Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Tissue Res ; 393(1): 133-147, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37178194

RESUMEN

Variations in the gene encoding filamin-A-interacting protein 1 (FILIP1) were identified to be associated with a combination of neurological and muscular symptoms. While FILIP1 was shown to regulate motility of brain ventricular zone cells, a process important for corticogenesis, the function of the protein in muscle cells has been less well characterized. The expression of FILIP1 in regenerating muscle fibres predicted a role in early muscle differentiation. Here we analysed expression and localization of FILIP1 and its binding partners filamin-C (FLNc) and microtubule plus-end-binding protein EB3 in differentiating cultured myotubes and adult skeletal muscle. Prior to the development of cross-striated myofibrils, FILIP1 is associated with microtubules and colocalizes with EB3. During further myofibril maturation its localization changes, and FILIP1 localizes to myofibrillar Z-discs together with the actin-binding protein FLNc. Forced contractions of myotubes by electrical pulse stimulation (EPS) induce focal disruptions in myofibrils and translocation of both proteins from Z-discs to these lesions, suggesting a role in induction and/or repair of these structures. The immediate proximity of tyrosylated, dynamic microtubules and EB3 to lesions implies that also these play a role in these processes. This implication is supported by the fact that in nocodazole-treated myotubes that lack functional microtubules, the number of lesions induced by EPS is significantly reduced. In summary, we here show that FILIP1 is a cytolinker protein that is associated with both microtubules and actin filaments, and might play a role in the assembly of myofibrils and their stabilization upon mechanical stress to protect them from damage.


Asunto(s)
Microtúbulos , Miofibrillas , Miofibrillas/metabolismo , Filaminas/análisis , Filaminas/genética , Filaminas/metabolismo , Estrés Mecánico , Microtúbulos/metabolismo , Diferenciación Celular , Músculo Esquelético/metabolismo
2.
Cells ; 13(1)2023 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-38201288

RESUMEN

Synaptopodin-2 (SYNPO2) is a protein associated with the Z-disc in striated muscle cells. It interacts with α-actinin and filamin C, playing a role in Z-disc maintenance under stress by chaperone-assisted selective autophagy (CASA). In smooth muscle cells, SYNPO2 is a component of dense bodies. Furthermore, it has been proposed to play a role in tumor cell proliferation and metastasis in many different kinds of cancers. Alternative transcription start sites and alternative splicing predict the expression of six putative SYNPO2 isoforms differing by extended amino- and/or carboxy-termini. Our analyses at mRNA and protein levels revealed differential expression of SYNPO2 isoforms in cardiac, skeletal and smooth muscle cells. We identified synemin, an intermediate filament protein, as a novel binding partner of the PDZ-domain in the amino-terminal extension of the isoforms mainly expressed in cardiac and smooth muscle cells, and demonstrated colocalization of SYNPO2 and synemin in both cell types. A carboxy-terminal extension, mainly expressed in smooth muscle cells, is sufficient for association with dense bodies and interacts with α-actinin. SYNPO2 therefore represents an additional and novel link between intermediate filaments and the Z-discs in cardiomyocytes and dense bodies in smooth muscle cells, respectively. In pathological skeletal muscle samples, we identified SYNPO2 in the central and intermediate zones of target fibers of patients with neurogenic muscular atrophy, and in nemaline bodies. Our findings help to understand distinct functions of individual SYNPO2 isoforms in different muscle tissues, but also in tumor pathology.


Asunto(s)
Actinina , Miocitos del Músculo Liso , Humanos , Miocitos Cardíacos , Isoformas de Proteínas , Sarcómeros
3.
Nat Commun ; 12(1): 3575, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117258

RESUMEN

An amino acid exchange (P209L) in the HSPB8 binding site of the human co-chaperone BAG3 gives rise to severe childhood cardiomyopathy. To phenocopy the disease in mice and gain insight into its mechanisms, we generated humanized transgenic mouse models. Expression of human BAG3P209L-eGFP in mice caused Z-disc disintegration and formation of protein aggregates. This was accompanied by massive fibrosis resulting in early-onset restrictive cardiomyopathy with increased mortality as observed in patients. RNA-Seq and proteomics revealed changes in the protein quality control system and increased autophagy in hearts from hBAG3P209L-eGFP mice. The mutation renders hBAG3P209L less soluble in vivo and induces protein aggregation, but does not abrogate hBAG3 binding properties. In conclusion, we report a mouse model mimicking the human disease. Our data suggest that the disease mechanism is due to accumulation of hBAG3P209L and mouse Bag3, causing sequestering of components of the protein quality control system and autophagy machinery leading to sarcomere disruption.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Cardiomiopatía Restrictiva/genética , Cardiomiopatía Restrictiva/metabolismo , Animales , Autofagia , Sitios de Unión , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/terapia , Cardiomiopatía Restrictiva/terapia , Niño , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Terapia Genética , Corazón , Proteínas de Choque Térmico , Humanos , Ratones , Ratones Transgénicos , Chaperonas Moleculares/metabolismo , Mutación , Unión Proteica , Proteómica , Sarcómeros/metabolismo
4.
J Muscle Res Cell Motil ; 42(2): 381-397, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33710525

RESUMEN

Hypertrophic cardiomyopathy (HCM) often leads to heart failure. Mutations in sarcomeric proteins are most frequently the cause of HCM but in many patients the gene defect is not known. Here we report on a young man who was diagnosed with HCM shortly after birth. Whole exome sequencing revealed a mutation in the FLNC gene (c.7289C > T; p.Ala2430Val) that was previously shown to cause aggregation of the mutant protein in transfected cells. Myocardial tissue from patients with this mutation has not been analyzed before and thus, the underlying etiology is not well understood. Myocardial tissue of our patient obtained during myectomy at the age of 23 years was analyzed in detail by histochemistry, immunofluorescence staining, electron microscopy and western blot analysis. Cardiac histology showed a pathology typical for myofibrillar myopathy with myofibril disarray and abnormal protein aggregates containing BAG3, desmin, HSPB5 and filamin C. Analysis of sarcomeric and intercalated disc proteins showed focally reduced expression of the gap junction protein connexin43 and Xin-positive sarcomeric lesions in the cardiomyocytes of our patient. In addition, autophagy pathways were altered with upregulation of LC3-II, WIPI1 and HSPB5, 6, 7 and 8. We conclude that the p.Ala2430Val mutation in FLNC most probably is associated with HCM characterized by abnormal intercalated discs, disarray of myofibrils and aggregates containing Z-disc proteins similar to myofibrillar myopathy, which supports the pathological effect of the mutation.


Asunto(s)
Cardiomiopatía Hipertrófica , Filaminas , Miopatías Estructurales Congénitas , Proteínas Adaptadoras Transductoras de Señales , Adulto , Proteínas Reguladoras de la Apoptosis , Cardiomiopatía Hipertrófica/genética , Filaminas/genética , Humanos , Masculino , Mutación , Miocitos Cardíacos , Adulto Joven
5.
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
6.
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
7.
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
8.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA