Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Muscle Nerve ; 65(5): 560-567, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35179228

RESUMO

INTRODUCTION/AIMS: Myotonic dystrophy type 1 (DM1) is known to affect cognitive function, but the best methods to assess central nervous system involvement in multicenter studies have not been determined. In this study our primary aim was to evaluate the potential of computerized cognitive tests to assess cognition in DM1. METHODS: We conducted a prospective, longitudinal, observational study of 113 adults with DM1 at six sites. Psychomotor speed, attention, working memory, and executive functioning were assessed at baseline, 3 months, and 12 months using computerized cognitive tests. Results were compared with assessments of muscle function and patient reported outcomes (PROs), including the Myotonic Dystrophy Health Index (MDHI) and the 5-dimension EuroQol (EQ-5D-5L) questionnaire. RESULTS: Based on intraclass correlation coefficients, computerized cognitive tests had moderate to good reliability for psychomotor speed (0.76), attention (0.82), working memory speed (0.79), working memory accuracy (0.65), and executive functioning (0.87). Performance at baseline was lowest for working memory accuracy (P < .0001). Executive function performance improved from baseline to 3 months (P < .0001), without further changes over 1 year. There was a moderate correlation between poorer executive function and larger CTG repeat size (r = -0.433). There were some weak associations between PROs and cognitive performance. DISCUSSION: Computerized tests of cognition are feasible in multicenter studies of DM1. Poor performance was exhibited in working memory, which may be a useful variable in clinical trials. Learning effects may have contributed to the improvement in executive functioning. The relationship between PROs and cognitive impairment in DM1 requires further study.


Assuntos
Distrofia Miotônica , Adulto , Cognição , Computadores , Humanos , Estudos Longitudinais , Distrofia Miotônica/complicações , Distrofia Miotônica/diagnóstico , Estudos Prospectivos , Reprodutibilidade dos Testes
2.
J Neurol Neurosurg Psychiatry ; 92(11): 1186-1196, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34103343

RESUMO

BACKGROUND: We used a multimodal approach including detailed phenotyping, whole exome sequencing (WES) and candidate gene filters to diagnose rare neurological diseases in individuals referred by tertiary neurology centres. METHODS: WES was performed on 66 individuals with neurogenetic diseases using candidate gene filters and stringent algorithms for assessing sequence variants. Pathogenic or likely pathogenic missense variants were interpreted using in silico prediction tools, family segregation analysis, previous publications of disease association and relevant biological assays. RESULTS: Molecular diagnosis was achieved in 39% (n=26) including 59% of childhood-onset cases and 27% of late-onset cases. Overall, 37% (10/27) of myopathy, 41% (9/22) of neuropathy, 22% (2/9) of MND and 63% (5/8) of complex phenotypes were given genetic diagnosis. Twenty-seven disease-associated variants were identified including ten novel variants in FBXO38, LAMA2, MFN2, MYH7, PNPLA6, SH3TC2 and SPTLC1. Single-nucleotide variants (n=10) affected conserved residues within functional domains and previously identified mutation hot-spots. Established pathogenic variants (n=16) presented with atypical features, such as optic neuropathy in adult polyglucosan body disease, facial dysmorphism and skeletal anomalies in cerebrotendinous xanthomatosis, steroid-responsive weakness in congenital myasthenia syndrome 10. Potentially treatable rare diseases were diagnosed, improving the quality of life in some patients. CONCLUSIONS: Integrating deep phenotyping, gene filter algorithms and biological assays increased diagnostic yield of exome sequencing, identified novel pathogenic variants and extended phenotypes of difficult to diagnose rare neurogenetic disorders in an outpatient clinic setting.


Assuntos
Sequenciamento do Exoma , Doenças Genéticas Inatas/diagnóstico , Mutação , Doenças do Sistema Nervoso/diagnóstico , Doenças Raras/diagnóstico , Adolescente , Adulto , Idoso , Doenças Genéticas Inatas/genética , Humanos , Pessoa de Meia-Idade , Técnicas de Diagnóstico Molecular , Doenças do Sistema Nervoso/genética , Linhagem , Fenótipo , Doenças Raras/genética , Adulto Jovem
3.
Acta Neuropathol ; 139(6): 1089-1104, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32236737

RESUMO

RYR1 encodes the type 1 ryanodine receptor, an intracellular calcium release channel (RyR1) on the skeletal muscle sarcoplasmic reticulum (SR). Pathogenic RYR1 variations can destabilize RyR1 leading to calcium leak causing oxidative overload and myopathy. However, the effect of RyR1 leak has not been established in individuals with RYR1-related myopathies (RYR1-RM), a broad spectrum of rare neuromuscular disorders. We sought to determine whether RYR1-RM affected individuals exhibit pathologic, leaky RyR1 and whether variant location in the channel structure can predict pathogenicity. Skeletal muscle biopsies were obtained from 17 individuals with RYR1-RM. Mutant RyR1 from these individuals exhibited pathologic SR calcium leak and increased activity of calcium-activated proteases. The increased calcium leak and protease activity were normalized by ex-vivo treatment with S107, a RyR stabilizing Rycal molecule. Using the cryo-EM structure of RyR1 and a new dataset of > 2200 suspected RYR1-RM affected individuals we developed a method for assigning pathogenicity probabilities to RYR1 variants based on 3D co-localization of known pathogenic variants. This study provides the rationale for a clinical trial testing Rycals in RYR1-RM affected individuals and introduces a predictive tool for investigating the pathogenicity of RYR1 variants of uncertain significance.


Assuntos
Cálcio/metabolismo , Doenças Musculares/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Citoplasma/metabolismo , Humanos , Músculo Esquelético/metabolismo , Doenças Musculares/terapia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Retículo Sarcoplasmático/metabolismo
4.
Muscle Nerve ; 57(5): 749-755, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28981955

RESUMO

INTRODUCTION: This study analyzes and describes atypical presentations of Charcot-Marie-Tooth disease type 4C (CMT4C). METHODS: We present clinical and physiologic features of 5 patients with CMT4C caused by biallelic private mutations of SH3TC2. RESULTS: All patients manifested scoliosis, and nerve conduction study indicated results in the demyelinating range. All patients exhibited signs of motor impairment within the first years of life. We describe 2 or more different genetic diseases in the same patient, atypical presentations of CMT, and 3 new mutations in CMT4C patients. DISCUSSION: A new era of unbiased genetic testing has led to this small case series of individuals with CMT4C and highlights the recognition of different genetic diseases in CMT4C patients for accurate diagnosis, genetic risk identification, and therapeutic intervention. The phenotype of CMT4C, in addition, appears to be enriched by a number of features unusual for the broad CMT category. Muscle Nerve 57: 749-755, 2018.


Assuntos
Doença de Charcot-Marie-Tooth , Mutação/genética , Proteínas/genética , Adolescente , Adulto , Animais , Animais Recém-Nascidos , Doença de Charcot-Marie-Tooth/complicações , Doença de Charcot-Marie-Tooth/diagnóstico , Doença de Charcot-Marie-Tooth/genética , Criança , Doenças Desmielinizantes/etiologia , Feminino , Testes Genéticos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/metabolismo , Escoliose/etiologia
5.
Biochemistry ; 56(14): 2061-2070, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28349680

RESUMO

Z-disc-associated, alternatively spliced, PDZ motif-containing protein (ZASP) is a principal component of the sarcomere. The three prevalent isoforms of ZASP in skeletal muscle are generated by alternative splicing of exons 9 and 10. The long isoforms, either having (ZASP-L) or lacking exon 10 (ZASP-LΔex10), include an N-terminal PDZ domain, an actin-binding region (ABR) with a conserved motif (ZM), and three C-terminal LIM domains. The short isoform (ZASP-S) lacks the LIM domains. Mutations, A147T and A165V, within the ZM of ZASP-LΔex10 cause myofibrillar myopathy, but the mechanism is unknown. We have prepared these proteins, their ABR, and the respective mutant variants in recombinant form, characterized them biophysically, and analyzed their actin-binding properties by surface plasmon resonance and electron microscopy. All the proteins were physically homogeneous and monomeric and had circular dichroic spectra consistent with partially folded conformations. Comparison of the NMR HSQC spectra of ZASP-S and the PDZ domain showed that the ABR is unstructured. ZASP-S and its mutant variants and ZASP-LΔex10 all bound to immobilized G-actin with high affinity (Kd ≈ 10-8 to 10-9 M). Constructs of the isolated actin-binding region missing exon 10 (ABRΔ10) bound with lower affinity (Kd ≈ 10-7 M), but those retaining exon 10 (ABR+10) did so only weakly (Kd ≈ 10-5 M). ZASP-S, and the ABRΔ10, also induced F-actin and array formation, even in conditions of low ionic strength and in the absence of KCl and Mg2+ ions. Interestingly, the ZM mutations A147T and A165V did not affect any of the results described above.


Assuntos
Actinas/química , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas com Domínio LIM/química , Actinas/genética , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Processamento Alternativo , Sítios de Ligação , Escherichia coli/genética , Escherichia coli/metabolismo , Éxons , Expressão Gênica , Humanos , Íntrons , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Mutação , Concentração Osmolar , Ligação Proteica , Domínios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sarcômeros/fisiologia , Relação Estrutura-Atividade
6.
Am J Hum Genet ; 93(5): 976-83, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24207122

RESUMO

Spinal muscular atrophies (SMAs) are a heterogeneous group of inherited disorders characterized by degeneration of anterior horn cells and progressive muscle weakness. In two unrelated families affected by a distinct form of autosomal-dominant distal SMA initially manifesting with calf weakness, we identified by genetic linkage analysis and exome sequencing a heterozygous missense mutation, c.616T>C (p.Cys206Arg), in F-box protein 38 (FBXO38). FBXO38 is a known coactivator of the transcription factor Krüppel-like factor 7 (KLF7), which regulates genes required for neuronal axon outgrowth and repair. The p.Cys206Arg substitution did not alter the subcellular localization of FBXO38 but did impair KLF7-mediated transactivation of a KLF7-responsive promoter construct and endogenous KLF7 target genes in both heterologously expressing human embryonic kidney 293T cells and fibroblasts derived from individuals with the FBXO38 missense mutation. This transcriptional dysregulation was associated with an impairment of neurite outgrowth in primary motor neurons. Together, these results suggest that a transcriptional regulatory pathway that has a well-established role in axonal development could also be critical for neuronal maintenance and highlight the importance of FBXO38 and KLF7 activity in motor neurons.


Assuntos
Proteínas F-Box/genética , Atrofia Muscular Espinal/genética , Mutação de Sentido Incorreto , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Células do Corno Anterior/metabolismo , Células do Corno Anterior/patologia , Axônios/metabolismo , Axônios/patologia , Exoma , Feminino , Fibroblastos/citologia , Fibroblastos/patologia , Ligação Genética , Células HEK293 , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Atrofia Muscular Espinal/patologia , Linhagem , Adulto Jovem
7.
J Biol Chem ; 289(19): 13615-26, 2014 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-24668811

RESUMO

The core of skeletal muscle Z-discs consists of actin filaments from adjacent sarcomeres that are cross-linked by α-actinin homodimers. Z-disc-associated, alternatively spliced, PDZ motif-containing protein (ZASP)/Cypher interacts with α-actinin, myotilin, and other Z-disc proteins via the PDZ domain. However, these interactions are not sufficient to maintain the Z-disc structure. We show that ZASP directly interacts with skeletal actin filaments. The actin-binding domain is between the modular PDZ and LIM domains. This ZASP region is alternatively spliced so that each isoform has unique actin-binding domains. All ZASP isoforms contain the exon 6-encoded ZASP-like motif that is mutated in zaspopathy, a myofibrillar myopathy (MFM), whereas the exon 8-11 junction-encoded peptide is exclusive to the postnatal long ZASP isoform (ZASP-LΔex10). MFM is characterized by disruption of skeletal muscle Z-discs and accumulation of myofibrillar degradation products. Wild-type and mutant ZASP interact with α-actin, α-actinin, and myotilin. Expression of mutant, but not wild-type, ZASP leads to Z-disc disruption and F-actin accumulation in mouse skeletal muscle, as in MFM. Mutations in the actin-binding domain of ZASP-LΔex10, but not other isoforms, cause disruption of the actin cytoskeleton in muscle cells. These isoform-specific mutation effects highlight the essential role of the ZASP-LΔex10 isoform in F-actin organization. Our results show that MFM-associated ZASP mutations in the actin-binding domain have deleterious effects on the core structure of the Z-discs in skeletal muscle.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas com Domínio LIM/metabolismo , Mutação de Sentido Incorreto , Miofibrilas/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/patologia , Actinina/genética , Actinina/metabolismo , Actinas/genética , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Motivos de Aminoácidos , Animais , Linhagem Celular , Conectina/genética , Conectina/metabolismo , Humanos , Proteínas com Domínio LIM/genética , Camundongos , Proteínas dos Microfilamentos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miofibrilas/genética , Miopatias Congênitas Estruturais/genética , Miopatias Congênitas Estruturais/metabolismo , Miopatias Congênitas Estruturais/patologia , Estrutura Terciária de Proteína
8.
Sci Rep ; 13(1): 270, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36609526

RESUMO

LIM domain-binding 3 (LDB3) is a member of the Enigma family of PDZ-LIM proteins. LDB3 has been reported as a striated muscle-specific Z-band alternatively spliced protein that plays an important role in mechanosensory actin cytoskeleton remodeling. This study shows that LDB3 is broadly expressed in the central and peripheral nervous system of human and mouse. LDB3 is predominantly expressed in the adult stages compared to early development and at a significantly higher level in the spinal cord than in the brain. As in skeletal muscle and heart, LDB3 is extensively alternatively spliced in the neurons. Three novel splice isoforms were identified suggesting splicing-dependent regulation of LDB3 expression in the nervous system. Expression of LDB3 in the motor cortex, cerebellum, spinal motor neuron, peripheral nerve, and neuromuscular junction in addition to skeletal muscle indicates important roles for this PDZ-LIM family protein in motor planning and execution. Moreover, expression in the hippocampal neurons suggests roles for LDB3 in learning and memory. LDB3 interactors filamin C and myotilin are also expressed in the spinal motor neuron, nerve, and neuromuscular junction, thereby providing the basis for neurogenic manifestations in myopathies associated with mutations in these so-called muscle proteins.


Assuntos
Proteínas com Domínio LIM , Músculo Estriado , Camundongos , Humanos , Animais , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Músculo Esquelético/metabolismo , Músculo Estriado/metabolismo , Ligação Proteica , Proteínas Musculares/metabolismo , Fatores de Transcrição/metabolismo , Sistema Nervoso/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
9.
Neurobiol Dis ; 45(1): 539-46, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21964252

RESUMO

The genetic basis of oculopharyngeal muscular dystrophy (OPMD) is a short expansion of a polyalanine tract (normal allele: 10 alanines, mutant allele: 11-17 alanines) in the nuclear polyadenylate binding protein PABPN1 which is essential for controlling poly(A) tail length in messenger RNA. Mutant PABPN1 forms nuclear inclusions in OPMD muscle. To investigate the pathogenic role of mutant PABPN1 in vivo, we generated a ligand-inducible transgenic mouse model by using the mifepristone-inducible gene expression system. Induction of ubiquitous expression of mutant PABPN1 resulted in skeletal and cardiac myopathy. Histological changes of degenerative myopathy were preceded by nuclear inclusions of insoluble PABPN1. Downregulation of mutant PABPN1 expression attenuated the myopathy and reduced the nuclear burden of insoluble PABPN1. These results support association between mutant PABPN1 accumulation and degenerative myopathy in mice. Resolution of myopathy in mice suggests that the disease process in OPMD patients may be treatable.


Assuntos
Núcleo Celular/patologia , Músculo Esquelético/patologia , Distrofia Muscular Oculofaríngea/patologia , Alelos , Animais , Núcleo Celular/genética , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Poli A/genética , Poli A/metabolismo
10.
Neuromuscul Disord ; 32(4): 321-331, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35305880

RESUMO

Grip myotonia and weakness are attractive treatment response biomarkers in clinical trials of myotonic dystrophy type 1 (DM1). There is a need to develop simple, patient-friendly and reproducible methods of quantifying grip myotonia in multisite trial settings. We designed a HandClench Relaxometer (HCR) that measures grip myotonia and strength. In contrast with the existing quantitative myometry (QMA) setup, the HCR is portable, economical, can be used with any laptop and generates automated command prompts. We demonstrate the feasibility and reliability of HCR device in twenty DM1 individuals and ten age-matched controls; patients returned for follow up within two months. The device showed excellent day to day reproducibility (ICC >0.80) in patients. The HCR device detected myotonia in milder muscle disease and measured longer myotonia duration than QMA indicating enhanced sensitivity for quantifying myotonia in DM1. The reaction time to the relax but not squeeze command was delayed and showed warm up similar to myotonia in DM1. HCR outcomes were correlated with key pinch strength, hand dexterity test, and fat replacement in the MRI of the long finger flexor muscles. Use of the HCR is warranted for grip myotonia and strength measurements in longitudinal observational and interventional studies of DM1.


Assuntos
Miotonia , Distrofia Miotônica , Eletromiografia , Força da Mão/fisiologia , Humanos , Lactente , Miotonia/diagnóstico , Distrofia Miotônica/diagnóstico , Reprodutibilidade dos Testes
11.
Neurology ; 96(5): e798-e808, 2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33219145

RESUMO

OBJECTIVE: To characterize muscle involvement and evaluate disease severity in patients with GNE myopathy using skeletal muscle MRI and proton magnetic resonance spectroscopy (1H-MRS). METHODS: Skeletal muscle imaging of the lower extremities was performed in 31 patients with genetically confirmed GNE myopathy, including T1-weighted and short tau inversion recovery (STIR) images, T1 and T2 mapping, and 1H-MRS. Measures evaluated included longitudinal relaxation time (T1), transverse relaxation time (T2), and 1H-MRS fat fraction (FF). Thigh muscle volume was correlated with relevant measures of strength, function, and patient-reported outcomes. RESULTS: The cohort was representative of a wide range of disease progression. Contractile thigh muscle volume ranged from 5.51% to 62.95% and correlated with thigh strength (r = 0.91), the 6-minute walk test (r = 0.82), the adult myopathy assessment tool (r = 0.83), the activities-specific balance confidence scale (r = 0.65), and the inclusion body myositis functional rating scale (r = 0.62). Four stages of muscle involvement were distinguished by qualitative (T1W and STIR images) and quantitative methods: stage I: unaffected muscle (T1 = 1,033 ± 74.2 ms, T2 = 40.0 ± 1.9 ms, FF = 7.4 ± 3.5%); stage II: STIR hyperintense muscle with minimal or no fat infiltration (T1 = 1,305 ± 147 ms, T2 = 50.2 ± 3.5 ms, FF = 27.6 ± 12.7%); stage III: fat infiltration and STIR hyperintensity (T1 = 1,209 ± 348 ms, T2 = 73.3 ± 12.6 ms, FF = 57.5 ± 10.6%); and stage IV: complete fat replacement (T1 = 318 ± 39.9 ms, T2 = 114 ± 21.2 ms, FF = 85.6 ± 4.2%). 1H-MRS showed a significant decrease in intramyocellular lipid and trimethylamines between stage I and II, suggesting altered muscle metabolism at early stages. CONCLUSION: MRI biomarkers can monitor muscle involvement and determine disease severity noninvasively in patients with GNE myopathy. CLINICALTRIALSGOV IDENTIFIER: NCT01417533.


Assuntos
Miopatias Distais/diagnóstico por imagem , Metabolismo dos Lipídeos , Força Muscular , Músculo Esquelético/diagnóstico por imagem , Adulto , Idoso , Progressão da Doença , Miopatias Distais/metabolismo , Miopatias Distais/patologia , Miopatias Distais/fisiopatologia , Feminino , Músculos Isquiossurais/diagnóstico por imagem , Músculos Isquiossurais/metabolismo , Músculos Isquiossurais/patologia , Músculos Isquiossurais/fisiopatologia , Humanos , Perna (Membro) , Lipídeos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Complexos Multienzimáticos/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Tamanho do Órgão , Medidas de Resultados Relatados pelo Paciente , Espectroscopia de Prótons por Ressonância Magnética , Músculo Quadríceps/diagnóstico por imagem , Músculo Quadríceps/metabolismo , Músculo Quadríceps/patologia , Índice de Gravidade de Doença , Coxa da Perna , Teste de Caminhada , Adulto Jovem
12.
Commun Biol ; 4(1): 355, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33742095

RESUMO

Mechanical stress induced by contractions constantly threatens the integrity of muscle Z-disc, a crucial force-bearing structure in striated muscle. The PDZ-LIM proteins have been proposed to function as adaptors in transducing mechanical signals to preserve the Z-disc structure, however the underlying mechanisms remain poorly understood. Here, we show that LDB3, a well-characterized striated muscle PDZ-LIM protein, modulates mechanical stress signaling through interactions with the mechanosensing domain in filamin C, its chaperone HSPA8, and PKCα in the Z-disc of skeletal muscle. Studies of Ldb3Ala165Val/+ mice indicate that the myopathy-associated LDB3 p.Ala165Val mutation triggers early aggregation of filamin C and its chaperones at muscle Z-disc before aggregation of the mutant protein. The mutation causes protein aggregation and eventually Z-disc myofibrillar disruption by impairing PKCα and TSC2-mTOR, two important signaling pathways regulating protein stability and disposal of damaged cytoskeletal components at a major mechanosensor hub in the Z-disc of skeletal muscle.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas com Domínio LIM/genética , Mecanotransdução Celular , Músculo Esquelético/enzimologia , Miopatias Congênitas Estruturais/enzimologia , Mutação Puntual , Proteína Quinase C-alfa/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Autofagia , Modelos Animais de Doenças , Regulação para Baixo , Filaminas/metabolismo , Proteínas de Choque Térmico HSC70/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Contração Muscular , Força Muscular , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Miopatias Congênitas Estruturais/genética , Miopatias Congênitas Estruturais/patologia , Miopatias Congênitas Estruturais/fisiopatologia , Agregados Proteicos , Agregação Patológica de Proteínas , Proteína Quinase C-alfa/genética , Serina-Treonina Quinases TOR/genética , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo
13.
Neuromuscul Disord ; 30(9): 742-749, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32819793

RESUMO

Patients with bi-allelic loss-of-function mutations in the gene ANO5 most commonly present with muscular dystrophy. In some studies, patients with ANO5-related dystrophy (ANO5-RD) had evidence of mild cardiac abnormalities; however, cardiac magnetic resonance imaging (MRI) has not been used for myocardial characterization. Ten patients with genetically confirmed ANO5-RD were enrolled in a phenotyping study to better characterize cardiac involvement. Evaluations included medical history, neurological examination and cardiac evaluations (electrocardiogram, echocardiogram and cardiac MRI). All patients were clinically asymptomatic from a cardiac perspective. Muscle MRI was consistent with previous studies of ANO5-RD with increased T1 signal in the posterior and medial compartments of the upper leg and the posterior compartment of the lower leg. Cardiac studies using echocardiography and cardiac MRI revealed dilation of the aortic root and thickening of the aortic valve without significant stenosis in 3/10 patients. There was evidence of abnormal late gadolinium enhancement (LGE) on cardiac MRI in 2/10 patients. In ANO5-RD, the development of cardiac fibrosis, edema or inflammation as demonstrated by LGE has not yet been reported. Cardiac MRI can characterize cardiac tissue and may detect subtle changes before they appear on echocardiography, with potential prognostic implications.


Assuntos
Meios de Contraste/farmacologia , Gadolínio/metabolismo , Imagem Cinética por Ressonância Magnética/métodos , Imageamento por Ressonância Magnética , Anoctaminas/genética , Cardiomiopatias/classificação , Cardiomiopatias/patologia , Eletrocardiografia , Feminino , Coração/fisiopatologia , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Miocárdio/patologia
14.
J Neuromuscul Dis ; 7(2): 153-166, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32039858

RESUMO

BACKGROUND: Extensive genetic screening results in the identification of thousands of rare variants that are difficult to interpret. Because of its sheer size, rare variants in the titin gene (TTN) are detected frequently in any individual. Unambiguous interpretation of molecular findings is almost impossible in many patients with myopathies or cardiomyopathies. OBJECTIVE: To refine the current classification framework for TTN-associated skeletal muscle disorders and standardize the interpretation of TTN variants. METHODS: We used the guidelines issued by the American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology (AMP) to re-analyze TTN genetic findings from our patient cohort. RESULTS: We identified in the classification guidelines three rules that are not applicable to titin-related skeletal muscle disorders; six rules that require disease-/gene-specific adjustments and four rules requiring quantitative thresholds for a proper use. In three cases, the rule strength need to be modified. CONCLUSIONS: We suggest adjustments are made to the guidelines. We provide frequency thresholds to facilitate filtering of candidate causative variants and guidance for the use and interpretation of functional data and co-segregation evidence. We expect that the variant classification framework for TTN-related skeletal muscle disorders will be further improved along with a better understanding of these diseases.


Assuntos
Cardiomiopatias , Conectina/genética , Doenças Musculares , Guias de Prática Clínica como Assunto/normas , Cardiomiopatias/classificação , Cardiomiopatias/congênito , Cardiomiopatias/genética , Humanos , Doenças Musculares/classificação , Doenças Musculares/congênito , Doenças Musculares/genética
15.
J Gen Physiol ; 129(1): 79-94, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17158949

RESUMO

Muscle degeneration and myotonia are clinical hallmarks of myotonic dystrophy type 1 (DM1), a multisystemic disorder caused by a CTG repeat expansion in the 3' untranslated region of the myotonic dystrophy protein kinase (DMPK) gene. Transgenic mice engineered to express mRNA with expanded (CUG)(250) repeats (HSA(LR) mice) exhibit prominent myotonia and altered splicing of muscle chloride channel gene (Clcn1) transcripts. We used whole-cell patch clamp recordings and nonstationary noise analysis to compare and biophysically characterize the magnitude, kinetics, voltage dependence, and single channel properties of the skeletal muscle chloride channel (ClC-1) in individual flexor digitorum brevis (FDB) muscle fibers isolated from 1-3-wk-old wild-type and HSA(LR) mice. The results indicate that peak ClC-1 current density at -140 mV is reduced >70% (-48.5 +/- 3.6 and -14.0 +/- 1.6 pA/pF, respectively) and the kinetics of channel deactivation increased in FDB fibers obtained from 18-20- d-old HSA(LR) mice. Nonstationary noise analysis revealed that the reduction in ClC-1 current density in HSA(LR) FDB fibers results from a large reduction in ClC-1 channel density (170 +/- 21 and 58 +/- 11 channels/pF in control and HSA(LR) fibers, respectively) and a modest decrease in maximal channel open probability(0.91 +/- 0.01 and 0.75 +/- 0.03, respectively). Qualitatively similar results were observed for ClC-1 channel activity in knockout mice for muscleblind-like 1 (Mbnl1(DeltaE3/DeltaE3)), a second murine model of DM1 that exhibits prominent myotonia and altered Clcn1 splicing (Kanadia et al., 2003). These results support a molecular mechanism for myotonia in DM1 in which a reduction in both the number of functional sarcolemmal ClC-1 and maximal channel open probability, as well as an acceleration in the kinetics of channel deactivation, results from CUG repeat-containing mRNA molecules sequestering Mbnl1 proteins required for proper CLCN1 pre-mRNA splicing and chloride channel function.


Assuntos
Canais de Cloreto/genética , Canais de Cloreto/fisiologia , Distrofia Miotônica/genética , Distrofia Miotônica/fisiopatologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiologia , Técnicas de Patch-Clamp , Splicing de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Neurol India ; 56(3): 298-304, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18974556

RESUMO

Myotonia reflects a state of muscle fiber hyperexcitability. Impaired transmembrane conductance of either chloride or sodium ions results in myotonia. Myotonic disorders include the myotonic dystrophies and nondystrophic myotonias. Mutations in the genes encoding chloride (ClC-1) or sodium (SCN4A) channels expressed exclusively in skeletal muscle cause nondystrophic myotonias. Genetic defects in the myotonic dystrophies do not involve ion channel or its regulator proteins. Recent research supports a novel RNA-mediated disease mechanism of myotonia in the myotonic dystrophies. Myotonic dystrophy Type 1 is caused by CTG repeat expansion in the 3' untranslated region in the Dystrophia Myotonica Protein Kinase (DMPK) gene. Myotonic dystrophy Type 2 is caused by CCTG repeat expansion in the first intron in Zinc Finger Protein 9 (ZNF9) gene. The expanded repeat is transcribed in RNA and forms discrete inclusions in nucleus in both types of myotonic dystrophies. Mutant RNA sequesters MBNL1, a splice regulator protein and depletes MBNL1 from the nucleoplasm. Loss of MBNL1 results in altered splicing of ClC-1 mRNA. Altered splice products do not encode functional ClC-1 protein. Subsequent loss of chloride conductance in muscle membrane causes myotonia in the myotonic dystrophies. The purpose of this review is to discuss the clinical presentation, recent advances in understanding the disease mechanism with particular emphasis on myotonic dystrophies and potential therapy options in myotonic disorders.


Assuntos
Transtornos Miotônicos/genética , Transtornos Miotônicos/fisiopatologia , Canais de Cloreto/genética , Eletromiografia , Humanos , Mutação/genética , Transtornos Miotônicos/diagnóstico , Transtornos Miotônicos/terapia , Canal de Sódio Disparado por Voltagem NAV1.4 , Canais de Sódio/genética , Expansão das Repetições de Trinucleotídeos/genética
17.
Front Neurol ; 9: 118, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29556213

RESUMO

The ryanodine receptor 1-related congenital myopathies (RYR1-RM) comprise a spectrum of slow, rare neuromuscular diseases. Affected individuals present with a mild-to-severe symptomatology ranging from proximal muscle weakness, hypotonia and joint contractures to scoliosis, ophthalmoplegia, and respiratory involvement. Although there is currently no FDA-approved treatment for RYR1-RM, our group recently conducted the first clinical trial in this patient population (NCT02362425). This study aimed to characterize novel RYR1 variants with regard to genetic, laboratory, muscle magnetic resonance imaging (MRI), and clinical findings. Genetic and histopathology reports were obtained from participant's medical records. Alamut Visual Software was used to determine if participant's variants had been previously reported and to assess predicted pathogenicity. Physical exams, pulmonary function tests, T1-weighted muscle MRI scans, and blood measures were completed during the abovementioned clinical trial. Six novel variants (two de novo, three dominant, and one recessive) were identified in individuals with RYR1-RM. Consistent with established RYR1-RM histopathology, cores were observed in all biopsies, except Case 6 who exhibited fiber-type disproportion. Muscle atrophy and impaired mobility with Trendelenburg gait were the most common clinical symptoms and were identified in all cases. Muscle MRI revealed substantial inter-individual variation in fatty infiltration corroborating the heterogeneity of the disease. Two individuals with dominant RYR1 variants exhibited respiratory insufficiency: a clinical symptom more commonly associated with recessive RYR1-RM cases. This study demonstrates that a genetics-led approach is suitable for the diagnosis of suspected RYR1-RM which can be corroborated through histopathology, muscle MRI and clinical examination.

18.
Neurol Clin Pract ; 8(6): 507-520, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30588381

RESUMO

PURPOSE OF REVIEW: Myotonic dystrophy type 1 (DM1) is a severe, progressive genetic disease that affects between 1 in 3,000 and 8,000 individuals globally. No evidence-based guideline exists to inform the care of these patients, and most do not have access to multidisciplinary care centers staffed by experienced professionals, creating a clinical care deficit. RECENT FINDINGS: The Myotonic Dystrophy Foundation (MDF) recruited 66 international clinicians experienced in DM1 patient care to develop consensus-based care recommendations. MDF created a 2-step methodology for the project using elements of the Single Text Procedure and the Nominal Group Technique. The process generated a 4-page Quick Reference Guide and a comprehensive, 55-page document that provides clinical care recommendations for 19 discrete body systems and/or care considerations. SUMMARY: The resulting recommendations are intended to help standardize and elevate care for this patient population and reduce variability in clinical trial and study environments.

19.
Circ Res ; 97(11): 1152-5, 2005 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-16254211

RESUMO

The disease mechanism underlying myotonic dystrophy type 1 (DM1) pathogenesis in skeletal muscle may involve sequestration of RNA binding proteins in nuclear foci of expanded poly(CUG) RNA. Here we report evidence for a parallel mechanism in the heart. Accumulation of expanded poly(CUG) RNA in nuclear foci is associated with sequestration of muscleblind proteins and abnormal regulation of alternative splicing in DM1 cardiac muscle. A toxic effect of RNA with an expanded repeat may contribute to cardiac disease in DM1.


Assuntos
Núcleo Celular/metabolismo , Miócitos Cardíacos/metabolismo , Distrofia Miotônica/genética , RNA Nuclear/metabolismo , Proteínas de Ligação a RNA/metabolismo , Expansão das Repetições de Trinucleotídeos , Processamento Alternativo , Humanos , Pessoa de Meia-Idade , Proteínas Musculares/genética , Miotonina Proteína Quinase , Canal de Sódio Disparado por Voltagem NAV1.5 , Proteínas Serina-Treonina Quinases/genética , Proteínas de Ligação a RNA/análise , Canais de Sódio/genética
20.
J Med Imaging (Bellingham) ; 4(4): 041310, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29226176

RESUMO

Duchenne muscular dystrophy (DMD) is a childhood-onset neuromuscular disease that results in the degeneration of muscle, starting in the extremities, before progressing to more vital areas, such as the lungs. Respiratory failure and pneumonia due to respiratory muscle weakness lead to hospitalization and early mortality. However, tracking the disease in this region can be difficult, as current methods are based on breathing tests and are incapable of distinguishing between muscle involvements. Cine MRI scans give insight into respiratory muscle movements, but the images suffer due to low spatial resolution and poor signal-to-noise ratio. Thus, a robust lung segmentation method is required for accurate analysis of the lung and respiratory muscle movement. We deployed a deep learning approach that utilizes sequence-specific prior information to assist the segmentation of lung in cine MRI. More specifically, we adopt a holistically nested network to conduct image-to-image holistic training and prediction. One frame of the cine MRI is used in the training and applied to the remainder of the sequence ([Formula: see text] frames). We applied this method to cine MRIs of the lung in the axial, sagittal, and coronal planes. Characteristic lung motion patterns during the breathing cycle were then derived from the segmentations and used for diagnosis. Our data set consisted of 31 young boys, age [Formula: see text] years, 15 of whom suffered from DMD. The remaining 16 subjects were age-matched healthy volunteers. For validation, slices from inspiratory and expiratory cycles were manually segmented and compared with results obtained from our method. The Dice similarity coefficient for the deep learning-based method was [Formula: see text] for the sagittal view, [Formula: see text] for the axial view, and [Formula: see text] for the coronal view. The holistic neural network approach was compared with an approach using Demon's registration and showed superior performance. These results suggest that the deep learning-based method reliably and accurately segments the lung across the breathing cycle.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA