Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Medicina (Kaunas) ; 60(7)2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-39064489

RESUMO

Muscular dystrophies (MDs) are a heterogeneous group of diseases of genetic origin characterized by progressive skeletal muscle degeneration and weakness. There are several types of MDs, varying in terms of age of onset, severity, and pattern of the affected muscles. However, all of them worsen over time, and many patients will eventually lose their ability to walk. In addition to skeletal muscle effects, patients with MDs may present cardiac and respiratory disorders, generating complications that could lead to death. Interdisciplinary management is required to improve the surveillance and quality of life of patients with an MD. At present, pharmacological therapy is only available for Duchene muscular dystrophy (DMD)-the most common type of MD-and is mainly based on the use of corticosteroids. Other MDs caused by alterations in dystrophin-associated proteins (DAPs) are less frequent but represent an important group within these diseases. Pharmacological alternatives with clinical potential in patients with MDs and other proteins associated with dystrophin have been scarcely explored. This review focuses on drugs and molecules that have shown beneficial effects, mainly in experimental models involving alterations in DAPs. The mechanisms associated with the effects leading to promising results regarding the recovery or maintenance of muscle strength and reduction in fibrosis in the less-common MDs (i.e., with respect to DMD) are explored, and other therapeutic targets that could contribute to maintaining the homeostasis of muscle fibers, involving different pathways, such as calcium regulation, hypertrophy, and maintenance of satellite cell function, are also examined. It is possible that some of the drugs explored here could be used to affordably improve the muscular function of patients until a definitive treatment for MDs is developed.


Assuntos
Distrofias Musculares , Humanos , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/fisiopatologia , Distrofina , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/fisiopatologia , Complexo de Proteínas Associadas Distrofina
2.
Nat Commun ; 15(1): 4935, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858388

RESUMO

Cell polarity mechanisms allow the formation of specialized membrane domains with unique protein compositions, signalling properties, and functional characteristics. By analyzing the localization of potassium channels and proteins belonging to the dystrophin-associated protein complex, we reveal the existence of distinct planar-polarized membrane compartments at the surface of C. elegans muscle cells. We find that muscle polarity is controlled by a non-canonical Wnt signalling cascade involving the ligand EGL-20/Wnt, the receptor CAM-1/Ror, and the intracellular effector DSH-1/Dishevelled. Interestingly, classical planar cell polarity proteins are not required for this process. Using time-resolved protein degradation, we demonstrate that -while it is essentially in place by the end of embryogenesis- muscle polarity is a dynamic state, requiring continued presence of DSH-1 throughout post-embryonic life. Our results reveal the unsuspected complexity of the C. elegans muscle membrane and establish a genetically tractable model system to study cellular polarity and membrane compartmentalization in vivo.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Polaridade Celular , Distrofina , Músculos , Via de Sinalização Wnt , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Distrofina/metabolismo , Distrofina/genética , Músculos/metabolismo , Proteínas Desgrenhadas/metabolismo , Proteínas Desgrenhadas/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Membrana Celular/metabolismo , Complexo de Proteínas Associadas Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/genética , Proteínas Wnt/metabolismo , Transdução de Sinais
3.
Mass Spectrom Rev ; 43(1): 90-105, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-36420714

RESUMO

The dystrophin-associated protein complex (DAPC) is a highly organized multiprotein complex that plays a pivotal role in muscle fiber structure integrity and cell signaling. The complex is composed of three distinct interacting subgroups, intracellular peripheral proteins, transmembrane glycoproteins, and extracellular glycoproteins subcomplexes. Dystrophin protein nucleates the DAPC and is important for connecting the intracellular actin cytoskeletal filaments to the sarcolemma glycoprotein complex that is connected to the extracellular matrix via laminin, thus stabilizing the sarcolemma during muscle fiber contraction and relaxation. Genetic mutations that lead to lack of expression or altered expression of any of the DAPC proteins are associated with different types of muscle diseases. Hence characterization of this complex in healthy and dystrophic muscle might bring insights into its role in muscle pathogenesis. This review highlights the role of mass spectrometry in characterizing the DAPC interactome as well as post-translational glycan modifications of some of its components such as α-dystroglycan. Detection and quantification of dystrophin using targeted mass spectrometry are also discussed in the context of healthy versus dystrophic skeletal muscle.


Assuntos
Complexo de Proteínas Associadas Distrofina , Distrofina , Distrofina/análise , Distrofina/genética , Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/análise , Complexo de Proteínas Associadas Distrofina/metabolismo , Laminina/análise , Laminina/metabolismo , Sarcolema/química , Sarcolema/metabolismo , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Glicoproteínas/análise
4.
Gene Ther ; 29(9): 520-535, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35105949

RESUMO

Duchenne muscular dystrophy (DMD) is a muscle wasting disorder caused by mutations in the gene encoding dystrophin. Gene therapy using micro-dystrophin (MD) transgenes and recombinant adeno-associated virus (rAAV) vectors hold great promise. To overcome the limited packaging capacity of rAAV vectors, most MD do not include dystrophin carboxy-terminal (CT) domain. Yet, the CT domain is known to recruit α1- and ß1-syntrophins and α-dystrobrevin, a part of the dystrophin-associated protein complex (DAPC), which is a signaling and structural mediator of muscle cells. In this study, we explored the impact of inclusion of the dystrophin CT domain on ΔR4-23/ΔCT MD (MD1), in DMDmdx rats, which allows for relevant evaluations at muscular and cardiac levels. We showed by LC-MS/MS that MD1 expression is sufficient to restore the interactions at a physiological level of most DAPC partners in skeletal and cardiac muscles, and that inclusion of the CT domain increases the recruitment of some DAPC partners at supra-physiological levels. In parallel, we demonstrated that inclusion of the CT domain does not improve MD1 therapeutic efficacy on DMD muscle and cardiac pathologies. Our work highlights new evidences of the therapeutic potential of MD1 and strengthens the relevance of this candidate for gene therapy of DMD.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Animais , Cromatografia Líquida , Distrofina/genética , Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/metabolismo , Terapia Genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Ratos , Espectrometria de Massas em Tandem
5.
Protein Expr Purif ; 167: 105525, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31682967

RESUMO

Mutation of the gene encoding γ-sarcoglycan (SGCG), an integral membrane protein responsible for maintaining the integrity of the muscle cell sarcolemma, results in Limb-Girdle Muscular Dystrophy (LGMD), a congenital disease with no current treatment options. This member of the sarcoglycan glycoprotein family is a vital component of the Dystrophin Complex, which together facilitate normal muscle function. However, very little is known about the structure and dynamics of these proteins, and of membrane glycoproteins in general. This is due to a number of factors, including their complexity, heterogeneity and highly-specific native environments. The expression, purification, and structural study of membrane proteins is further impeded by their hydrophobic nature and consequent propensity to aggregate in aqueous solutions. Here, we report the first successful expression and purification of milligram quantities of full-length recombinant SGCG, utilizing fusion protein-guided overexpression to inclusion bodies in Escherichia coli. Purification of SGCG from the fusion protein, TrpΔLE, was facilitated using chemical cleavage. Cleavage products were then isolated by size-exclusion chromatography. Successful purification of the protein was confirmed using SDS-PAGE and mass spectroscopy. Finally, solution nuclear magnetic resonance spectroscopy of uniformly 15N-labeled SGCG in detergent environments was performed, yielding the first spectra of the full-length membrane glycoprotein, SGCG. These results represent the initial structural studies of SGCG, laying the foundation for further investigation on the interaction and dynamics of other integral membrane proteins. More specifically, this data allows for opportunities in the future for enhanced treatment modalities and cures for LGMD.


Assuntos
Sarcoglicanas , Cromatografia em Gel , Clonagem Molecular/métodos , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/isolamento & purificação , Complexo de Proteínas Associadas Distrofina/metabolismo , Escherichia coli , Glicoproteínas/biossíntese , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/isolamento & purificação , Humanos , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Proteínas de Membrana/análise , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular do Cíngulo dos Membros/etiologia , Mutação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Sarcoglicanas/biossíntese , Sarcoglicanas/química , Sarcoglicanas/genética , Sarcoglicanas/isolamento & purificação , Sarcolema/metabolismo , Solubilidade
6.
Sci Rep ; 9(1): 2770, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808964

RESUMO

The dystrophin-glycoprotein complex (DGC) links the muscle cytoskeleton to the extracellular matrix and is responsible for force transduction and protects the muscle fibres from contraction induced damage. Mutations in components of the DGC are responsible for muscular dystrophies and congenital myopathies. Expression of DGC components have been shown to be altered in many myopathies. In contrast we have very little evidence of whether adaptive changes in muscle impact on DGC expression. In this study we investigated connection between muscle fibre phenotype and the DGC. Our study reveals that the levels of DGC proteins at the sarcolemma differ in highly glycolytic muscle compared to wild-type and that these changes can be normalised by the super-imposition of an oxidative metabolic programme. Importantly we show that the metabolic properties of the muscle do not impact on the total amount of DGC components at the protein level. Our work shows that the metabolic property of a muscle fibre is a key factor in regulating the expression of DGC proteins at the sarcolemma.


Assuntos
Complexo de Proteínas Associadas Distrofina/metabolismo , Distrofina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Animais , Colágeno Tipo IV/metabolismo , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Miostatina/deficiência , Miostatina/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Sarcoglicanas/metabolismo
7.
Methods Mol Biol ; 1828: 327-342, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30171551

RESUMO

Exon-skipping antisense oligonucleotides (AOs) are promising treatments for muscle-related genetic ailments including Duchenne muscular dystrophy (DMD), but clinical translation is unfortunately hampered by insufficient systemic delivery. Here we describe that how one can employ a glucose-fructose injection mixture to improve muscle uptake and functional outcomes of DMD AOs in energy-deficient peripheral muscles of mdx mice. The potentiating effect of glucose-fructose on AOs in energy-deficient muscles offers a simple and economical method for enhancing AO potency, reducing screening costs for researchers and accelerating the translation of nucleic acid-based therapeutics in DMD and other muscular dystrophies.


Assuntos
Éxons , Frutose/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Splicing de RNA , Trifosfato de Adenosina/metabolismo , Animais , Clatrina/metabolismo , Distrofina/genética , Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/genética , Complexo de Proteínas Associadas Distrofina/metabolismo , Endocitose , Metabolismo Energético , Frutose/metabolismo , Glucose/metabolismo , Humanos , Imuno-Histoquímica , Redes e Vias Metabólicas , Camundongos , Camundongos Endogâmicos mdx , Morfolinos/administração & dosagem , Morfolinos/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/genética
8.
Biomaterials ; 183: 54-66, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30149230

RESUMO

Duchenne Muscular Dystrophy (DMD) is a fatal genetic disorder currently having no cure. Here we report that culture substrates patterned with nanogrooves and functionalized with Matrigel (or laminin) present an engineered cell microenvironment to allow myotubes derived from non-diseased, less-affected DMD, and severely-affected DMD human induced pluripotent stem cells (hiPSCs) to exhibit prominent differences in alignment and orientation, providing a sensitive phenotypic biomarker to potentially facilitate DMD drug development and early diagnosis. We discovered that myotubes differentiated from myogenic progenitors derived from non-diseased hiPSCs align nearly perpendicular to nanogrooves, a phenomenon not reported previously. We further found that myotubes derived from hiPSCs of a dystrophin-null DMD patient orient randomly, and those from hiPSCs of a patient carrying partially functional dystrophin align approximately 14° off the alignment direction of non-diseased myotubes. Substrates engineered with micron-scale grooves and/or cell adhesion molecules only interacting with integrins all guide parallel myotube alignment to grooves and lose the ability to distinguish different cell types. Disruption of the interaction between the Dystrophin-Associated-Protein-Complex (DAPC) and laminin by heparin or anti-α-dystroglycan antibody IIH6 disenables myotubes to align perpendicular to nanogrooves, suggesting that this phenotype is controlled by the DAPC-mediated cytoskeleton-extracellular matrix linkage.


Assuntos
Fibras Musculares Esqueléticas/fisiologia , Distrofia Muscular de Duchenne/diagnóstico , Nanoestruturas/química , Biomarcadores , Diferenciação Celular , Células Cultivadas , Microambiente Celular , Colágeno/metabolismo , Citoesqueleto/metabolismo , Combinação de Medicamentos , Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/metabolismo , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Laminina/metabolismo , Desenvolvimento Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Mioblastos/metabolismo , Mioblastos/patologia , Fenótipo , Proteoglicanas/metabolismo
9.
Matrix Biol ; 68-69: 628-636, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29475025

RESUMO

Endplate acetylcholinesterase (AChE) deficiency is a form of congenital myasthenic syndrome (CMS) caused by mutations in COLQ, which encodes collagen Q (ColQ). ColQ is an extracellular matrix (ECM) protein that anchors AChE to the synaptic basal lamina. Biglycan, encoded by BGN, is another ECM protein that binds to the dystrophin-associated protein complex (DAPC) on skeletal muscle, which links the actin cytoskeleton and ECM proteins to stabilize the sarcolemma during repeated muscle contractions. Upregulation of biglycan stabilizes the DPAC. Gene therapy can potentially ameliorate any disease that can be recapitulated in cultured cells. However, the difficulty of tissue-specific and developmental stage-specific regulated expression of transgenes, as well as the difficulty of introducing a transgene into all cells in a specific tissue, prevents us from successfully applying gene therapy to many human diseases. In contrast to intracellular proteins, an ECM protein is anchored to the target tissue via its specific binding affinity for protein(s) expressed on the cell surface within the target tissue. Exploiting this unique feature of ECM proteins, we developed protein-anchoring therapy in which a transgene product expressed even in remote tissues can be delivered and anchored to a target tissue using specific binding signals. We demonstrate the application of protein-anchoring therapy to two disease models. First, intravenous administration of adeno-associated virus (AAV) serotype 8-COLQ to Colq-deficient mice, resulting in specific anchoring of ectopically expressed ColQ-AChE at the NMJ, markedly improved motor functions, synaptic transmission, and the ultrastructure of the neuromuscular junction (NMJ). In the second example, Mdx mice, a model for Duchenne muscular dystrophy, were intravenously injected with AAV8-BGN. The treatment ameliorated motor deficits, mitigated muscle histopathologies, decreased plasma creatine kinase activities, and upregulated expression of utrophin and DAPC component proteins. We propose that protein-anchoring therapy could be applied to hereditary/acquired defects in ECM and secreted proteins, as well as therapeutic overexpression of such factors.


Assuntos
Acetilcolinesterase/genética , Biglicano/genética , Colágeno/genética , Terapia Genética/métodos , Proteínas Musculares/genética , Distrofia Muscular de Duchenne/terapia , Síndromes Miastênicas Congênitas/terapia , Acetilcolinesterase/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Biglicano/metabolismo , Colágeno/metabolismo , Dependovirus/química , Modelos Animais de Doenças , Complexo de Proteínas Associadas Distrofina/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Proteínas Ligadas por GPI/metabolismo , Vetores Genéticos/administração & dosagem , Humanos , Camundongos , Proteínas Musculares/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Síndromes Miastênicas Congênitas/genética , Síndromes Miastênicas Congênitas/metabolismo
10.
J Cell Physiol ; 233(7): 5142-5159, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-28464259

RESUMO

Dystrophin protein in association with several other cellular proteins and glycoproteins leads to the formation of a large multifaceted protein complex at the cell membrane referred to as dystrophin glycoprotein complex (DGC), that serves distinct functions in cell signaling and maintaining the membrane stability as well as integrity. In accordance with this, several findings suggest exquisite role of DGC in signaling pathways associated with cell development and/or maintenance of homeostasis. In the present review, we summarize the established facts about the various components of this complex with emphasis on recent insights into specific contribution of the DGC in cell signaling at the membrane. We have also discussed the recent advances made in exploring the molecular associations of DGC components within the cells and the functional implications of these interactions. Our review would help to comprehend the composition, role, and functioning of DGC and may lead to a deeper understanding of its role in several human diseases.


Assuntos
Membrana Celular/genética , Complexo de Proteínas Associadas Distrofina/genética , Distrofina/genética , Glicoproteínas/genética , Membrana Celular/química , Distrofina/química , Complexo de Proteínas Associadas Distrofina/química , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA