactate were elevated. Magnetic resonance imaging brain had evidence of an acute infarction in the right occipital territory. Sanger sequencing for common mitochondrial variants of mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes confirmed this diagnosis. Whole exome sequencing revealed pathogenic compound heterozygous variants in NEB gene implicating in coexisting nemaline myopathy. Acute presentation was managed with supportive care, antiepileptics, and mitochondrial supplementation. Currently he is stable on daily supplementation of arginine and limb-strengthening physiotherapy. He is being monitored closely clinically and with serum lactate level. CONCLUSION: Genetic diseases are rare. Coexistence of two genetic conditions is even rarer. Genetic confirmation of diagnosis is imperative for prediction of complications, accurate management, and genetic counseling.


Subject(s)
Myopathies, Nemaline , Humans , Male , Myopathies, Nemaline/genetics , Myopathies, Nemaline/complications , Child , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/complications , Mitochondrial Encephalomyopathies/diagnosis , Magnetic Resonance Imaging , Stroke , Sri Lanka , Acidosis, Lactic/genetics , Exome Sequencing
2.
Neuromuscul Disord ; 43: 29-38, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39180840

ABSTRACT

Nemaline myopathy (NM) is a congenital myopathy with generalised muscle weakness, most pronounced in neck flexor, bulbar and respiratory muscles. The aim of this cross-sectional study was to assess the Dutch NM patient cohort. We assessed medical history, physical examination, quality of life (QoL), fatigue severity, motor function (MFM), and respiratory muscle function. We included 18 of the 28 identified patients (13 females (11-67 years old); five males (31-74 years old)) with typical or mild NM and eight different genotypes. Nine patients (50 %) used a wheelchair, eight patients (44 %) used mechanical ventilation, and four patients (22 %) were on tube feeding. Spinal deformities were found in 14 patients (78 %). The median Medical Research Council (MRC) sum score was 38/60 [interquartile range 32-51] in typical and 48/60 [44-50] in mild NM. The experienced QoL was lower and fatigue severity was higher than reference values of the healthy population. The total MFM score was 55 % [49-94] in typical and 88 % [72-93] in mild NM. Most of the patients who performed spirometry had a restrictive lung function pattern (11/15). This identification and characterisation of the Dutch NM patient cohort is important for international collaboration and can guide the design of future clinical trials.


Subject(s)
Myopathies, Nemaline , Quality of Life , Humans , Male , Female , Cross-Sectional Studies , Middle Aged , Netherlands , Adult , Myopathies, Nemaline/genetics , Myopathies, Nemaline/physiopathology , Adolescent , Aged , Young Adult , Child , Severity of Illness Index , Fatigue/physiopathology , Respiratory Muscles/physiopathology
3.
BMJ Case Rep ; 17(6)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866581

ABSTRACT

Nemaline rod myopathy is an extremely rare muscle disease responsible for hypotonia and poor muscle strength in infants. The disease has variable phenotypic presentations across different ages, ranging from neonatal to the adult onset and from severe to asymptomatic varieties. Clinical features, muscle biopsy and genetic testing help in diagnosis. The histopathological examination shows the presence of rod-like structures or nemaline bodies in muscles. Management remains mainly supportive, and currently, there is no available curative treatment. This case report describes an infant presenting with gross hypotonia, poor handling of secretions and multiple extubation failures who was diagnosed by clinical exome sequencing. The patient harboured compound heterozygous variants in the NEB gene suggestive of nemaline rod myopathy. The newborn showed significant improvement in muscle strength after he was started on dietary L-tyrosine supplementation. This case highlights the emerging role of L-tyrosine in the supportive care of infants with nemaline rod myopathy.


Subject(s)
Myopathies, Nemaline , Tyrosine , Humans , Infant , Male , Muscle Hypotonia/drug therapy , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Myopathies, Nemaline/genetics , Myopathies, Nemaline/drug therapy , Treatment Outcome , Tyrosine/therapeutic use
4.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38869008

ABSTRACT

Cofilin, an actin-severing protein, plays key roles in muscle sarcomere addition and maintenance. Our previous work found that Drosophila cofilin (DmCFL) knockdown in muscle causes progressive deterioration of muscle structure and function and produces features seen in nemaline myopathy caused by cofilin mutations. We hypothesized that disruption of actin cytoskeleton dynamics by DmCFL knockdown would impact other aspects of muscle development, and, thus, conducted an RNA-sequencing analysis that unexpectedly revealed upregulated expression of numerous neuromuscular junction (NMJ) genes. We found that DmCFL is enriched in the muscle postsynaptic compartment and that DmCFL muscle knockdown causes F-actin disorganization in this subcellular domain prior to the sarcomere defects observed later in development. Despite NMJ gene expression changes, we found no significant changes in gross presynaptic Bruchpilot active zones or total postsynaptic glutamate receptor levels. However, DmCFL knockdown resulted in mislocalization of GluRIIA class glutamate receptors in more deteriorated muscles and strongly impaired NMJ transmission strength. These findings expand our understanding of the roles of cofilin in muscle to include NMJ structural development and suggest that NMJ defects may contribute to the pathophysiology of nemaline myopathy.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Neuromuscular Junction , Synaptic Transmission , Animals , Neuromuscular Junction/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Actin Depolymerizing Factors/metabolism , Actin Depolymerizing Factors/genetics , Actins/metabolism , Sarcomeres/metabolism , Gene Knockdown Techniques , Actin Cytoskeleton/metabolism , Myopathies, Nemaline/metabolism , Myopathies, Nemaline/genetics , Myopathies, Nemaline/pathology
5.
Stem Cell Res ; 77: 103435, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733812

ABSTRACT

We used a non-integrated reprogramming approach to establish a human induced pluripotent stem cell (hiPSC) line (INNDSUi004-A) from the skin fibroblasts of a 13-year-old female individual with Congenital Nemaline Myopath. The cells obtained have typical characteristics of embryonic stem cells, show expression of specific pluripotency markers, and can differentiate into three germ layers in vitro. This iPSC cell line has the genetic information of the patient and is a good model for studying disease mechanisms and developing novel therapies.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Myopathies, Nemaline , Induced Pluripotent Stem Cells/metabolism , Humans , Myopathies, Nemaline/pathology , Myopathies, Nemaline/genetics , Female , Cell Line , Adolescent , Fibroblasts/metabolism , Cellular Reprogramming
6.
Prenat Diagn ; 44(8): 996-998, 2024 07.
Article in English | MEDLINE | ID: mdl-38666792

ABSTRACT

We present a case of fetal akinesia deformation sequence due to nemaline myopathy (NM). In addition to the muscle manifestations, prenatal observations included an enlarged subarachnoid space and delayed cortical development. Trio whole-exome sequencing revealed a de novo novel pathogenic variant in the ACTA1 gene, which encodes skeletal muscle alpha-actin. Our findings suggest that brain abnormalities can occur prenatally in NM and support the potential role of skeletal muscle alpha-actin in the central nervous system.


Subject(s)
Actins , Myopathies, Nemaline , Humans , Female , Actins/genetics , Pregnancy , Adult , Myopathies, Nemaline/genetics , Exome Sequencing , Ultrasonography, Prenatal , Malformations of Cortical Development/genetics , Malformations of Cortical Development/diagnostic imaging , Arthrogryposis
7.
Acta Neuropathol ; 147(1): 72, 2024 04 18.
Article in English | MEDLINE | ID: mdl-38634969

ABSTRACT

Nebulin, a critical protein of the skeletal muscle thin filament, plays important roles in physiological processes such as regulating thin filament length (TFL), cross-bridge cycling, and myofibril alignment. Pathogenic variants in the nebulin gene (NEB) cause NEB-based nemaline myopathy (NEM2), a genetically heterogeneous disorder characterized by hypotonia and muscle weakness, currently lacking curative therapies. In this study, we examined a cohort of ten NEM2 patients, each with unique pathogenic variants, aiming to understand their impact on mRNA, protein, and functional levels. Results show that pathogenic truncation variants affect NEB mRNA stability and lead to nonsense-mediated decay of the mutated transcript. Moreover, a high incidence of cryptic splice site activation was found in patients with pathogenic splicing variants that are expected to disrupt the actin-binding sites of nebulin. Determination of protein levels revealed patients with either relatively normal or markedly reduced nebulin. We observed a positive relation between the reduction in nebulin and a reduction in TFL, or reduction in tension (both maximal and submaximal tension). Interestingly, our study revealed a pathogenic duplication variant in nebulin that resulted in a four-copy gain in the triplicate region of NEB and a much larger nebulin protein and longer TFL. Additionally, we investigated the effect of Omecamtiv mecarbil (OM), a small-molecule activator of cardiac myosin, on force production of type 1 muscle fibers of NEM2 patients. OM treatment substantially increased submaximal tension across all NEM2 patients ranging from 87 to 318%, with the largest effects in patients with the lowest level of nebulin. In summary, this study indicates that post-transcriptional or post-translational mechanisms regulate nebulin expression. Moreover, we propose that the pathomechanism of NEM2 involves not only shortened but also elongated thin filaments, along with the disruption of actin-binding sites resulting from pathogenic splicing variants. Significantly, our findings highlight the potential of OM treatment to improve skeletal muscle function in NEM2 patients, especially those with large reductions in nebulin levels.


Subject(s)
Myopathies, Nemaline , Urea , Humans , Actins , Muscle Weakness , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myopathies, Nemaline/drug therapy , Myopathies, Nemaline/genetics , Myopathies, Nemaline/pathology , Urea/analogs & derivatives , Muscle Proteins/genetics , Muscle Proteins/metabolism
9.
BMJ Case Rep ; 17(3)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38471704

ABSTRACT

HIV-associated myopathies include HIV-associated polymyositis, inclusion body myositis, diffuse infiltrative lymphocytosis syndrome and sporadic late-onset nemaline myopathy (HIV-NM). HIV-NM typically manifests as a painless, progressive proximal and axial muscle weakness with characteristic histological findings of intracytoplasmic rods, or nemaline bodies, seen in atrophic muscle fibres. HIV-NM presents prior to or shortly after initiation of antiretroviral therapy (ART) and is treated with intravenous immunoglobulin, glucocorticoids or immunosuppression. We present a case of HIV-NM in a patient with well-controlled HIV on decades-long ART with progressive bent spine syndrome, or camptocormia. This case highlights the importance of considering HIV-associated myopathies such as HIV-NM in patients with HIV who present with musculoskeletal complaints.


Subject(s)
HIV Infections , Muscular Atrophy, Spinal , Myopathies, Nemaline , Myositis, Inclusion Body , Spinal Curvatures , Humans , HIV Infections/complications , Muscle Fibers, Skeletal , Muscle, Skeletal/pathology , Myopathies, Nemaline/complications , Myopathies, Nemaline/pathology , Myopathies, Nemaline/therapy
10.
Hum Mol Genet ; 33(12): 1036-1054, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38493359

ABSTRACT

Nemaline myopathy (NM) is a rare congenital neuromuscular disorder characterized by muscle weakness and hypotonia, slow gross motor development, and decreased respiratory function. Mutations in at least twelve genes, all of each encode proteins that are either components of the muscle thin filament or regulate its length and stability, have been associated with NM. Mutations in Nebulin (NEB), a giant filamentous protein localized in the sarcomere, account for more than 50% of NM cases. At present, there remains a lack of understanding of whether NEB genotype influences nebulin function and NM-patient phenotypes. In addition, there is a lack of therapeutically tractable models that can enable drug discovery and address the current unmet treatment needs of patients. To begin to address these gaps, here we have characterized five new zebrafish models of NEB-related NM. These mutants recapitulate most aspects of NEB-based NM, showing drastically reduced survival, defective muscle structure, reduced contraction force, shorter thin filaments, presence of electron-dense structures in myofibers, and thickening of the Z-disks. This study represents the first extensive investigation of an allelic series of nebulin mutants, and thus provides an initial examination in pre-clinical models of potential genotype-phenotype correlations in human NEB patients. It also represents the first utilization of a set of comprehensive outcome measures in zebrafish, including correlation between molecular analyses, structural and biophysical investigations, and phenotypic outcomes. Therefore, it provides a rich source of data for future studies exploring the NM pathomechanisms, and an ideal springboard for therapy identification and development for NEB-related NM.


Subject(s)
Alleles , Disease Models, Animal , Muscle Proteins , Muscle, Skeletal , Mutation , Myopathies, Nemaline , Phenotype , Sarcomeres , Zebrafish , Myopathies, Nemaline/genetics , Myopathies, Nemaline/pathology , Myopathies, Nemaline/physiopathology , Zebrafish/genetics , Animals , Muscle Proteins/genetics , Muscle Proteins/metabolism , Sarcomeres/genetics , Sarcomeres/metabolism , Sarcomeres/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Humans , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
J Gen Physiol ; 156(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38376469

ABSTRACT

Nemaline myopathies are the most common form of congenital myopathies. Variants in ACTA1 (NEM3) comprise 15-25% of all nemaline myopathy cases. Patients harboring variants in ACTA1 present with a heterogeneous disease course characterized by stable or progressive muscle weakness and, in severe cases, respiratory failure and death. To date, no specific treatments are available. Since NEM3 is an actin-based thin filament disease, we tested the ability of tirasemtiv, a fast skeletal muscle troponin activator, to improve skeletal muscle function in a mouse model of NEM3, harboring the patient-based p.Asp286Gly variant in Acta1. Acute and long-term tirasemtiv treatment significantly increased muscle contractile capacity at submaximal stimulation frequencies in both fast-twitch extensor digitorum longus and gastrocnemius muscle, and intermediate-twitch diaphragm muscle in vitro and in vivo. Additionally, long-term tirasemtiv treatment in NEM3 mice resulted in a decreased respiratory rate with preserved minute volume, suggesting more efficient respiration. Altogether, our data support the therapeutic potential of fast skeletal muscle troponin activators in alleviating skeletal muscle weakness in a mouse model of NEM3 caused by the Acta1:p.Asp286Gly variant.


Subject(s)
Imidazoles , Myopathies, Nemaline , Pyrazines , Humans , Animals , Mice , Myopathies, Nemaline/drug therapy , Myopathies, Nemaline/genetics , Muscle Tonus , Actins/genetics , Muscle, Skeletal , Disease Models, Animal , Troponin
14.
Genes Genomics ; 46(5): 613-620, 2024 05.
Article in English | MEDLINE | ID: mdl-38363456

ABSTRACT

BACKGROUND: Nemaline Myopathy (NM) is a rare genetic disorder that affects muscle function and is characterized by the presence of nemaline rods in muscle fibers. These rods are abnormal structures that interfere with muscle contraction and can cause muscle weakness, respiratory distress, and other complications. NM is caused by variants in several genes, including TNNT1, which encodes the protein troponin T1. NM is inherited in an autosomal recessive pattern. The prevalence of heterozygous TNNT1 variants has been reported to be 1/152,000, indicating that the disease is relatively rare. OBJECTIVE: Investigation of TNNT1 gene variants that may cause cretin kinase elevation. METHODS: Detailed family histories and clinical data were recorded. Whole exome sequencing was performed and family segregation was done by Sanger sequencing. RESULTS: In this study, we report a 5-year-old girl with a novel variant recessive congenital TNNT1 myopathy. The patient had a novel homozygous (c.271_273del) deletion in the TNNT1 gene that is associated with creatine kinase elevation, which is a marker of muscle damage. CONCLUSION: This case expands the phenotypic spectrum of TNNT1 myopathy and highlights the importance of genetic testing and counseling for families affected by this rare disorder. In this study provides valuable insights into the genetic basis of NM and highlights the importance of early diagnosis and management for patients with this rare disorder. Further research is needed to better understand the pathophysiology of TNNT1 myopathy and to develop effective treatments for this debilitating condition.


Subject(s)
Myopathies, Nemaline , Female , Humans , Child, Preschool , Myopathies, Nemaline/genetics , Myopathies, Nemaline/diagnosis , Creatine Kinase/genetics , Homozygote , Genetic Testing , Troponin T/genetics
15.
Genes (Basel) ; 15(2)2024 02 05.
Article in English | MEDLINE | ID: mdl-38397198

ABSTRACT

BACKGROUND: Mutations in the KLHL40 gene are a common cause of severe or even lethal nemaline myopathy. Some cases with mild forms have been described, although the cases are still anecdotal. The aim of this paper was to systematically review the cases described in the literature and to describe a 12-year clinical and imaging follow-up in an Italian patient with KLHL40- related myopathy in order to suggest possible follow-up measurements. METHODS: Having searched through three electronic databases (PubMed, Scopus, and EBSCO), 18 articles describing 65 patients with homozygous or compound heterozygous KLHL40 mutations were selected. A patient with a KLHL40 homozygous mutation (c.1582G>A/p.E528K) was added and clinical and genetic data were collected. RESULTS: The most common mutation identified in our systematic review was the (c.1516A>C) followed by the (c.1582G>A). In our review, 60% percent of the patients died within the first 4 years of life. Clinical features were similar across the sample. Unfortunately, however, there is no record of the natural history data in the surviving patients. The 12-year follow-up of our patient revealed a slow improvement in her clinical course, identifying muscle MRI as the only possible marker of disease progression. CONCLUSIONS: Due to its clinical and genotype homogeneity, KLHL40-related myopathy may be a condition that would greatly benefit from the development of new gene therapies; muscle MRI could be a good biomarker to monitor disease progression.


Subject(s)
Muscle, Skeletal , Myopathies, Nemaline , Humans , Female , Muscle, Skeletal/diagnostic imaging , Follow-Up Studies , Muscle Proteins/genetics , Myopathies, Nemaline/genetics , Biomarkers , Disease Progression
16.
Sci Rep ; 14(1): 4306, 2024 02 21.
Article in English | MEDLINE | ID: mdl-38383731

ABSTRACT

Rare or novel missense variants in large genes such as TTN and NEB are frequent in the general population, which hampers the interpretation of putative disease-causing biallelic variants in patients with sporadic neuromuscular disorders. Often, when the first initial genetic analysis is performed, the reconstructed haplotype, i.e. phasing information of the variants is missing. Segregation analysis increases the diagnostic turnaround time and is not always possible if samples from family members are lacking. To overcome this difficulty, we investigated how well the linked-read technology succeeded to phase variants in these large genes, and whether it improved the identification of structural variants. Linked-read sequencing data of nemaline myopathy, distal myopathy, and proximal myopathy patients were analyzed for phasing, single nucleotide variants, and structural variants. Variant phasing was successful in the large muscle genes studied. The longest continuous phase blocks were gained using high-quality DNA samples with long DNA fragments. Homozygosity increased the number of phase blocks, especially in exome sequencing samples lacking intronic variation. In our cohort, linked-read sequencing added more information about the structural variation but did not lead to a molecular genetic diagnosis. The linked-read technology can support the clinical diagnosis of neuromuscular and other genetic disorders.


Subject(s)
Muscular Diseases , Myopathies, Nemaline , Neuromuscular Diseases , Humans , Haplotypes/genetics , Neuromuscular Diseases/diagnosis , Neuromuscular Diseases/genetics , DNA , High-Throughput Nucleotide Sequencing
17.
PLoS Genet ; 20(1): e1011117, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38198522

ABSTRACT

During striated muscle development the first periodically repeated units appear in the premyofibrils, consisting of immature sarcomeres that must undergo a substantial growth both in length and width, to reach their final size. Here we report that, beyond its well established role in sarcomere elongation, the Sarcomere length short (SALS) protein is involved in Z-disc formation and peripheral growth of the sarcomeres. Our protein localization data and loss-of-function studies in the Drosophila indirect flight muscle strongly suggest that radial growth of the sarcomeres is initiated at the Z-disc. As to thin filament elongation, we used a powerful nanoscopy approach to reveal that SALS is subject to a major conformational change during sarcomere development, which might be critical to stop pointed end elongation in the adult muscles. In addition, we demonstrate that the roles of SALS in sarcomere elongation and radial growth are both dependent on formin type of actin assembly factors. Unexpectedly, when SALS is present in excess amounts, it promotes the formation of actin aggregates highly resembling the ones described in nemaline myopathy patients. Collectively, these findings helped to shed light on the complex mechanisms of SALS during the coordinated elongation and thickening of the sarcomeres, and resulted in the discovery of a potential nemaline myopathy model, suitable for the identification of genetic and small molecule inhibitors.


Subject(s)
Myopathies, Nemaline , Sarcomeres , Animals , Humans , Sarcomeres/metabolism , Formins/metabolism , Actins/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Drosophila/metabolism
18.
Neuromuscul Disord ; 35: 29-32, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38219297

ABSTRACT

Patients with myopathies caused by pathogenic variants in tropomyosin genes TPM2 and TPM3 usually have muscle hypotonia and weakness, their muscle biopsies often showing fibre size disproportion and nemaline bodies. Here, we describe a series of patients with hypercontractile molecular phenotypes, high muscle tone, and mostly non-specific myopathic biopsy findings without nemaline bodies. Three of the patients had trismus, whilst in one patient, the distal joints of her fingers flexed on extension of the wrists. In one biopsy from a patient with a rare TPM3 pathogenic variant, cores and minicores were observed, an unusual finding in TPM3-caused myopathy. The variants alter conserved contact sites between tropomyosin and actin.


Subject(s)
Muscular Diseases , Myopathies, Nemaline , Humans , Female , Muscle, Skeletal/pathology , Tropomyosin/genetics , Muscular Diseases/pathology , Muscle Hypertonia/pathology , Phenotype , Myopathies, Nemaline/genetics , Myopathies, Nemaline/pathology , Mutation
19.
Acta Neurol Belg ; 124(1): 91-99, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37525074

ABSTRACT

BACKGROUND: Nemaline myopathy, the most common of the congenital myopathies, is caused by various genetic mutations. In this study, we attempted to investigate the clinical features, muscle pathology and genetic features of 15 patients with nemaline myopathy. RESULTS: Among the 15 patients, there were 9 (60.00%) males and 6 (40.00%) females, and 9 (60.00%) of them came from three families respectively. The age of seeing a doctor ranged from 9 to 52 years old, the age of onset was from 5 to 23 years old, and the duration of disease ranged from 3 to 35 years. Ten out of the 15 patients had high arched palate and elongated face. Only one patient had mild respiratory muscle involvement and none had dysphagia. Muscle biopsies were performed in 9 out of the 15 patients. Pathologically, muscle fibers of different sizes, atrophic muscle fibers and compensatory hypertrophic fibers could be found, and occasionally degenerated and necrotic muscle fibers were observed. Different degrees of nemaline bodies aggregation could be seen in all 9 patients. The distribution of type I and type II muscle fibers were significantly abnormal in patients with nemaline myopathy caused by NEB gene, however, it was basically normal in patients with nemaline myopathy caused by TPM3 gene and ACTA1 gene. Electron microscopic analysis of 6 patients showed that nemaline bodies aggregated between myofibrils were found in 5(83.33%) cases, and most of them were located near the Z band, but no intranuclear rods were found. The gene analysis of 15 NM patients showed that three NM-related genes were harbored, including 11 (73.33%) patients with NEB, 3 (20.00%) patients with TPM3, and 1 (6.67%) patient with ACTA1, respectively. A total of 12 mutation sites were identified and included 10 (83.33%) mutations in exon and 2(16.67%) mutations in intron. CONCLUSIONS: The clinical phenotype of nemaline myopathy is highly heterogeneous. Muscle pathology shows that nemaline bodies aggregation is an important feature for the diagnosis of NM. NEB is the most frequent causative gene in this cohort. The splicing mutation, c.21522 + 3A > G may be the hotspot mutation of the NEB gene in Chinese NM patients.


Subject(s)
Muscular Diseases , Myopathies, Nemaline , Male , Female , Humans , Child , Adolescent , Young Adult , Adult , Middle Aged , Myopathies, Nemaline/genetics , Myopathies, Nemaline/pathology , Muscle, Skeletal/pathology , Mutation , China
20.
Neurol Sci ; 45(3): 1225-1231, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37851294

ABSTRACT

BACKGROUND: Inherited nemaline myopathy is one of the most common congenital myopathies. This genetically heterogeneous disease is defined by the presence of nemaline bodies in muscle biopsy. The phenotypic spectrum is wide and cognitive involvement has been reported, although not extensively evaluated. METHODS: We report two nemaline myopathy patients presenting pronounced central nervous system involvement leading to functional compromise and novel facial and skeletal dysmorphic findings, possibly expanding the disease phenotype. RESULTS: One patient had two likely pathogenic NEB variants, c.2943G > A and c.8889 + 1G > A, and presented cognitive impairment and dysmorphic features, and the other had one pathogenic variant in ACTA1, c.169G > C (p.Gly57Arg), presenting autism spectrum disorder and corpus callosum atrophy. Both patients had severe cognitive involvement despite milder motor dysfunction. CONCLUSION: We raise the need for further studies regarding the role of thin filament proteins in the central nervous system and for a systematic cognitive assessment of congenital myopathy patients.


Subject(s)
Autism Spectrum Disorder , Myopathies, Nemaline , Humans , Myopathies, Nemaline/genetics , Myopathies, Nemaline/pathology , Muscle, Skeletal/pathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Central Nervous System , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL