Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Circ Res ; 135(5): 554-574, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39011635

RESUMO

BACKGROUND: Cardiac hypertrophy compensates for increased biomechanical stress of the heart induced by prevalent cardiovascular pathologies but can result in heart failure if left untreated. Here, we hypothesized that the membrane fusion and repair protein dysferlin is critical for the integrity of the transverse-axial tubule (TAT) network inside cardiomyocytes and contributes to the proliferation of TAT endomembranes during pressure overload-induced cardiac hypertrophy. METHODS: Stimulated emission depletion and electron microscopy were used to localize dysferlin in mouse and human cardiomyocytes. Data-independent acquisition mass spectrometry revealed the cardiac dysferlin interactome and proteomic changes of the heart in dysferlin-knockout mice. After transverse aortic constriction, we compared the hypertrophic response of wild-type versus dysferlin-knockout hearts and studied TAT network remodeling mechanisms inside cardiomyocytes by live-cell membrane imaging. RESULTS: We localized dysferlin in a vesicular compartment in nanometric proximity to contact sites of the TAT network with the sarcoplasmic reticulum, a.k.a. junctional complexes for Ca2+-induced Ca2+ release. Interactome analyses demonstrated a novel protein interaction of dysferlin with the membrane-tethering sarcoplasmic reticulum protein juncophilin-2, a putative interactor of L-type Ca2+ channels and ryanodine receptor Ca2+ release channels in junctional complexes. Although the dysferlin-knockout caused a mild progressive phenotype of dilated cardiomyopathy, global proteome analysis revealed changes preceding systolic failure. Following transverse aortic constriction, dysferlin protein expression was significantly increased in hypertrophied wild-type myocardium, while dysferlin-knockout animals presented markedly reduced left-ventricular hypertrophy. Live-cell membrane imaging showed a profound reorganization of the TAT network in wild-type left-ventricular myocytes after transverse aortic constriction with robust proliferation of axial tubules, which critically depended on the increased expression of dysferlin within newly emerging tubule components. CONCLUSIONS: Dysferlin represents a new molecular target in cardiac disease that protects the integrity of tubule-sarcoplasmic reticulum junctional complexes for regulated excitation-contraction coupling and controls TAT network reorganization and tubular membrane proliferation in cardiomyocyte hypertrophy induced by pressure overload.


Assuntos
Cardiomegalia , Disferlina , Camundongos Knockout , Miócitos Cardíacos , Retículo Sarcoplasmático , Animais , Disferlina/metabolismo , Disferlina/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/genética , Cardiomegalia/fisiopatologia , Humanos , Camundongos , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/patologia , Camundongos Endogâmicos C57BL , Masculino , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proliferação de Células , Células Cultivadas , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Quinase de Cadeia Leve de Miosina
2.
J Physiol ; 602(9): 1893-1910, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38615232

RESUMO

Dysferlin is a 237 kDa membrane-associated protein characterised by multiple C2 domains with a diverse role in skeletal and cardiac muscle physiology. Mutations in DYSF are known to cause various types of human muscular dystrophies, known collectively as dysferlinopathies, with some patients developing cardiomyopathy. A myriad of in vitro membrane repair studies suggest that dysferlin plays an integral role in the membrane repair complex in skeletal muscle. In comparison, less is known about dysferlin in the heart, but mounting evidence suggests that dysferlin's role is similar in both muscle types. Recent findings have shown that dysferlin regulates Ca2+ handling in striated muscle via multiple mechanisms and that this becomes more important in conditions of stress. Maintenance of the transverse (t)-tubule network and the tight coordination of excitation-contraction coupling are essential for muscle contractility. Dysferlin regulates the maintenance and repair of t-tubules, and it is suspected that dysferlin regulates t-tubules and sarcolemmal repair through a similar mechanism. This review focuses on the emerging complexity of dysferlin's activity in striated muscle. Such insights will progress our understanding of the proteins and pathways that regulate basic heart and skeletal muscle function and help guide research into striated muscle pathology, especially that which arises due to dysferlin dysfunction.


Assuntos
Cálcio , Disferlina , Humanos , Cálcio/metabolismo , Disferlina/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Músculo Estriado/metabolismo , Músculo Estriado/fisiologia
3.
Lipids Health Dis ; 23(1): 247, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39138561

RESUMO

BACKGROUND: Dysferlin-deficient limb-girdle muscular dystrophy type 2B (Dysf) mice are notorious for their mild phenotype. Raising plasma total cholesterol (CHOL) via apolipoprotein E (ApoE) knockout (KO) drastically exacerbates muscle wasting in Dysf mice. However, dysferlinopathic patients have abnormally reduced plasma high-density lipoprotein cholesterol (HDL-C) levels. The current study aimed to determine whether HDL-C lowering can exacerbate the mild phenotype of dysferlin-null mice. METHODS: Human cholesteryl ester transfer protein (CETP), a plasma lipid transfer protein not found in mice that reduces HDL-C, and/or its optimal adapter protein human apolipoprotein B (ApoB), were overexpressed in Dysf mice. Mice received a 2% cholesterol diet from 2 months of age and characterized through ambulatory and hanging functional tests, plasma analyses, and muscle histology. RESULTS: CETP/ApoB expression in Dysf mice caused reduced HDL-C (54.5%) and elevated ratio of CHOL/HDL-C (181.3%) compared to control Dysf mice in plasma, but without raising CHOL. Compared to the severe muscle pathology found in high CHOL Dysf/ApoE double knockout mice, Dysf/CETP/ApoB mice did not show significant changes in ambulation, hanging capacity, increases in damaged area, collagen deposition, or decreases in cross-sectional area and healthy myofibre coverage. CONCLUSIONS: CETP/ApoB over-expression in Dysf mice decreases HDL-C without increasing CHOL or exacerbating muscle pathology. High CHOL or nonHDL-C caused by ApoE KO, rather than low HDL-C, likely lead to rodent muscular dystrophy phenotype humanization.


Assuntos
Apolipoproteínas E , Proteínas de Transferência de Ésteres de Colesterol , HDL-Colesterol , Disferlina , Camundongos Knockout , Distrofia Muscular do Cíngulo dos Membros , Animais , Proteínas de Transferência de Ésteres de Colesterol/genética , Proteínas de Transferência de Ésteres de Colesterol/deficiência , Disferlina/genética , Disferlina/deficiência , HDL-Colesterol/sangue , Camundongos , Apolipoproteínas E/genética , Apolipoproteínas E/deficiência , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/patologia , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/genética , Atrofia Muscular/patologia , Atrofia Muscular/metabolismo , Masculino , Apolipoproteínas B/sangue , Apolipoproteínas B/genética , Modelos Animais de Doenças
4.
BMC Musculoskelet Disord ; 25(1): 241, 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38539162

RESUMO

BACKGROUND: Limb girdle muscular dystrophies (LGMDs) constitute a heterogeneous group of neuromuscular disorders with a very variable clinical presentation and overlapping traits. The clinical symptoms of LGMD typically appear in adolescence or early adulthood. Genetic variation in the dysferlin gene (DYSF) has been associated with LGMD. METHODS: We characterized a recessive LGMD in a young adult from consanguineous Irani families using whole-exome sequencing (WES) technology. Sanger sequencing was performed to verify the identified variant. Computational modeling and protein-protein docking were used to investigate the impact of the variant on the structure and function of the DYSF protein. RESULTS: By WES, we identified a novel homozygous missense variant in DYSF (NM_003494.4: c.5876T > C: p. Leu1959Pro) previously been associated with LGMD phenotypes. CONCLUSIONS: The identification and validation of new pathogenic DYSF variant in the present study further highlight the importance of this gene in LGMD.


Assuntos
Distrofia Muscular do Cíngulo dos Membros , Adulto , Humanos , Adulto Jovem , Disferlina/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Mutação de Sentido Incorreto , Fenótipo
5.
Curr Genomics ; 24(5): 330-335, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-38235354

RESUMO

Background: Dysferlinopathies represent a group of limb girdle or distal muscular dystrophies with an autosomal-recessive inheritance pattern resulting from the presence of pathogenic variants in the dysferlin gene (DYSF). Objective: In this work, we describe a population from a small city in Brazil carrying the c.5979dupA pathogenic variant of DYSF responsible for limb girdle muscular dystrophy type 2R and distal muscular dystrophy. Methods: Genotyping analyses were performed by qPCR using customized probe complementary to the region with the duplication under analysis in the DYSF. Results: A total of 104 individuals were examined. c.5979dupA was identified in 48 (46.15%) individuals. Twenty-three (22%) were homozygotes, among whom 13 (56.5%) were female. A total of 91.3% (21) of homozygous individuals had a positive family history, and seven (30.4%) reported consanguineous marriages. Twenty-five (24%) individuals were heterozygous (25.8±16 years) for the same variant, among whom 15 (60%) were female. The mean CK level was 697 IU for homozygotes, 140.5 IU for heterozygotes and 176 IU for wild-type homo-zygotes. The weakness distribution pattern showed 17.3% of individuals with a proximal pattern, 13% with a distal pattern and 69.6% with a mixed pattern. Fatigue was present in 15 homozygotes and one heterozygote. Conclusion: The high prevalence of this variant in individuals from this small community can be explained by a possible founder effect associated with historical, geographical and cultural aspects.

6.
Matrix Biol ; 129: 44-58, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38582404

RESUMO

Extracellular matrix (ECM) pathologic remodeling underlies many disorders, including muscular dystrophy. Tissue decellularization removes cellular components while leaving behind ECM components. We generated "on-slide" decellularized tissue slices from genetically distinct dystrophic mouse models. The ECM of dystrophin- and sarcoglycan-deficient muscles had marked thrombospondin 4 deposition, while dysferlin-deficient muscle had excess decorin. Annexins A2 and A6 were present on all dystrophic decellularized ECMs, but annexin matrix deposition was excessive in dysferlin-deficient muscular dystrophy. Muscle-directed viral expression of annexin A6 resulted in annexin A6 in the ECM. C2C12 myoblasts seeded onto decellularized matrices displayed differential myoblast mobility and fusion. Dystrophin-deficient decellularized matrices inhibited myoblast mobility, while dysferlin-deficient decellularized matrices enhanced myoblast movement and differentiation. Myoblasts treated with recombinant annexin A6 increased mobility and fusion like that seen on dysferlin-deficient decellularized matrix and demonstrated upregulation of ECM and muscle cell differentiation genes. These findings demonstrate specific fibrotic signatures elicit effects on myoblast activity.


Assuntos
Diferenciação Celular , Movimento Celular , Disferlina , Matriz Extracelular , Mioblastos , Sarcoglicanas , Animais , Mioblastos/metabolismo , Mioblastos/citologia , Matriz Extracelular/metabolismo , Camundongos , Sarcoglicanas/genética , Sarcoglicanas/metabolismo , Disferlina/genética , Disferlina/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Distrofina/genética , Distrofina/metabolismo , Anexina A2/genética , Anexina A2/metabolismo , Decorina/genética , Decorina/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Músculo Esquelético/metabolismo
7.
Sci Rep ; 14(1): 19267, 2024 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-39164335

RESUMO

Miyoshi myopathy/dysferlinopathy (MMD) is a rare muscle disease caused by DYSF gene mutations. Apart from skeletal muscles, DYSF is also expressed in the brain. However, the impact of MMD-causing DYSF variants on brain structure and function remains unexplored. To investigate this, we utilized magnetic resonance (MR) modalities (MR volumetry and 31P MR spectroscopy) in a family with seven children, four of whom have the illness. The MMD siblings showed distinct differences from healthy controls: (1) a significant (p < 0.001) right-sided volume asymmetry (+ 232 mm3) of the inferior lateral ventricles; and (2) a significant (p < 0.001) decrease in [Mg2+], along with a modified energy metabolism profile and altered membrane turnover in the hippocampus and motor and premotor cortices. The patients' [Mg2+], energy metabolism, and membrane turnover measures returned to those of healthy relatives after a month of 400 mg/day magnesium supplementation. This work is the first to describe anatomical and functional abnormalities characteristic of neurodegeneration in the MMD brain. Therefore, we call for further examination of brain functions in larger cohorts of MMD patients and testing of magnesium supplementation, which has proven to be an effective corrective approach in our study.


Assuntos
Encéfalo , Magnésio , Humanos , Masculino , Feminino , Criança , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Magnésio/metabolismo , Disferlina/metabolismo , Disferlina/genética , Imageamento por Ressonância Magnética , Metabolismo Energético , Adolescente , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Distrofia Muscular do Cíngulo dos Membros/patologia , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Espectroscopia de Ressonância Magnética , Adulto , Atrofia Muscular , Miopatias Distais
8.
Adv Sci (Weinh) ; 11(31): e2400188, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38887849

RESUMO

Dysferlin is a multi-functional protein that regulates membrane resealing, calcium homeostasis, and lipid metabolism in skeletal muscle. Genetic loss of dysferlin results in limb girdle muscular dystrophy 2B/2R (LGMD2B/2R) and other dysferlinopathies - rare untreatable muscle diseases that lead to permanent loss of ambulation in humans. The mild disease severity in dysferlin-deficient mice and diverse genotype-phenotype relationships in LGMD2B patients have prompted the development of new in vitro models for personalized studies of dysferlinopathy. Here the first 3-D tissue-engineered hiPSC-derived skeletal muscle ("myobundle") model of LGMD2B is described that exhibits compromised contractile function, calcium-handling, and membrane repair, and transcriptomic changes indicative of impaired oxidative metabolism and mitochondrial dysfunction. In response to the fatty acid (FA) challenge, LGMD2B myobundles display mitochondrial deficits and intracellular lipid droplet (LD) accumulation. Treatment with the ryanodine receptor (RyR) inhibitor dantrolene or the dissociative glucocorticoid vamorolone restores LGMD2B contractility, improves membrane repair, and reduces LD accumulation. Lastly, it is demonstrated that chemically induced chronic RyR leak in healthy myobundles phenocopies LGMD2B contractile and metabolic deficit, but not the loss of membrane repair capacity. Together, these results implicate intramyocellular Ca2+ leak as a critical driver of dysferlinopathic phenotype and validate the myobundle system as a platform to study LGMD2B pathogenesis.


Assuntos
Cálcio , Músculo Esquelético , Distrofia Muscular do Cíngulo dos Membros , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Humanos , Cálcio/metabolismo , Músculo Esquelético/metabolismo , Contração Muscular/genética , Disferlina/genética , Disferlina/metabolismo , Animais
9.
Skelet Muscle ; 14(1): 19, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39123261

RESUMO

BACKGROUND: Dysferlinopathies are a clinically heterogeneous group of muscular dystrophies caused by gene mutations resulting in deficiency of the membrane-associated protein dysferlin. They manifest post-growth and are characterised by muscle wasting (primarily in the limb and limb-gridle muscles), inflammation, and replacement of myofibres with adipose tissue. The precise pathomechanism for dysferlinopathy is currently unclear; as such there are no treatments currently available. Glucocorticoids (GCs) are widely used to reduce inflammation and treat muscular dystrophies, but when administered to patients with dysferlinopathy, they have unexpected adverse effects, with accelerated loss of muscle strength. METHODS: To investigate the mechanistic basis for the adverse effects of GCs in dysferlinopathy, the potent GC dexamethasone (Dex) was administered for 4-5 weeks (0.5-0.75 µg/mL in drinking water) to dysferlin-deficient BLA/J and normal wild-type (WT) male mice, sampled at 5 (Study 1) or 10 months (Study 2) of age. A wide range of analyses were conducted. Metabolism- and immune-related gene expression was assessed in psoas muscles at both ages and in quadriceps at 10 months of age. For the 10-month-old mice, quadriceps and psoas muscle histology was assessed. Additionally, we investigated the impact of Dex on the predominantly slow and fast-twitch soleus and extensor digitorum longus (EDL) muscles (respectively) in terms of contractile function, myofibre-type composition, and levels of proteins related to contractile function and metabolism, plus glycogen. RESULTS: At both ages, many complement-related genes were highly expressed in BLA/J muscles, and WT mice were generally more responsive to Dex than BLA/J. The effects of Dex on BLA/J mice included (i) increased expression of inflammasome-related genes in muscles (at 5 months) and (ii) exacerbated histopathology of quadriceps and psoas muscles at 10 months. A novel observation was pronounced staining for glycogen in many myofibres of the damaged quadriceps muscles, with large pale vacuolated myofibres, suggesting possible myofibre death by oncosis. CONCLUSION: These pilot studies provide a new focus for further investigation into the adverse effects of GCs on dysferlinopathic muscles.


Assuntos
Dexametasona , Disferlina , Glucocorticoides , Músculo Esquelético , Distrofia Muscular do Cíngulo dos Membros , Animais , Disferlina/genética , Disferlina/metabolismo , Dexametasona/efeitos adversos , Dexametasona/farmacologia , Masculino , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Distrofia Muscular do Cíngulo dos Membros/patologia , Glucocorticoides/efeitos adversos , Projetos Piloto , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Animais de Doenças , Força Muscular/efeitos dos fármacos
10.
Mol Ther Methods Clin Dev ; 32(2): 101257, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38779337

RESUMO

Mutations in the DYSF gene, encoding the protein dysferlin, lead to several forms of muscular dystrophy. In healthy skeletal muscle, dysferlin concentrates in the transverse tubules and is involved in repairing the sarcolemma and stabilizing Ca2+ signaling after membrane disruption. The DYSF gene encodes 7-8 C2 domains, several Fer and Dysf domains, and a C-terminal transmembrane sequence. Because its coding sequence is too large to package in adeno-associated virus, the full-length sequence is not amenable to current gene delivery methods. Thus, we have examined smaller versions of dysferlin, termed "nanodysferlins," designed to eliminate several C2 domains, specifically C2 domains D, E, and F; B, D, and E; and B, D, E, and F. We also generated a variant by replacing eight amino acids in C2G in the nanodysferlin missing domains D through F. We electroporated dysferlin-null A/J mouse myofibers with Venus fusion constructs of these variants, or as untagged nanodysferlins together with GFP, to mark transfected fibers We found that, although these nanodysferlins failed to concentrate in transverse tubules, three of them supported membrane repair after laser wounding while all four bound the membrane repair protein, TRIM72/MG53, similar to WT dysferlin. By contrast, they failed to suppress Ca2+ waves after myofibers were injured by mild hypoosmotic shock. Our results suggest that the internal C2 domains of dysferlin are required for normal t-tubule localization and Ca2+ signaling and that membrane repair does not require these C2 domains.

11.
Biomolecules ; 14(3)2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38540676

RESUMO

Dysferlinopathies refer to a spectrum of muscular dystrophies that cause progressive muscle weakness and degeneration. They are caused by mutations in the DYSF gene, which encodes the dysferlin protein that is crucial for repairing muscle membranes. This review delves into the clinical spectra of dysferlinopathies, their molecular mechanisms, and the spectrum of emerging therapeutic strategies. We examine the phenotypic heterogeneity of dysferlinopathies, highlighting the incomplete understanding of genotype-phenotype correlations and discussing the implications of various DYSF mutations. In addition, we explore the potential of symptomatic, pharmacological, molecular, and genetic therapies in mitigating the disease's progression. We also consider the roles of diet and metabolism in managing dysferlinopathies, as well as the impact of clinical trials on treatment paradigms. Furthermore, we examine the utility of animal models in elucidating disease mechanisms. By culminating the complexities inherent in dysferlinopathies, this write up emphasizes the need for multidisciplinary approaches, precision medicine, and extensive collaboration in research and clinical trial design to advance our understanding and treatment of these challenging disorders.


Assuntos
Distrofia Muscular do Cíngulo dos Membros , Distrofias Musculares , Animais , Proteínas Musculares/genética , Proteínas de Membrana/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Distrofias Musculares/genética , Mutação
12.
Physiol Rep ; 11(24): e15887, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38110300

RESUMO

Muscular dystrophy (MD) is a genetic disorder that causes progressive muscle weakness and degeneration. Limb-girdle muscular dystrophy (LGMD) is a type of MD that mainly causes muscle atrophy within the shoulder and pelvic girdles. LGMD is classified into autosomal dominant (LGMD-D) and autosomal recessive (LGMD-R) inheritance patterns. Mutations in the Dysferlin gene (DYSF) are common causes of LGMD-R. However, genetic screening of DYSF mutations is rare in Taiwan. Herein, we identified a novel c.2867_2871del ACCAG deletion and a previously reported c.937+1G>A mutation in DYSF from a Taiwanese family with LGMD. The primary symptoms of both siblings were difficulty climbing stairs, walking on the toes, and gradually worsening weakness in the proximal muscles and increased creatine kinase level. Through pedigree analysis and sequencing, two siblings from this family were found to have compound heterozygous DYSF mutations (c. 937+1G>A and c. 2867_2871del ACCAG) within the separated alleles. These mutations induced early stop codons; if translated, truncated DYSF proteins will be expressed. Or, the mRNA products of these two mutations will merit the nonsense-mediated decay, might result in no dysferlin protein expressed. To our knowledge, this is the first report of a novel c.2867_2871del ACCAG deletion in DYSF. Further research is required to examine the effects of the novel DYSF mutation in Taiwanese patients with LGMD.


Assuntos
Distrofia Muscular do Cíngulo dos Membros , Humanos , Disferlina/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Atrofia Muscular , Padrões de Herança
13.
Chinese Journal of Neurology ; (12): 573-581, 2019.
Artigo em Chinês | WPRIM | ID: wpr-756039

RESUMO

Limb girdle muscular dystrophy (LGMD) is characterized by progressive proximal muscle weakness with high genetic heterogeneity.LGMD is the fourth prevalent form of muscular dystrophies in the adult neurology department.Since most patients are juvenile-or adult-onset and present as limb muscle weakness,it would be easily misdiagnosed as myositis or metabolic myopathies.The final diagnosis depends on muscle immunohistochemical staining,Western blotting and genetic screening.In China,LGMD2B and LGMD2A are the most prevalent forms,accounting for 74.3% in overall LGMD.Patients with LGMD2B usually have onset age between 19-27 years old.LGMD2B patients present as asymptomatic hyper creatine kinase emia (CK) at the early stage,and later develop to typical proximal muscle weakness with bilateral calf atrophy and extremely high serum CK.The onset age of LGMD2A patients is between 7-18 years old.LGMD2A patients presented as proximal muscle weakness with or without bilateral scapular winging and Achilles tendon contractures.Serum CK can be moderately or highly elevated.Current therapies are mainly supportive and the effective treatment is insufficient.The ongoing global elinical history study and gene therapy bring us new hope for treating LGMD in the coming future.

14.
Artigo em Inglês | WPRIM | ID: wpr-25818

RESUMO

Dysferlinopathy is caused by mutations in the DYSF gene. To characterize the clinical spectrum, we investigated the characteristics of 31 Korean dysferlinopathy patients confirmed by immunohistochemistry. The mean age of symptom onset was 22.23 +/- 7.34 yr. The serum creatine kinase (CK) was highly increased (4- to 101-fold above normal). The pathological findings of muscle specimens showed nonspecific dystrophic features and frequent inflammatory cell infiltration. Muscle imaging studies showed fatty atrophic changes dominantly in the posterolateral muscles of the lower limb. The patients with dysferlinopathy were classified by initial muscle weakness: fifteen patients with Miyoshi myopathy phenotype (MM), thirteen patients with limb girdle muscular dystrophy 2B phenotype (LGMD2B), two patients with proximodistal phenotype, and one asymptomatic patient. There were no differences between LGMD2B and MM groups in terms of onset age, serum CK levels and pathological findings. Dysferlinopathy patients usually have young adult onset and high serum CK levels. However, heterogeneity of clinical presentations and pathologic findings upon routine staining makes it difficult to diagnose dysferlinopathy. These limitations make immunohistochemistry currently the most important method for the diagnosis of dysferlinopathy.


Assuntos
Adolescente , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Idade de Início , Creatina Quinase/sangue , Miopatias Distais/patologia , Imuno-Histoquímica , Proteínas de Membrana/genética , Proteínas Musculares/genética , Atrofia Muscular/patologia , Distrofia Muscular do Cíngulo dos Membros/diagnóstico , Mutação , Fenótipo , República da Coreia , Tomografia Computadorizada por Raios X
15.
Artigo em Coreano | WPRIM | ID: wpr-158631

RESUMO

BACKGROUND: Dysferlin is a 230 kDa protein of the sarcolemma. This encoding gene is mutated in patients with dysferlinopathy (limb-girdle muscular dystrophy 2B and Miyoshi myopathy), which is characterized byan active muscle degeneration and regeneration process. Dysferlin is known to play an essential role in muscle signaling and muscle fiber repair. We studied the gene to define its functional role in muscle repair and differentiation in human skeletal muscle of the patients with myopathies and cultured human myoblast. METHODS: An immunohistochemical analysis of dysferlin and N-CAM in biopsied muscle tissue obtained from eleven patients with myopathies [six patients with Duchenne muscular dystrophy (DMD), two patients with dermatomyositis (DM), two patients with polymyositis (PM), and one patient with dysferlinopathy (MM)] and eight normal controls. Cultured human myoblast obtained from normal muscle tissue was also analyzed by the expression of dysferlin through immunocytochemical staining and western blot. RESULTS: The immunoreactivity of dysferlin was strongly expressed in regenerative muscle fibers of myopathies except dysferlinpathy, which was co-localization with N-CAM by double immunohistochemistry. By western blot analysis, the expression level of dysferlin was variable in myopathies compared to normal controls, but no expression in dyferlinopathy. The expression of dysferlin in myotubes was significantly increased compared to that in myoblast by immunostaining and western blot analysis. CONCLUSIONS: These results indicated that the expression of dysferlin increased in regenerative and degenerative muscle fibers and also increased in myoblast differentiation. Our study supports that dysferlin not only has a role in skeletal muscle development but also in regeneration/repair process.


Assuntos
Humanos , Humanos , Western Blotting , Dermatomiosite , Imuno-Histoquímica , Fibras Musculares Esqueléticas , Músculo Esquelético , Doenças Musculares , Distrofias Musculares , Distrofia Muscular de Duchenne , Mioblastos , Polimiosite , Regeneração , Sarcolema
16.
Artigo em Coreano | WPRIM | ID: wpr-151275

RESUMO

BACKGROUND: Recent genetic analyses have shown that Miyoshi myopathy (MM) is caused by a mutation in the DYSF, which induces the dysfunction of dysferlin. We identified the deficiency of dysferlin by immunohistochemistry and Western blot in four patients with clinically diagnosed MM, and investigated the clinical and pathological characteristics of MM. METHODS: A muscle biopsy was performed in four patients who were diagnosed with MM by clinical and electrophysiological study. Immunostaining of muscle specimens for dyferlin, dystrophin, alpha, beta, gamma, sigma-sarcoglycan, beta-dystroglycan, and caveolin-3 were performed in all four patients. We analyzed the quantitative analysis for dysferlin by Western blot in three of four patients. RESULTS: All four patients showed clinical onset during adolescence or early adulthood (15-26 year old), a slowly progressive course, and a relatively high serum creatine kinase level (2240-6400 IU/L). Routine pathological studies showed non-specific myopathic changes. On immunocytochemistry, there was negative immunoreacticity for dysferlin on muscle specimens in all patients. The immunoreactivities for dystrophin, alpha, beta, gamma, sigma-sarcoglycan, beta-dystroglycan, and caveolin-3 were normal. On Western blotting, complete loss of dysferlin was noted in all three patients with MM CONCLUSIONS: Identification of isolated deficiency of dysferlin on immunocytochemistry or Western blot is important for the confirmative diagnosis of MM.


Assuntos
Adolescente , Humanos , Biópsia , Western Blotting , Caveolina 3 , Creatina Quinase , Diagnóstico , Distroglicanas , Distrofina , Imuno-Histoquímica , Doenças Musculares
17.
Artigo em Inglês | WPRIM | ID: wpr-124466

RESUMO

Limb-girdle muscular dystrophy type 2B (LGMD2B), a subtype of autosomal recessive limb-girdle muscular dystrophy (ARLGMD), is characterized by a relatively late onset and slow progressive course. LGMD2B is known to be caused by the loss of the dysferlin protein at sarcolemma in muscle fibers. In this study, the clinical and pathological characteristics of Korean LGMD2B patients were investigated. Seventeen patients with ARLGMD underwent muscle biopsy and the histochemical examination was performed. For the immunocytochemistry, a set of antibodies against dystrophin, alpha, beta, gamma, delta-sarcoglycans, dysferlin, caveolin-3, and beta-dystroglycan was used. Four patients (24%) showed selective loss of immunoreactivity against dysferlin at the sarcolemma on the muscle specimens. Therefore, they were classified into the LGMD2B category. The age at the onset of disease ranged from 9 yr to 33 yr, and none of the patients was wheelchair bound at the neurological examination. The serum creatine kinase (CK) was high in all the patients (4010-5310 IU/L). The pathologic examination showed mild to moderate dystrophic features. These are the first Korean LGMD2B cases with a dysferlin deficiency confirmed by immunocytochemistry. The clinical, pathological, and immunocytochemical findings of the patients with LGMD2B in this study were in accordance with those of other previous reports.


Assuntos
Adolescente , Adulto , Criança , Feminino , Humanos , Masculino , Idade de Início , Creatina Quinase/sangue , Progressão da Doença , Imuno-Histoquímica , Coreia (Geográfico) , Proteínas de Membrana/biossíntese , Proteínas Musculares/biossíntese , Músculos/patologia , Distrofias Musculares/diagnóstico , Fatores de Tempo
18.
Yonsei Medical Journal ; : 927-930, 2004.
Artigo em Inglês | WPRIM | ID: wpr-203758

RESUMO

Recent genetic and immunohistochemical analyses have shown that Miyoshi myopathy (MM) is caused by a mutation in the DYSF gene, which induces dysfunction of dysferlin. The author described one patient showing characteristic MM phenotype with deficiency of dysferlin on immunohistochemistry. Direct DNA sequencing of whole exons of DYSF gene revealed one homozygous missense mutation (G1165C) on exon 12, which let to an amino acid substitution from the glutamic acid to glutamine at the 389 of the peptide sequence in this patient. This is the first reported case of MM confirmed by immunohistochemical and genetic analyses in Korea.


Assuntos
Adulto , Humanos , Masculino , Caveolinas/análise , Miopatias Distais/genética , Imuno-Histoquímica , Proteínas de Membrana/química , Proteínas Musculares/química , Mutação
19.
Artigo em Chinês | WPRIM | ID: wpr-594027

RESUMO

Objective To investigate the clinical and pathological features of Miyoshi myopathy(MM) with dysferlin protein deficient. Methods The clinical and pathological data of the 3 patients with MM were analysed. Results 3 patients were onset at youngster.The clinical manifestation were myastheria and myoatrophy in distal of lower limbs.1 case combined myalgia and tumefaction in lower limbs at early stage of onset;1 case showed myathenia in proximal of lower limbs.The level of serum creatine phosphokinase (CK) was significantly ligher in the 3 cases (7543 IU/L, 5657 IU/L, 8721 IU/L respectively). The level of serum lactic dehydrogenase (LDH) was significantly higher in the 2 cases (456 IU/L ,636 IU/L respectively).The result of muscle pathology was showed myogenic damage in all the cases. The expression of dysferlin protain in membrane of muscle cells was completely deficient, although the expression of dystrophin was normal. Inflammatory cells infiltration was found in 1 case's muscle tissue. Conclusions The clinical characters of MM patient are onset at youngster,myasthenia and myoatrophy in lower limbs.The deficit of dysferlin protain can be found by pathology.

20.
Artigo em Inglês | WPRIM | ID: wpr-197469

RESUMO

Dysferlin is a plasma membrane protein of skeletal muscle whose deficiency causes Miyoshi myopathy, limb girdle muscular dystrophy 2B and distal anterior compartment myopathy. Recent studies have reported that dysferlin is implicated in membrane repair mechanism and coimmunoprecipitates with caveolin 3 in human skeletal muscle. Caveolin 3 is a principal structural protein of caveolae membrane domains in striated muscle cells and cardiac myocytes. Mutations of caveolin 3 gene (CAV3) cause different diseases and where caveolin 3 expression is defective, dysferlin localization is abnormal. We describe the alteration of dysferlin expression and localization in skeletal muscle from a patient with raised serum creatine kinase (hyperCKaemia), whose reduction of caveolin 3 is caused by a CAV3 P28L mutation. Moreover, we performed a study on dysferlin interaction with caveolin 3 in C2C12 cells. We show the association of dysferlin to cellular membrane of C2C12 myotubes and the low affinity link between dysferlin and caveolin 3 by immunoprecipitation techniques. We also reproduced caveolinopathy conditions in C2C12 cells by a selective p38 MAP kinase inhibition with SB203580, which blocks the expression of caveolin 3. In this model, myoblasts do not fuse into myotubes and we found that dysferlin expression is reduced. These results underline the importance of dysferlin-caveolin 3 relationship for skeletal muscle integrity and propose a cellular model to clarify the dysferlin alteration mechanisms in caveolinopathies.


Assuntos
Animais , Humanos , Camundongos , Biópsia , Caveolina 3 , Caveolinas/genética , Linhagem Celular , Creatina Quinase/sangue , Inibidores Enzimáticos/farmacologia , Imidazóis/farmacologia , Insulina/farmacologia , Proteínas de Membrana/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Musculares/metabolismo , Músculo Esquelético/citologia , Mutação/genética , Ligação Proteica , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa