Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurol Genet ; 7(3): e590, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34235269

RESUMEN

OBJECTIVE: To determine whether a new indel mutation in the dimerization domain of filamin C (FLNc) causes a hereditary myopathy with protein aggregation in muscle fibers, we clinically and molecularly studied a German family with autosomal dominant myofibrillar myopathy (MFM). METHODS: We performed mutational analysis in 3 generations, muscle histopathology, and proteomic studies of IM protein aggregates. Functional consequences of the FLNC mutation were investigated with interaction and transfection studies and biophysics molecular analysis. RESULTS: Eight patients revealed clinical features of slowly progressive proximal weakness associated with a heterozygous c.8025_8030delCAAGACinsA (p.K2676Pfs*3) mutation in FLNC. Two patients exhibited a mild cardiomyopathy. MRI of skeletal muscle revealed lipomatous changes typical for MFM with FLNC mutations. Muscle biopsies showed characteristic MFM findings with protein aggregation and lesion formation. The proteomic profile of aggregates was specific for MFM-filaminopathy and indicated activation of the ubiquitin-proteasome system (UPS) and autophagic pathways. Functional studies revealed that mutant FLNc is misfolded, unstable, and incapable of forming homodimers and heterodimers with wild-type FLNc. CONCLUSIONS: This new MFM-filaminopathy family confirms that expression of mutant FLNC leads to an adult-onset muscle phenotype with intracellular protein accumulation. Mutant FLNc protein is biochemically compromised and leads to dysregulation of protein quality control mechanisms. Proteomic analysis of MFM protein aggregates is a potent method to identify disease-relevant proteins, differentiate MFM subtypes, evaluate the relevance of gene variants, and identify novel MFM candidate genes.

2.
Front Immunol ; 12: 670219, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34290699

RESUMEN

Orientia (O.) tsutsugamushi, the causative agent of scrub typhus, is a neglected, obligate intracellular bacterium that has a prominent tropism for monocytes and macrophages. Complications often involve the lung, where interstitial pneumonia is a typical finding. The severity of scrub typhus in humans has been linked to altered plasma concentrations of chemokines which are known to act as chemoattractants for myeloid cells. The trafficking and function of monocyte responses is critically regulated by interaction of the CC chemokine ligand 2 (CCL2) and its CC chemokine receptor CCR2. In a self-healing mouse model of intradermal infection with the human-pathogenic Karp strain of O. tsutsugamushi, we investigated the role of CCR2 on bacterial dissemination, development of symptoms, lung histology and monocyte subsets in blood and lungs. CCR2-deficient mice showed a delayed onset of disease and resolution of symptoms, higher concentrations and impaired clearance of bacteria in the lung and the liver, accompanied by a slow infiltration of interstitial macrophages into the lungs. In the blood, we found an induction of circulating monocytes that depended on CCR2, while only a small increase in Ly6Chi monocytes was observed in CCR2-/- mice. In the lung, significantly higher numbers of Ly6Chi and Ly6Clo monocytes were found in the C57BL/6 mice compared to CCR2-/- mice. Both wildtype and CCR2-deficient mice developed an inflammatory milieu as shown by cytokine and inos/arg1 mRNA induction in the lung, but with delayed kinetics in CCR2-deficient mice. Histopathology revealed that infiltration of macrophages to the parenchyma, but not into the peribronchial tissue, depended on CCR2. In sum, our data suggest that in Orientia infection, CCR2 drives blood monocytosis and the influx and activation of Ly6Chi and Ly6Clo monocytes into the lung, thereby accelerating bacterial replication and development of interstitial pulmonary inflammation.


Asunto(s)
Antígenos Ly/metabolismo , Pulmón/microbiología , Macrófagos/microbiología , Monocitos/microbiología , Orientia tsutsugamushi/patogenicidad , Receptores CCR2/deficiencia , Tifus por Ácaros/microbiología , Animales , Carga Bacteriana , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Hígado/inmunología , Hígado/metabolismo , Hígado/microbiología , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Orientia tsutsugamushi/crecimiento & desarrollo , Orientia tsutsugamushi/inmunología , Receptores CCR2/genética , Tifus por Ácaros/genética , Tifus por Ácaros/inmunología , Tifus por Ácaros/metabolismo
3.
Acta Neuropathol Commun ; 8(1): 154, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32887649

RESUMEN

Filamin C (FLNc) is mainly expressed in striated muscle cells where it localizes to Z-discs, myotendinous junctions and intercalated discs. Recent studies have revealed numerous mutations in the FLNC gene causing familial and sporadic myopathies and cardiomyopathies with marked clinical variability. The most frequent myopathic mutation, p.W2710X, which is associated with myofibrillar myopathy, deletes the carboxy-terminal 16 amino acids from FLNc and abolishes the dimerization property of Ig-like domain 24. We previously characterized "knock-in" mice heterozygous for this mutation (p.W2711X), and have now investigated homozygous mice using protein and mRNA expression analyses, mass spectrometry, and extensive immunolocalization and ultrastructural studies. Although the latter mice display a relatively mild myopathy under normal conditions, our analyses identified major mechanisms causing the pathophysiology of this disease: in comparison to wildtype animals (i) the expression level of FLNc protein is drastically reduced; (ii) mutant FLNc is relocalized from Z-discs to particularly mechanically strained parts of muscle cells, i.e. myotendinous junctions and myofibrillar lesions; (iii) the number of lesions is greatly increased and these lesions lack Bcl2-associated athanogene 3 (BAG3) protein; (iv) the expression of heat shock protein beta-7 (HSPB7) is almost completely abolished. These findings indicate grave disturbances of BAG3-dependent and -independent autophagy pathways that are required for efficient lesion repair. In addition, our studies reveal general mechanisms of lesion formation and demonstrate that defective FLNc dimerization via its carboxy-terminal domain does not disturb assembly and basic function of myofibrils. An alternative, more amino-terminally located dimerization site might compensate for that loss. Since filamins function as stress sensors, our data further substantiate that FLNc is important for mechanosensing in the context of Z-disc stabilization and maintenance.


Asunto(s)
Filaminas/genética , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología , Sarcómeros/patología , Animales , Técnicas de Sustitución del Gen , Homocigoto , Ratones , Mutación , Miopatías Estructurales Congénitas/metabolismo , Sarcómeros/metabolismo
4.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 62-75, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27756573

RESUMEN

The cochaperone BAG3 is a central protein homeostasis factor in mechanically strained mammalian cells. It mediates the degradation of unfolded and damaged forms of the actin-crosslinker filamin through chaperone-assisted selective autophagy (CASA). In addition, BAG3 stimulates filamin transcription in order to compensate autophagic disposal and to maintain the actin cytoskeleton under strain. Here we demonstrate that BAG3 coordinates protein synthesis and autophagy through spatial regulation of the mammalian target of rapamycin complex 1 (mTORC1). The cochaperone utilizes its WW domain to contact a proline-rich motif in the tuberous sclerosis protein TSC1 that functions as an mTORC1 inhibitor in association with TSC2. Interaction with BAG3 results in a recruitment of TSC complexes to actin stress fibers, where the complexes act on a subpopulation of mTOR-positive vesicles associated with the cytoskeleton. Local inhibition of mTORC1 is essential to initiate autophagy at sites of filamin unfolding and damage. At the same time, BAG3-mediated sequestration of TSC1/TSC2 relieves mTORC1 inhibition in the remaining cytoplasm, which stimulates protein translation. In human muscle, an exercise-induced association of TSC1 with the cytoskeleton coincides with mTORC1 activation in the cytoplasm. The spatial regulation of mTORC1 exerted by BAG3 apparently provides the basis for a simultaneous induction of autophagy and protein synthesis to maintain the proteome under mechanical strain.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/genética , Complejos Multiproteicos/genética , Músculo Esquelético/metabolismo , Miocitos del Músculo Liso/metabolismo , Estrés Mecánico , Serina-Treonina Quinasas TOR/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Fenómenos Biomecánicos , Línea Celular , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Filaminas/genética , Filaminas/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/metabolismo , Músculo Esquelético/citología , Miocitos del Músculo Liso/ultraestructura , Unión Proteica , Biosíntesis de Proteínas , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
5.
Exp Cell Res ; 348(1): 95-105, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27639425

RESUMEN

Pressure overload induces cardiac remodeling involving both the contractile machinery and intercalated disks (IDs). Filamin C (FlnC) and Xin actin-binding repeat-containing proteins (XIRPs) are multi-adapters localizing in IDs of higher vertebrates. Knockout of the gene encoding Xin (Xirp1) in mice leads to a mild cardiac phenotype with ID mislocalization. In order to amplify this phenotype, we performed transverse aortic constriction (TAC) on control and Xirp1-deficient mice. TAC induced similar left ventricular hypertrophy in both genotypes, suggesting that the lack of Xin does not lead to higher susceptibility to cardiac overload. However, in both genotypes, FlnC appeared in "streaming" localizations across multiple sarcomeres proximal to the IDs, suggesting a remodeling response. Furthermore, FlnC-positive areas of remodeling, reminiscent of sarcomeric lesions previously described for skeletal muscles (but so far unreported in the heart), were also observed. These adaptations reflect a similarly strong effect of the pressure induced by TAC in both genotypes. However, 2 weeks post-operation TAC-treated knockout hearts had reduced levels of connexin43 and slightly increased incidents of ventricular tachycardia compared to their wild-type (WT) counterparts. Our findings highlight the FlnC-positive sarcomeric lesions and ID-proximal streaming as general remodeling responses in cardiac overload-induced hypertrophy.


Asunto(s)
Cardiomegalia/patología , Sarcómeros/patología , Animales , Aorta/patología , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/diagnóstico por imagen , Arritmias Cardíacas/patología , Cardiomegalia/complicaciones , Cardiomegalia/diagnóstico por imagen , Conexina 43/metabolismo , Constricción Patológica , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Electrocardiografía , Femenino , Filaminas/metabolismo , Hipertrofia Ventricular Izquierda/complicaciones , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/patología , Ratones , Miocardio/metabolismo , Miocardio/patología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Fenotipo , Taquicardia/complicaciones , Taquicardia/diagnóstico por imagen , Taquicardia/patología
6.
Eur J Hum Genet ; 24(12): 1771-1777, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27485408

RESUMEN

We describe a new early-onset neuromuscular disorder due to a homozygous loss-of-function variant in the kyphoscoliosis peptidase gene (KY). A 7.5-year-old girl with walking difficulties from 2 years of age presented with generalized muscle weakness; mild contractures in the shoulders, hips and feet; cavus feet; and lordosis but no scoliosis. She had previously been operated with Achilles tendon elongation. Whole-body MRI showed atrophy and fatty infiltration in the calf muscles. Biopsy of the vastus lateralis muscle showed variability in fiber size, with some internalized nuclei and numerous very small fibers with variable expression of developmental myosin heavy chain isoforms. Some small fibers showed abnormal sarcomeres with thickened Z-discs and small nemaline rods. Whole-exome sequencing revealed a homozygous one-base deletion (c.1071delG, p.(Thr358Leufs*3)) in KY, predicted to result in a truncated protein. Analysis of an RNA panel showed that KY is predominantly expressed in skeletal muscle in humans. A recessive variant in the murine ortholog Ky was previously described in a spontaneously generated mouse mutant with kyphoscoliosis, which developed postnatally and was caused by dystrophy of postural muscles. The abnormal distribution of Xin and Ky-binding partner filamin C in the muscle fibers of our patient was highly similar to their altered localization in ky/ky mouse muscle fibers. We describe the first human case of disease associated with KY inactivation. As in the mouse model, the affected child showed a neuromuscular disorder - but in contrast, no kyphoscoliosis.


Asunto(s)
Cifosis/genética , Proteínas Musculares/genética , Péptido Hidrolasas/genética , Escoliosis/genética , Edad de Inicio , Niño , Codón sin Sentido , Femenino , Filaminas/metabolismo , Humanos , Cifosis/diagnóstico por imagen , Cifosis/patología , Proteínas Musculares/deficiencia , Péptido Hidrolasas/deficiencia , Músculo Cuádriceps/metabolismo , Músculo Cuádriceps/patología , Escoliosis/diagnóstico por imagen , Escoliosis/patología
7.
Sci Rep ; 6: 19614, 2016 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-26804343

RESUMEN

Eccentric exercise leads to focal disruptions in the myofibrils, referred to as "lesions". These structures are thought to contribute to the post-exercise muscle weakness, and to represent areas of mechanical damage and/or remodelling. Lesions have been investigated in human biopsies and animal samples after exercise. However, this approach does not examine the mechanisms behind lesion formation, or their behaviour during contraction. To circumvent this, we used electrical pulse stimulation (EPS) to simulate exercise in C2C12 myotubes, combined with live microscopy. EPS application led to the formation of sarcomeric lesions in the myotubes, resembling those seen in exercised mice, increasing in number with the time of application or stimulation intensity. Furthermore, transfection with an EGFP-tagged version of the lesion and Z-disc marker filamin-C allowed us to observe the formation of lesions using live cell imaging. Finally, using the same technique we studied the behaviour of these structures during contraction, and observed them to be passively stretching. This passive behaviour supports the hypothesis that lesions contribute to the post-exercise muscle weakness, protecting against further damage. We conclude that EPS can be reliably used as a model for the induction and study of sarcomeric lesions in myotubes in vitro.


Asunto(s)
Músculo Esquelético/ultraestructura , Condicionamiento Físico Animal , Sarcómeros/ultraestructura , Animales , Biopsia , Humanos , Ratones , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/fisiología , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/fisiopatología , Sarcómeros/patología , Sarcómeros/fisiología
8.
Hum Mol Genet ; 24(25): 7207-20, 2015 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-26472074

RESUMEN

Filamin C (FLNC) mutations in humans cause myofibrillar myopathy (MFM) and cardiomyopathy, characterized by protein aggregation and myofibrillar degeneration. We generated the first patient-mimicking knock-in mouse harbouring the most common disease-causing filamin C mutation (p.W2710X). These heterozygous mice developed muscle weakness and myofibrillar instability, with formation of filamin C- and Xin-positive lesions streaming between Z-discs. These lesions, which are distinct from the classical MFM protein aggregates by their morphology and filamentous appearance, were greatly increased in number upon acute physical exercise in the mice. This pathology suggests that mutant filamin influences the mechanical stability of myofibrillar Z-discs, explaining the muscle weakness in mice and humans. Re-evaluation of biopsies from MFM-filaminopathy patients with different FLNC mutations revealed a similar, previously unreported lesion pathology, in addition to the classical protein aggregates, and suggested that structures previously interpreted as aggregates may be in part sarcomeric lesions. We postulate that these lesions define preclinical disease stages, preceding the formation of protein aggregates.


Asunto(s)
Músculo Esquelético/patología , Miofibrillas/patología , Animales , Filaminas/genética , Genotipo , Ratones , Microscopía Electrónica , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Distrofias Musculares/genética , Miofibrillas/genética , Fenotipo
9.
J Mol Biol ; 427(12): 2151-8, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-25868382

RESUMEN

The Drosophila indirect flight muscles (IFM) can be used as a model for the study of sarcomere assembly. Here we use a transgenic line with a green fluorescent protein (GFP) exon inserted into the Z-disc-proximal portion of sallimus (Sls), also known as Drosophila titin, to observe sarcomere assembly during IFM development. Firstly, we confirm that Sls-GFP can be used in the heterozygote state without an obvious phenotype in IFM and other muscles. We then use Sls-GFP in the IFM to show that sarcomeres grow individually and uniformly throughout the fibre, growing linearly in length and in diameter. Finally, we show that limiting the amounts of Sls in the IFM using RNAi leads to sarcomeres with smaller Z-discs in their core, whilst the thick/thin filament lattice can form peripherally without a Z-disc. Thick filament preparations from those muscles show that although the Z-disc-containing core has thick filaments of a regular length, filaments from the peripheral lattice are longer and asymmetrical around the bare zone. Therefore, the Z-disc and Sls are required for thick filament length specification but not for the assembly of the thin/thick filament lattice.


Asunto(s)
Conectina/metabolismo , Drosophila/enzimología , Drosophila/fisiología , Sarcómeros/metabolismo , Animales , Animales Modificados Genéticamente , Drosophila/crecimiento & desarrollo , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Músculos/enzimología , Músculos/fisiología , Coloración y Etiquetado/métodos
10.
J Cell Sci ; 127(Pt 16): 3578-92, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24963132

RESUMEN

Filamin C (FLNc) and Xin actin-binding repeat-containing proteins (XIRPs) are multi-adaptor proteins that are mainly expressed in cardiac and skeletal muscles and which play important roles in the assembly and repair of myofibrils and their attachment to the membrane. We identified the dystrophin-binding protein aciculin (also known as phosphoglucomutase-like protein 5, PGM5) as a new interaction partner of FLNc and Xin. All three proteins colocalized at intercalated discs of cardiac muscle and myotendinous junctions of skeletal muscle, whereas FLNc and aciculin also colocalized in mature Z-discs. Bimolecular fluorescence complementation experiments in developing cultured mammalian skeletal muscle cells demonstrated that Xin and aciculin also interact in FLNc-containing immature myofibrils and areas of myofibrillar remodeling and repair induced by electrical pulse stimulation (EPS). Fluorescence recovery after photobleaching (FRAP) experiments showed that aciculin is a highly dynamic and mobile protein. Aciculin knockdown in myotubes led to failure in myofibril assembly, alignment and membrane attachment, and a massive reduction in myofibril number. A highly similar phenotype was found upon depletion of aciculin in zebrafish embryos. Our results point to a thus far unappreciated, but essential, function of aciculin in myofibril formation, maintenance and remodeling.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Filaminas/metabolismo , Miofibrillas/metabolismo , Proteínas Nucleares/metabolismo , Fosfoglucomutasa/metabolismo , Animales , Línea Celular , Células Cultivadas , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Filaminas/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mioblastos/metabolismo , Miofibrillas/genética , Proteínas Nucleares/genética , Fosfoglucomutasa/genética , Unión Proteica
11.
PLoS Genet ; 10(2): e1004166, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586196

RESUMEN

During muscle development, myosin and actin containing filaments assemble into the highly organized sarcomeric structure critical for muscle function. Although sarcomerogenesis clearly involves the de novo formation of actin filaments, this process remained poorly understood. Here we show that mouse and Drosophila members of the DAAM formin family are sarcomere-associated actin assembly factors enriched at the Z-disc and M-band. Analysis of dDAAM mutants revealed a pivotal role in myofibrillogenesis of larval somatic muscles, indirect flight muscles and the heart. We found that loss of dDAAM function results in multiple defects in sarcomere development including thin and thick filament disorganization, Z-disc and M-band formation, and a near complete absence of the myofibrillar lattice. Collectively, our data suggest that dDAAM is required for the initial assembly of thin filaments, and subsequently it promotes filament elongation by assembling short actin polymers that anneal to the pointed end of the growing filaments, and by antagonizing the capping protein Tropomodulin.


Asunto(s)
Citoesqueleto de Actina/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Drosophila/genética , Desarrollo de Músculos/genética , Sarcómeros/genética , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Diferenciación Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Ratones , Desarrollo de Músculos/fisiología , Miocardio/metabolismo , Miofibrillas/genética , Miofibrillas/metabolismo , Miosinas/genética , Sarcómeros/fisiología , Sarcómeros/ultraestructura
12.
Mol Biol Cell ; 24(20): 3215-26, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23985323

RESUMEN

The Xin actin-binding repeat-containing proteins Xin and XIRP2 are exclusively expressed in striated muscle cells, where they are believed to play an important role in development. In adult muscle, both proteins are concentrated at attachment sites of myofibrils to the membrane. In contrast, during development they are localized to immature myofibrils together with their binding partner, filamin C, indicating an involvement of both proteins in myofibril assembly. We identify the SH3 domains of nebulin and nebulette as novel ligands of proline-rich regions of Xin and XIRP2. Precise binding motifs are mapped and shown to bind both SH3 domains with micromolar affinity. Cocrystallization of the nebulette SH3 domain with the interacting XIRP2 peptide PPPTLPKPKLPKH reveals selective interactions that conform to class II SH3 domain-binding peptides. Bimolecular fluorescence complementation experiments in cultured muscle cells indicate a temporally restricted interaction of Xin-repeat proteins with nebulin/nebulette during early stages of myofibril development that is lost upon further maturation. In mature myofibrils, this interaction is limited to longitudinally oriented structures associated with myofibril development and remodeling. These data provide new insights into the role of Xin actin-binding repeat-containing proteins (together with their interaction partners) in myofibril assembly and after muscle damage.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/metabolismo , Miofibrillas/metabolismo , Proteínas Nucleares/metabolismo , Animales , Sitios de Unión , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas con Dominio LIM/química , Proteínas con Dominio LIM/genética , Ligandos , Ratones , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteínas Musculares/química , Proteínas Musculares/genética , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Miofibrillas/química , Miofibrillas/ultraestructura , Proteínas Nucleares/química , Proteínas Nucleares/genética , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Dominios Homologos src/genética
13.
J Muscle Res Cell Motil ; 34(1): 61-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23225103

RESUMEN

The assembly of striated muscle myofibrils is a multistep process in which a variety of proteins is involved. One of the first and most important steps in myofibrillogenesis is the arrangement of thin myofilaments into ordered I-Z-I brushes, requiring the coordinated activity of numerous actin binding proteins. The early expression of myopodin prior to sarcomeric α-actinin, as well as its binding to actin, α-actinin and filamin indicate an important role for this protein in actin cytoskeleton remodelling with the precise function of myopodin in this process yet remaining to be resolved. While myopodin was previously described as a protein capable of cross-linking actin filaments into thick bundles upon transient transfections, it has remained unclear whether myopodin alone is capable of bundling actin, or if additional proteins are involved. We have therefore investigated the in vitro actin binding properties of myopodin. High speed cosedimentation assays with skeletal muscle actin confirmed direct binding of myopodin to F-actin and showed that this interaction is mediated by at least two independent actin binding sites, found in all myopodin isoforms identified to date. Furthermore, low-speed cosedimentation assays revealed that not only full length myopodin, but also the fragment containing only the second binding site, bundles microfilaments in the absence of accessory proteins. Ultrastructural analysis demonstrated that this bundling activity resembled that of α-actinin. Biochemical experiments revealed that bundling was not achieved by myopodin's ability to dimerize, indicating the presence of two individual F-actin binding sites within the second binding segment. Thus full length myopodin contains at least three F-actin binding sites. These data provide further understanding of the mechanisms by which myopodin contributes to actin reorganization during myofibril assembly.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinina/metabolismo , Actinas/metabolismo , Multimerización de Proteína , Animales , Sitios de Unión , Pollos/metabolismo , Cromatografía en Gel , Clonación Molecular , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Complejos Multiproteicos/metabolismo , Músculo Esquelético/metabolismo , Unión Proteica , Técnicas del Sistema de Dos Híbridos
14.
J Cell Sci ; 126(Pt 1): 139-48, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23178940

RESUMEN

During muscle development myosin molecules form symmetrical thick filaments, which integrate with the thin filaments to produce the regular sarcomeric lattice. In Drosophila indirect flight muscles (IFMs) the details of this process can be studied using genetic approaches. The weeP26 transgenic line has a GFP-encoding exon inserted into the single Drosophila muscle myosin heavy chain gene, Mhc. The weeP26 IFM sarcomeres have a unique MHC-GFP-labelling pattern restricted to the sarcomere core, explained by non-translation of the GFP exon following alternative splicing. Characterisation of wild-type IFM MHC mRNA confirmed the presence of an alternately spliced isoform, expressed earlier than the major IFM-specific isoform. The two wild-type IFM-specific MHC isoforms differ by the presence of a C-terminal 'tailpiece' in the minor isoform. The sequential expression and assembly of these two MHCs into developing thick filaments suggest a role for the tailpiece in initiating A-band formation. The restriction of the MHC-GFP sarcomeric pattern in weeP26 is lifted when the IFM lack the IFM-specific myosin binding protein flightin, suggesting that it limits myosin dissociation from thick filaments. Studies of flightin binding to developing thick filaments reveal a progressive binding at the growing thick filament tips and in a retrograde direction to earlier assembled, proximal filament regions. We propose that this flightin binding restricts myosin molecule incorporation/dissociation during thick filament assembly and explains the location of the early MHC isoform pattern in the IFM A-band.


Asunto(s)
Proteínas de Drosophila/metabolismo , Músculo Esquelético/metabolismo , Isoformas de Proteínas/metabolismo , Empalme Alternativo/genética , Empalme Alternativo/fisiología , Animales , Drosophila , Proteínas de Drosophila/genética , Exones/genética , Filaminas , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miosinas/genética , Miosinas/metabolismo , Isoformas de Proteínas/genética , Sarcómeros/metabolismo
15.
J Muscle Res Cell Motil ; 29(6-8): 185-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19152055

RESUMEN

One of the advantages of using Drosophila melanogaster as a model organism is our knowledge of its genetics combined with its genetic versatility. Over the years, a range of transgenic tools have been developed that allow easy genetic manipulation such as the generation of novel mutants and the use of fusion proteins and protein traps. This work discusses examples of the above, and describes systems like GAL4-UAS and RNAi, before highlighting the importance and advantage of the indirect flight muscle as a model for muscle research.


Asunto(s)
Animales Modificados Genéticamente/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Músculo Esquelético/fisiología , Animales , Animales Modificados Genéticamente/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Músculo Esquelético/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...