Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Neuromuscul Disord ; 43: 29-38, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39180840

RESUMO

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.


Assuntos
Miopatias da Nemalina , Qualidade de Vida , Humanos , Masculino , Feminino , Estudos Transversais , Pessoa de Meia-Idade , Países Baixos , Adulto , Miopatias da Nemalina/genética , Miopatias da Nemalina/fisiopatologia , Adolescente , Idoso , Adulto Jovem , Criança , Índice de Gravidade de Doença , Fadiga/fisiopatologia , Músculos Respiratórios/fisiopatologia
2.
Hum Mol Genet ; 33(12): 1036-1054, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38493359

RESUMO

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.


Assuntos
Alelos , Modelos Animais de Doenças , Proteínas Musculares , Músculo Esquelético , Mutação , Miopatias da Nemalina , Fenótipo , Sarcômeros , Peixe-Zebra , Miopatias da Nemalina/genética , Miopatias da Nemalina/patologia , Miopatias da Nemalina/fisiopatologia , Peixe-Zebra/genética , Animais , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Sarcômeros/genética , Sarcômeros/metabolismo , Sarcômeros/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Humanos , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
3.
Neurology ; 96(10): e1425-e1436, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33397769

RESUMO

OBJECTIVE: Nemaline myopathy (NM) is a rare neuromuscular condition with clinical and genetic heterogeneity. To establish disease natural history, we performed a cross-sectional study of NM, complemented by longitudinal assessment and exploration of pilot outcome measures. METHODS: Fifty-seven individuals with NM were recruited at 2 family workshops, including 16 examined at both time points. Participants were evaluated by clinical history and physical examination. Functional outcome measures included the Motor Function Measure (MFM), pulmonary function tests (PFTs), myometry, goniometry, and bulbar assessments. RESULTS: The most common clinical classification was typical congenital (54%), whereas 42% had more severe presentations. Fifty-eight percent of individuals needed mechanical support, with 26% requiring wheelchair, tracheostomy, and feeding tube. The MFM scale was performed in 44 of 57 participants and showed reduced scores in most with little floor/ceiling effect. Of the 27 individuals completing PFTs, abnormal values were observed in 65%. Last, bulbar function was abnormal in all patients examined, as determined with a novel outcome measure. Genotypes included mutations in ACTA1 (18), NEB (20), and TPM2 (2). Seventeen individuals were genetically unresolved. Patients with pathogenic ACTA1 and NEB variants were largely similar in clinical phenotype. Patients without genetic resolution had more severe disease. CONCLUSION: We present a comprehensive cross-sectional study of NM. Our data identify significant disabilities and support a relatively stable disease course. We identify a need for further diagnostic investigation for the genetically unresolved group. MFM, PFTs, and the slurp test were identified as promising outcome measures for future clinical trials.


Assuntos
Miopatias da Nemalina/fisiopatologia , Actinas/genética , Adolescente , Adulto , Criança , Pré-Escolar , Estudos de Coortes , Estudos Transversais , Avaliação da Deficiência , Progressão da Doença , Nutrição Enteral , Feminino , Genótipo , Humanos , Lactente , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/genética , Miopatias da Nemalina/genética , Projetos Piloto , Desempenho Psicomotor , Testes de Função Respiratória , Sialorreia/epidemiologia , Sialorreia/etiologia , Traqueostomia/estatística & dados numéricos , Resultado do Tratamento , Cadeiras de Rodas/estatística & dados numéricos , Adulto Jovem
4.
Neuromuscul Disord ; 30(3): 207-212, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32008911

RESUMO

Fingerprint bodies are observed in a variety of clinical situations with no definite genetic cause identified so far. We report for the first time the association of fingerprint bodies with rods in a patient who developed a slowly progressive myopathy affecting the face and limb extremities. Ultrastructural examination first disclosed fingerprint bodies and on a second biopsy, associated cytoplasmic bodies and rods. Next Generation Sequencing panel of congenital nemaline myopathy genes allowed the identification of two novel variants, a deleterious missense variant (c.1628G>T, p.Arg543Leu) located in the WASP-homology 2 domain, and a deletion (c.366delG, p.Lys122AsnFs*6) in the LMOD3 gene, generally causing severe nemaline myopathy with antenatal onset and early death. Recently, a less severe phenotype similar to our case has been reported. Our study confirms the existence of milder phenotypes linked to LMOD3 mutations and underlines that fingerprint bodies, though not specific, may be an early ultrastructural marker that could be linked, among others, to nemaline myopathy.


Assuntos
Proteínas dos Microfilamentos/genética , Fibras Musculares Esqueléticas/ultraestrutura , Doenças Musculares , Miopatias da Nemalina , Adulto , Humanos , Masculino , Microscopia Eletrônica , Doenças Musculares/diagnóstico , Doenças Musculares/genética , Doenças Musculares/patologia , Doenças Musculares/fisiopatologia , Miopatias da Nemalina/diagnóstico , Miopatias da Nemalina/genética , Miopatias da Nemalina/patologia , Miopatias da Nemalina/fisiopatologia , Fenótipo , Adulto Jovem
5.
Ann Neurol ; 87(4): 568-583, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31970803

RESUMO

OBJECTIVE: Recessive null variants of the slow skeletal muscle troponin T1 (TNNT1) gene are a rare cause of nemaline myopathy that is fatal in infancy due to respiratory insufficiency. Muscle biopsy shows rods and fiber type disproportion. We report on 4 French Canadians with a novel form of recessive congenital TNNT1 core-rod myopathy. METHODS: Patients underwent full clinical characterization, lower limb magnetic resonance imaging (MRI), muscle biopsy, and genetic testing. A zebrafish loss-of-function model using morpholinos was created to assess the pathogenicity of the identified variant. Wild-type or mutated human TNNT1 mRNAs were coinjected with morpholinos to assess their abilities to rescue the morphant phenotype. RESULTS: Three adults and 1 child shared a novel missense homozygous variant in the TNNT1 gene (NM_003283.6: c.287T > C; p.Leu96Pro). They developed from childhood very slowly progressive limb-girdle weakness with rigid spine and disabling contractures. They suffered from restrictive lung disease requiring noninvasive mechanical ventilation in 3 patients, as well as recurrent episodes of rhabdomyolysis triggered by infections, which were relieved by dantrolene in 1 patient. Older patients remained ambulatory into their 60s. MRI of the leg muscles showed fibrofatty infiltration predominating in the posterior thigh and the deep posterior leg compartments. Muscle biopsies showed multiminicores and lobulated fibers, rods in half the patients, and no fiber type disproportion. Wild-type TNNT1 mRNA rescued the zebrafish morphants, but mutant transcripts failed to do so. INTERPRETATION: This study expands the phenotypic spectrum of TNNT1 myopathy and provides functional evidence for the pathogenicity of the newly identified missense mutation. ANN NEUROL 2020;87:568-583.


Assuntos
Músculo Esquelético/patologia , Miopatias da Nemalina/fisiopatologia , RNA Mensageiro/metabolismo , Troponina T/genética , Animais , Criança , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Morfolinos , Músculo Esquelético/ultraestrutura , Miopatias da Nemalina/genética , Miopatias da Nemalina/patologia , Rabdomiólise/genética , Rabdomiólise/fisiopatologia , Troponina T/metabolismo , Peixe-Zebra
6.
Muscle Nerve ; 61(3): 416-424, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31893464

RESUMO

INTRODUCTION: The conditional nebulin knockout mouse is a new model mimicking nemaline myopathy, a rare disease characterized by muscle weakness and rods within muscle fibers. We investigated the impact of nebulin (NEB) deficiency on muscle function in vivo. METHODS: Conditional nebulin knockout mice and control littermates were studied at 10 to 12 months. Muscle function (force and fatigue) and anatomy (muscles volume and fat content) were measured in vivo. Myosin heavy chain (MHC) composition and nebulin (NEB) protein expression were assessed by protein electrophoresis. RESULTS: Conditional nebulin knockout mice displayed a lower NEB level (-90%) leading to a 40% and 45% reduction in specific maximal force production and muscles volume, respectively. Nebulin deficiency was also associated with higher resistance to fatigue and increased MHC I content. DISCUSSION: Adult nebulin-deficient mice displayed severe muscle atrophy and weakness in vivo related to a low NEB content but an improved fatigue resistance due to a slower contractile phenotype.


Assuntos
Proteínas Musculares/genética , Músculo Esquelético/fisiopatologia , Miopatias da Nemalina/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Membro Posterior , Camundongos Knockout , Contração Muscular , Fadiga Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miopatias da Nemalina/genética , Miopatias da Nemalina/patologia , Cadeias Pesadas de Miosina/metabolismo
7.
Biochem Biophys Res Commun ; 523(1): 258-262, 2020 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-31864708

RESUMO

Ghost muscle fibres reconstituted with myosin heads labeled with the fluorescent probe 1,5-IAEDANS were used for analysis of muscle fibre dysfunction associated with the R133W mutation in ß-tropomyosin (Tpm2.2). By using polarized microscopy, we showed that at high Ca2+ the R133W mutation in both αß-Tpm heterodimers and ßß-Tpm homodimers decreases the amount of the myosin heads strongly bound to F-actin and the number of switched-on actin monomers, with this effect being stronger for ßß-Tpm. This mutation also inhibits the shifting of the R133W-Tpm strands towards the open position and the efficiency of the cross-bridge work. At low Ca2+, the amount of the strongly bound myosin heads is lower for R133W-Tpms than for WT-Tpms which may contribute to a low myofilament Ca2+-sensitivity of the R133W-Tpms. It is concluded that freezing of the mutant αß- or ßß-Tpm close to the blocked position inhibits the strong binding of the cross-bridges and the switching on of actin monomers which may be the reason for muscle weakness associated with the R133W mutation in ß-tropomyosin. The use of reagents that activate myosin may be appropriate to restore muscle function in patients with the R133W mutation.


Assuntos
Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Mutação , Tropomiosina/genética , Animais , Cálcio/metabolismo , Masculino , Debilidade Muscular/genética , Debilidade Muscular/fisiopatologia , Miopatias da Nemalina/genética , Miopatias da Nemalina/fisiopatologia , Coelhos , Tropomiosina/metabolismo
8.
PLoS One ; 14(11): e0224467, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31721788

RESUMO

Nemaline myopathy (NEM) is a congenital neuromuscular disorder primarily caused by nebulin gene (NEB) mutations. NEM is characterized by muscle weakness for which currently no treatments exist. In NEM patients a predominance of type I fibers has been found. Thus, therapeutic options targeting type I fibers could be highly beneficial for NEM patients. Because type I muscle fibers express the same myosin isoform as cardiac muscle (Myh7), the effect of omecamtiv mecarbil (OM), a small molecule activator of Myh7, was studied in a nebulin-based NEM mouse model (Neb cKO). Skinned single fibers were activated by exogenous calcium and force was measured at a wide range of calcium concentrations. Maximal specific force of type I fibers was much less in fibers from Neb cKO animals and calcium sensitivity of permeabilized single fibers was reduced (pCa50 6.12 ±0.08 (cKO) vs 6.36 ±0.08 (CON)). OM increased the calcium sensitivity of type I single muscle fibers. The greatest effect occurred in type I fibers from Neb cKO muscle where OM restored the calcium sensitivity to that of the control type I fibers. Forces at submaximal activation levels (pCa 6.0-6.5) were significantly increased in Neb cKO fibers (~50%) but remained below that of control fibers. OM also increased isometric force and power during isotonic shortening of intact whole soleus muscle of Neb cKO mice, with the largest effects at physiological stimulation frequencies. We conclude that OM has the potential to improve the quality of life of NEM patients by increasing the force of type I fibers at submaximal activation levels.


Assuntos
Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Proteínas Musculares/genética , Miopatias da Nemalina/tratamento farmacológico , Ureia/análogos & derivados , Animais , Cálcio/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Miopatias da Nemalina/genética , Miopatias da Nemalina/fisiopatologia , Ureia/farmacologia , Ureia/uso terapêutico
9.
Stem Cell Res ; 40: 101559, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31526942

RESUMO

Nemaline myopathy-4 (NEM4) is a very rare inherited muscle disorder caused by a heterozygous mutation in tropomyosin-2 (TPM2) gene. We established an induced pluripotent stem cell (iPSC) line from peripheral blood mononuclear cells of a 3-month-old girl with NEM4 carrying a heterozygous mutation (c.397C>T (p.R133W)) in TPM2 gene. This iPSC line showed a normal karyotype, expressed pluripotency markers, showed differentiation potential and harbored the original mutation of c.397C>T in the TPM2 gene.


Assuntos
Linhagem Celular/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Miopatias da Nemalina/genética , Tropomiosina/genética , Diferenciação Celular , Linhagem Celular/citologia , Feminino , Heterozigoto , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Lactente , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Mutação , Miopatias da Nemalina/metabolismo , Miopatias da Nemalina/fisiopatologia , Tropomiosina/metabolismo
10.
Neurology ; 93(3): e298-e305, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31167932

RESUMO

OBJECTIVE: To describe the clinical phenotype, long-term treatment outcome, and overall survival of sporadic late-onset nemaline myopathy (SLONM) with or without a monoclonal protein (MP). METHODS: We conducted a retrospective chart review of patients seen between September 2000 and June 2017 and collected clinical, laboratory, and survival data. Treatment response was classified as mild, moderate, or marked as adjudged by predefined criteria. RESULTS: We identified 28 patients with SLONM; 17 (61%) had an associated MP. Median age at symptom onset was 62 years. Diagnosis was often delayed by a median of 35 months from symptom onset. There was no difference in clinical or laboratory features between patients with or without MP. Although the majority of patients had proximal or axial weakness at onset, about 18% of patients had atypical presentations. A total of 7/9 (78%) patients receiving IV immunoglobulin (IVIg), 6/8 (75%) receiving hematologic therapy as either autologous stem cell transplant (ASCT) or chemotherapy, and 1/8 (13%) receiving immunosuppressive therapies responded to treatment (p = 0.001). All 3 patients with marked response were treated with IVIg; 2 of them had an MP. The 5-year and 10-year overall survival from symptom onset was 92% and 68%, respectively, with no difference between patients with or without MP. CONCLUSION: SLONM has a wide spectrum of clinical presentations. In this contemporary case series, overall survival of patients did not seem to be affected by the presence of an MP. Initial treatment with IVIg is reasonable in all patients, followed by ASCT or chemotherapy as second-line therapy in patients with an associated MP.


Assuntos
Imunoglobulinas Intravenosas/uso terapêutico , Imunossupressores/uso terapêutico , Miopatias da Nemalina/fisiopatologia , Paraproteinemias/terapia , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Fatores Imunológicos/uso terapêutico , Lenalidomida/uso terapêutico , Masculino , Pessoa de Meia-Idade , Miopatias da Nemalina/complicações , Miopatias da Nemalina/metabolismo , Miopatias da Nemalina/terapia , Paraproteinemias/complicações , Paraproteinemias/metabolismo , Estudos Retrospectivos , Transplante de Células-Tronco , Taxa de Sobrevida , Talidomida/uso terapêutico , Transplante Autólogo
11.
J Physiol ; 597(15): 3999-4012, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31148174

RESUMO

KEY POINTS: The pathogenic mechanism and the neuromuscular reflex-related phenotype (e.g. tremors accompanied by clonus) of Amish nemaline myopathy, as well as of other recessively inherited TNNT1 myopathies, remain to be clarified. The truncated slow skeletal muscle isoform of troponin T (ssTnT) encoded by the mutant TNNT1 gene is unable to incorporate into myofilaments and is degraded in muscle cells. By contrast to extrafusal muscle fibres, spindle intrafusal fibres of normal mice contain a significant level of cardiac TnT and a low molecular weight splice form of ssTnT. Intrafusal fibres of ssTnT-knockout mice have significantly increased cardiac TnT. Rotarod and balance beam tests have revealed abnormal neuromuscular co-ordination in ssTnT-knockout mice and a blunted response to a spindle sensitizer, succinylcholine. The loss of ssTnT and a compensatory increase of cardiac TnT in intrafusal nuclear bag fibres may increase myofilament Ca2+ -sensitivity and tension, impairing spindle function, thus identifying a novel mechanism for the development of targeted treatment. ABSTRACT: A nonsense mutation at codon Glu180 of TNNT1 gene causes Amish nemaline myopathy (ANM), a recessively inherited disease with infantile lethality. TNNT1 encodes the slow skeletal muscle isoform of troponin T (ssTnT). The truncated ssTnT is unable to incorporate into myofilament and is degraded in muscle cells. The symptoms of ANM include muscle weakness, atrophy, contracture and tremors accompanied by clonus. An ssTnT-knockout (KO) mouse model recapitulates key features of ANM such as atrophy of extrafusal slow muscle fibres and increased fatigability. However, the neuromuscular reflex-related symptoms of ANM have not been explained. By isolating muscle spindles from ssTnT-KO and control mice aiming to examine the composition of myofilament proteins, we found that, in contrast to extrafusal fibres, intrafusal fibres contain a significant level of cardiac TnT and the low molecular weight splice form of ssTnT. Intrafusal fibres from ssTnT-KO mice have significantly increased cardiac TnT. Rotarod and balance beam tests revealed impaired neuromuscular co-ordination in ssTnT-KO mice, indicating abnormality in spindle functions. Unlike the wild-type control, the beam running ability of ssTnT-KO mice had a blunted response to a spindle sensitizer, succinylcholine. Immunohistochemistry detected ssTnT and cardiac TnT in nuclear bag fibres, whereas fast skeletal muscle TnT was detected in nuclear chain fibres, and cardiac α-myosin was present in one of the two nuclear bag fibres. The loss of ssTnT and a compensatory increase of cardiac TnT in nuclear bag fibres would increase myofilament Ca2+ -sensitivity and tension, thus affecting spindle activities. This mechanism provides an explanation for the pathophysiology of ANM, as well as a novel target for treatment.


Assuntos
Fibras Musculares Esqueléticas/metabolismo , Fusos Musculares/metabolismo , Miopatias da Nemalina/genética , Troponina T/genética , Animais , Células Cultivadas , Locomoção , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/fisiologia , Miofibrilas/metabolismo , Miopatias da Nemalina/metabolismo , Miopatias da Nemalina/fisiopatologia
12.
Acta Neuropathol ; 138(3): 477-495, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31218456

RESUMO

Nemaline myopathy (NM) is a skeletal muscle disorder caused by mutations in genes that are generally involved in muscle contraction, in particular those related to the structure and/or regulation of the thin filament. Many pathogenic aspects of this disease remain largely unclear. Here, we report novel pathological defects in skeletal muscle fibres of mouse models and patients with NM: irregular spacing and morphology of nuclei; disrupted nuclear envelope; altered chromatin arrangement; and disorganisation of the cortical cytoskeleton. Impairments in contractility are the primary cause of these nuclear defects. We also establish the role of microtubule organisation in determining nuclear morphology, a phenomenon which is likely to contribute to nuclear alterations in this disease. Our results overlap with findings in diseases caused directly by mutations in nuclear envelope or cytoskeletal proteins. Given the important role of nuclear shape and envelope in regulating gene expression, and the cytoskeleton in maintaining muscle fibre integrity, our findings are likely to explain some of the hallmarks of NM, including contractile filament disarray, altered mechanical properties and broad transcriptional alterations.


Assuntos
Citoesqueleto/patologia , Contração Muscular/fisiologia , Músculo Esquelético/patologia , Miopatias da Nemalina/patologia , Adulto , Idoso , Animais , Núcleo Celular/patologia , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Músculo Esquelético/fisiopatologia , Miopatias da Nemalina/fisiopatologia , Adulto Jovem
13.
Hum Mol Genet ; 28(15): 2549-2560, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30986853

RESUMO

Nemaline myopathy (NM) is the most common form of congenital myopathy that results in hypotonia and muscle weakness. This disease is clinically and genetically heterogeneous, but three recently discovered genes in NM encode for members of the Kelch family of proteins. Kelch proteins act as substrate-specific adaptors for Cullin 3 (CUL3) E3 ubiquitin ligase to regulate protein turnover through the ubiquitin-proteasome machinery. Defects in thin filament formation and/or stability are key molecular processes that underlie the disease pathology in NM; however, the role of Kelch proteins in these processes in normal and diseases conditions remains elusive. Here, we describe a role of NM causing Kelch protein, KLHL41, in premyofibil-myofibil transition during skeletal muscle development through a regulation of the thin filament chaperone, nebulin-related anchoring protein (NRAP). KLHL41 binds to the thin filament chaperone NRAP and promotes ubiquitination and subsequent degradation of NRAP, a process that is critical for the formation of mature myofibrils. KLHL41 deficiency results in abnormal accumulation of NRAP in muscle cells. NRAP overexpression in transgenic zebrafish resulted in a severe myopathic phenotype and absence of mature myofibrils demonstrating a role in disease pathology. Reducing Nrap levels in KLHL41 deficient zebrafish rescues the structural and function defects associated with disease pathology. We conclude that defects in KLHL41-mediated ubiquitination of sarcomeric proteins contribute to structural and functional deficits in skeletal muscle. These findings further our understanding of how the sarcomere assembly is regulated by disease-causing factors in vivo, which will be imperative for developing mechanism-based specific therapeutic interventions.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miopatias da Nemalina/metabolismo , Ubiquitinação , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Camundongos , Miofibrilas/metabolismo , Miopatias da Nemalina/genética , Miopatias da Nemalina/fisiopatologia , Fenótipo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
14.
Hum Mol Genet ; 28(10): 1709-1725, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30689900

RESUMO

Nebulin is a large skeletal muscle protein wound around the thin filaments, with its C-terminus embedded within the Z-disk and its N-terminus extending out toward the thin filament pointed end. While nebulin's C-terminus has been implicated in both sarcomeric structure and function as well as the development of nemaline myopathy, the contributions of this region remain largely unknown. Additionally, the C-terminus is reported to contribute to muscle hypertrophy via the IGF-1 growth pathway. To study the functions of nebulin's C-terminus, we generated a mouse model deleting the final two unique C-terminal domains, the serine-rich region (SRR) and the SH3 domain (NebΔ163-165). Homozygous NebΔ163-165 mice that survive past the neonatal stage exhibit a mild weight deficit. Characterization of these mice revealed that the truncation caused a moderate myopathy phenotype reminiscent of nemaline myopathy despite the majority of nebulin being localized properly in the thin filaments. This phenotype included muscle weight loss, changes in sarcomere structure, as well as a decrease in force production. Glutathione S-transferase (GST) pull-down experiments found novel binding partners with the SRR, several of which are associated with myopathies. While the C-terminus does not appear to be a limiting step in muscle growth, the IGF-1 growth pathway remained functional despite the deleted domains being proposed to be essential for IGF-1 mediated hypertrophy. The NebΔ163-165 mouse model emphasizes that nebulin's C-terminus is necessary for proper sarcomeric development and shows that its loss is sufficient to induce myopathy.


Assuntos
Fator de Crescimento Insulin-Like I/genética , Proteínas Musculares/genética , Miopatias da Nemalina/genética , Sarcômeros/genética , Citoesqueleto de Actina/genética , Sequência de Aminoácidos/genética , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Homozigoto , Humanos , Hipertrofia/genética , Hipertrofia/patologia , Camundongos , Debilidade Muscular/genética , Debilidade Muscular/patologia , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/patologia , Miopatias da Nemalina/fisiopatologia , Fenótipo , Sarcômeros/química
16.
Hum Mol Genet ; 27(18): 3272-3282, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29931346

RESUMO

We describe the natural history of 'Amish' nemaline myopathy (ANM), an infantile-onset, lethal disease linked to a pathogenic c.505G>T nonsense mutation of TNNT1, which encodes the slow fiber isoform of troponin T (TNNT1; a.k.a. TnT). The TNNT1 c.505G>T allele has a carrier frequency of 6.5% within Old Order Amish settlements of North America. We collected natural history data for 106 ANM patients born between 1923 and 2017. Over the last two decades, mean age of molecular diagnosis was 16 ± 27 days. TNNT1 c.505G>T homozygotes were normal weight at birth but failed to thrive by age 9 months. Presenting neonatal signs were axial hypotonia, hip and shoulder stiffness, and tremors, followed by progressive muscle weakness, atrophy and contractures. Affected children developed thoracic rigidity, pectus carinatum and restrictive lung disease during infancy, and all succumbed to respiratory failure by 6 years of age (median survival 18 months, range 0.2-66 months). Muscle histology from two affected children showed marked fiber size variation owing to both Type 1 myofiber smallness (hypotrophy) and Type 2 fiber hypertrophy, with evidence of nemaline rods, myofibrillar disarray and vacuolar pathology in both fiber types. The truncated slow TNNT1 (TnT) fragment (p.Glu180Ter) was undetectable in ANM muscle, reflecting its rapid proteolysis and clearance from sarcoplasm. Similar functional and histological phenotypes were observed in other human cohorts and two transgenic murine models (Tnnt1-/- and Tnnt1 c.505G>T). These findings have implications for emerging molecular therapies, including the suitably of TNNT1 gene replacement for newborns with ANM or other TNNT1-associated myopathies.


Assuntos
Debilidade Muscular/genética , Músculo Esquelético/patologia , Miopatias da Nemalina/genética , Troponina T/genética , Amish/genética , Animais , Criança , Códon sem Sentido/genética , Feminino , Homozigoto , Humanos , Recém-Nascido , Masculino , Camundongos , Debilidade Muscular/diagnóstico , Debilidade Muscular/fisiopatologia , Músculo Esquelético/metabolismo , Miopatias da Nemalina/diagnóstico , Miopatias da Nemalina/fisiopatologia , Patologia Molecular , Fenótipo , Isoformas de Proteínas/genética
17.
J Neurol ; 265(3): 542-551, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29356967

RESUMO

Sporadic late-onset nemaline myopathy (SLONM) is a rare acquired myopathy characterized by rapid-onset proximal weakness in late adulthood, and the presence of nemaline bodies on muscle biopsy. In recent years, several therapeutic interventions, including immunomodulating agents and autologous stem cell transplantation, have shown variable degrees of efficacy in different patients, but no consensus has been reached to allow an effective tailoring of treatments in this severe disease. We performed a retrospective evaluation of clinical, pathological, laboratory, muscle MRI, and follow-up data of SLONM patients diagnosed in the period 2010-2015 in our neuromuscular center. Six patients (three males and three females) were identified. Average time elapsed from the onset of symptoms to referral to the neuromuscular specialist was 23.7 months. Monoclonal gammopathy was detectable in five patients. Nemaline bodies were detected in all the patients, and their abundance correlated with clinical severity. Signs of cardiac involvement were present in all the patients to different extents. Muscle MRI showed a preferential involvement of neck extensors, paraspinal, gluteal, hamstring and soleus muscles. All patients were treated with prednisone and repeated courses of intravenous immunoglobulins, and a favorable outcome was reached in five patients. Our experience confirms that SLONM is clinically characterized by subacute proximal and axial muscle weakness. Time to referral was relatively long and should be reduced with increasing awareness of the disease. Muscle MRI could be of help as a diagnostic tool to identify this potentially treatable myopathy. Cardiac evaluation should be warranted in all SLONM patients to detect subclinical heart involvement.


Assuntos
Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/patologia , Miopatias da Nemalina/diagnóstico por imagem , Miopatias da Nemalina/patologia , Adulto , Idoso , Creatina Quinase/sangue , Eletromiografia , Feminino , Seguimentos , Coração/fisiopatologia , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , Miopatias da Nemalina/tratamento farmacológico , Miopatias da Nemalina/fisiopatologia , Condução Nervosa , Encaminhamento e Consulta , Testes de Função Respiratória , Estudos Retrospectivos , Índice de Gravidade de Doença , Fatores de Tempo , Resultado do Tratamento
18.
Hum Mol Genet ; 27(4): 638-648, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29293963

RESUMO

Nemaline myopathy (NM) is a heterogeneous congenital skeletal muscle disease with cytoplasmic rod-like structures (nemaline bodies) in muscle tissue. While weakness in NM is related to contractile abnormalities, myofiber smallness is an additional abnormality in NM that may be treatable. We evaluated the effects of mRK35 (a myostatin inhibitor developed by Pfizer) treatment in the TgACTA1D286G mouse model of NM. mRK35 induced skeletal muscle growth that led to significant increases in animal bodyweight, forelimb grip strength and muscle fiber force, although it should be noted that animal weight and forelimb grip strength in untreated TgACTA1D286G mice was not different from controls. Treatment was also associated with an increase in the number of tubular aggregates found in skeletal muscle. These findings suggest that myostatin inhibition may be useful in promoting muscle growth and strength in Acta1-mutant muscle, while also further establishing the relationship between low levels of myostatin and tubular aggregate formation.


Assuntos
Actinas/metabolismo , Músculo Esquelético/metabolismo , Miopatias da Nemalina/metabolismo , Actinas/genética , Animais , Membro Anterior/metabolismo , Membro Anterior/fisiologia , Força da Mão/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Músculo Esquelético/fisiologia , Miopatias da Nemalina/fisiopatologia , Miostatina/metabolismo
19.
J Neurol Sci ; 383: 61-68, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29246625

RESUMO

Nemaline myopathy (NM), the most common of the congenital myopathies, is caused by various genetic mutations. In this study, we attempted to identify the causative mutations of NM and to reveal any specific genotype-phenotype relationship in Korean patients with this disease. We investigated the clinical features and genotypes in 15 pathologically diagnosed NM patients, using whole exome sequencing (WES) combined with targeted sequencing and array-based comparative genomic hybridization. This strategy revealed pathogenic causative mutations in seven patients (46.7%), among whom mutations in the nebulin gene (NEB) were the most frequent (5 patients, 33.3%). Copy number variation (CNV) abnormality in NEB was not observed in any of our patients. In those with NEB-associated NM, the clinical spectrum was highly variable regardless of the mutation type. However, the majority of patients showing anterior lower leg weakness were associated with mutations located between NEB exons 166 and 177. We concluded that the combination of WES and targeted Sanger sequencing is an effective strategy for analyzing genotypes in patients with NM, and that CNV in NEB may not be a frequent cause of this disease among Koreans.


Assuntos
Miopatias da Nemalina/genética , Miopatias da Nemalina/fisiopatologia , Adolescente , Adulto , Criança , Pré-Escolar , Variações do Número de Cópias de DNA , Feminino , Técnicas de Genotipagem , Humanos , Lactente , Extremidade Inferior/diagnóstico por imagem , Extremidade Inferior/patologia , Extremidade Inferior/fisiopatologia , Masculino , Proteínas Musculares/genética , Debilidade Muscular/diagnóstico por imagem , Debilidade Muscular/genética , Debilidade Muscular/patologia , Debilidade Muscular/fisiopatologia , Mutação , Miopatias da Nemalina/diagnóstico por imagem , Miopatias da Nemalina/patologia , República da Coreia , Sequenciamento do Exoma , Adulto Jovem
20.
Curr Opin Neurol ; 30(5): 457-463, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28678037

RESUMO

PURPOSE OF REVIEW: Sporadic late-onset nemaline myopathy (SLONM) with monoclonal gammopathy of undetermined significance (MGUS) is a rare subacute progressive muscle disease. The prognosis is poor due to severe respiratory insufficiency. Recently, however, autologous stem-cell transplantation following high-dose melphalan has been shown to be effective unless there is delay before the treatment. Therefore, early recognition of the disease is important. This review gives an overview of recent advances in SLONM-MGUS, which could help to understand clinical and pathological features and treatment. RECENT FINDINGS: Efficacy of autologous stem-cell transplantation following high-dose melphalan has been demonstrated in a long-term observation study. Subsequently, reports from other groups also have supported it. Furthermore, efficacy of chemotherapy toward plasma cell dyscrasia without stem-cell transplantation have been reported as well. A few case reports have suggested the presence of cardiac involvement related to SLONM-MGUS. SUMMARY: SLONM-MGUS is now considered as a treatable disease. Antiplasma cell dyscrasia therapy is a promising therapeutic option. Meanwhile, the pathomechanic link between muscle degeneration and monoclonal gammopathy remains unclear and further investigations are warranted.


Assuntos
Gamopatia Monoclonal de Significância Indeterminada/terapia , Miopatias da Nemalina/terapia , Humanos , Gamopatia Monoclonal de Significância Indeterminada/patologia , Gamopatia Monoclonal de Significância Indeterminada/fisiopatologia , Músculo Esquelético/patologia , Miopatias da Nemalina/patologia , Miopatias da Nemalina/fisiopatologia , Transplante de Células-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA