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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 133(3): 255-270, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37401464

RESUMEN

BACKGROUND: Increasing cardiomyocyte contraction during myocardial stretch serves as the basis for the Frank-Starling mechanism in the heart. However, it remains unclear how this phenomenon occurs regionally within cardiomyocytes, at the level of individual sarcomeres. We investigated sarcomere contractile synchrony and how intersarcomere dynamics contribute to increasing contractility during cell lengthening. METHODS: Sarcomere strain and Ca2+ were simultaneously recorded in isolated left ventricular cardiomyocytes during 1 Hz field stimulation at 37 °C, at resting length and following stepwise stretch. RESULTS: We observed that in unstretched rat cardiomyocytes, differential sarcomere deformation occurred during each beat. Specifically, while most sarcomeres shortened during the stimulus, ≈10% to 20% of sarcomeres were stretched or remained stationary. This nonuniform strain was not traced to regional Ca2+ disparities but rather shorter resting lengths and lower force production in systolically stretched sarcomeres. Lengthening of the cell recruited additional shortening sarcomeres, which increased contractile efficiency as less negative, wasted work was performed by stretched sarcomeres. Given the known role of titin in setting sarcomere dimensions, we next hypothesized that modulating titin expression would alter intersarcomere dynamics. Indeed, in cardiomyocytes from mice with titin haploinsufficiency, we observed greater variability in resting sarcomere length, lower recruitment of shortening sarcomeres, and impaired work performance during cell lengthening. CONCLUSIONS: Graded sarcomere recruitment directs cardiomyocyte work performance, and harmonization of sarcomere strain increases contractility during cell stretch. By setting sarcomere dimensions, titin controls sarcomere recruitment, and its lowered expression in haploinsufficiency mutations impairs cardiomyocyte contractility.


Asunto(s)
Miocitos Cardíacos , Sarcómeros , Ratas , Ratones , Animales , Sarcómeros/metabolismo , Conectina/genética , Conectina/metabolismo , Miocitos Cardíacos/metabolismo , Contracción Miocárdica/fisiología , Miocardio/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(39): 24545-24556, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32929035

RESUMEN

The relationship between oxidative stress and cardiac stiffness is thought to involve modifications to the giant muscle protein titin, which in turn can determine the progression of heart disease. In vitro studies have shown that S-glutathionylation and disulfide bonding of titin fragments could alter the elastic properties of titin; however, whether and where titin becomes oxidized in vivo is less certain. Here we demonstrate, using multiple models of oxidative stress in conjunction with mechanical loading, that immunoglobulin domains preferentially from the distal titin spring region become oxidized in vivo through the mechanism of unfolded domain oxidation (UnDOx). Via oxidation type-specific modification of titin, UnDOx modulates human cardiomyocyte passive force bidirectionally. UnDOx also enhances titin phosphorylation and, importantly, promotes nonconstitutive folding and aggregation of unfolded domains. We propose a mechanism whereby UnDOx enables the controlled homotypic interactions within the distal titin spring to stabilize this segment and regulate myocardial passive stiffness.


Asunto(s)
Miocardio/química , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Proteínas Quinasas/metabolismo , Animales , Elasticidad , Masculino , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocitos Cardíacos/química , Oxidación-Reducción , Fosforilación , Proteínas Quinasas/química , Proteínas Quinasas/genética
3.
Int J Mol Sci ; 24(7)2023 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-37047781

RESUMEN

BICD2 variants have been linked to neurodegenerative disorders like spinal muscular atrophy with lower extremity predominance (SMALED2) or hereditary spastic paraplegia (HSP). Recently, mutations in BICD2 were implicated in myopathies. Here, we present one patient with a known and six patients with novel BICD2 missense variants, further characterizing the molecular landscape of this heterogenous neurological disorder. A total of seven patients were genotyped and phenotyped. Skeletal muscle biopsies were analyzed by histology, electron microscopy, and protein profiling to define pathological hallmarks and pathogenicity markers with consecutive validation using fluorescence microscopy. Clinical and MRI-features revealed a typical pattern of distal paresis of the lower extremities as characteristic features of a BICD2-associated disorder. Histological evaluation showed myopathic features of varying severity including fiber size variation, lipofibromatosis, and fiber splittings. Proteomic analysis with subsequent fluorescence analysis revealed an altered abundance and localization of thrombospondin-4 and biglycan. Our combined clinical, histopathological, and proteomic approaches provide new insights into the pathophysiology of BICD2-associated disorders, confirming a primary muscle cell vulnerability. In this context, biglycan and thrombospondin-4 have been identified, may serve as tissue pathogenicity markers, and might be linked to perturbed protein secretion based on an impaired vesicular transportation.


Asunto(s)
Proteínas Asociadas a Microtúbulos , Atrofia Muscular Espinal , Humanos , Biglicano/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteómica , Atrofia Muscular Espinal/genética , Mutación , Músculo Esquelético/metabolismo
4.
J Biol Chem ; 294(18): 7202-7218, 2019 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-30737279

RESUMEN

Whereas myosin 18B (Myo18B) is known to be a critical sarcomeric protein, the function of myosin 18A (Myo18A) is unclear, although it has been implicated in cell motility and Golgi shape. Here, we show that homozygous deletion (homozygous tm1a, tm1b, or tm1d alleles) of Myo18a in mouse is embryonic lethal. Reminiscent of Myo18b, Myo18a was highly expressed in the embryo heart, and cardiac-restricted Myo18a deletion in mice was embryonic lethal. Surprisingly, using Western blot analysis, we were unable to detect the known isoforms of Myo18A, Myo18Aα and Myo18Aß, in mouse heart using a custom C-terminal antibody. However, alternative anti-Myo18A antibodies detected a larger than expected protein, and RNA-Seq analysis indicated that a novel Myo18A transcript is expressed in mouse ventricular myocytes (and human heart). Cloning and sequencing revealed that this cardiac isoform, denoted Myo18Aγ, lacks the PDZ-containing N terminus of Myo18Aα but includes an alternative N-terminal extension and a long serine-rich C terminus. EGFP-tagged Myo18Aγ expressed in ventricular myocytes localized to the level of A-bands in sarcomeres, and Myo18a knockout embryos at day 10.5 exhibited disorganized sarcomeres with wavy thick filaments. We additionally generated myeloid-restricted Myo18a knockout mice to investigate the role of Myo18A in nonmuscle cells, exemplified by macrophages, which express more Myo18Aß than Myo18Aα, but no defects in cell shape, motility, or Golgi shape were detected. In summary, we have identified a previously unrecognized sarcomere component, a large novel isoform (denoted Myo18Aγ) of Myo18A. Thus, both members of class XVIII myosins are critical components of cardiac sarcomeres.


Asunto(s)
Miocardio/metabolismo , Miosinas/metabolismo , Sarcómeros/metabolismo , Animales , Eliminación de Gen , Genes Letales , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Ratones Noqueados , Miosinas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
5.
Genes Dev ; 26(2): 114-9, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22241783

RESUMEN

Protein lysine methylation is one of the most widespread post-translational modifications in the nuclei of eukaryotic cells. Methylated lysines on histones and nonhistone proteins promote the formation of protein complexes that control gene expression and DNA replication and repair. In the cytoplasm, however, the role of lysine methylation in protein complex formation is not well established. Here we report that the cytoplasmic protein chaperone Hsp90 is methylated by the lysine methyltransferase Smyd2 in various cell types. In muscle, Hsp90 methylation contributes to the formation of a protein complex containing Smyd2, Hsp90, and the sarcomeric protein titin. Deficiency in Smyd2 results in the loss of Hsp90 methylation, impaired titin stability, and altered muscle function. Collectively, our data reveal a cytoplasmic protein network that employs lysine methylation for the maintenance and function of skeletal muscle.


Asunto(s)
Citoplasma/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Músculo Esquelético/metabolismo , Miofibrillas/metabolismo , Animales , Embrión de Pollo , Conectina , Citoplasma/enzimología , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Lisina/metabolismo , Metilación , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pez Cebra
6.
J Mol Cell Cardiol ; 129: 13-26, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30771306

RESUMEN

Cardiac functionality is dependent on a balanced protein turnover. Accordingly, regulated protein decay is critical to maintain cardiac function. Here we demonstrate that deficiency of SPRED2, an intracellular repressor of ERK-MAPK signaling markedly expressed in human heart, resulted in impaired autophagy, heart failure, and shortened lifespan. SPRED2-/- mice showed cardiomyocyte hypertrophy, cardiac fibrosis, impaired electrical excitability, and severe arrhythmias. Mechanistically, cardiomyocyte dysfunction resulted from ERK hyperactivation and dysregulated autophagy, observed as accumulation of vesicles, vacuolar structures, and degenerated mitochondria. The diminished autophagic flux in SPRED2-/- hearts was reflected by a reduced LC3-II/LC3-I ratio and by decreased Atg7, Atg4B and Atg16L expression. Furthermore, the autophagosomal adaptors p62/SQSTM1 and NBR1 and lysosomal Cathepsin D accumulated in SPRED2-/- hearts. In wild-type hearts, SPRED2 interacted physically with p62/SQSTM1, NBR1, and Cathepsin D, indicating that SPRED2 is required for autophagolysosome formation in regular autophagy. Restored inhibition of MAPK signaling by selumetinib led to an increase in autophagic flux in vivo. Therefore, our study identifies SPRED2 as a novel, indispensable regulator of cardiac autophagy. Vice versa, SPRED2 deficiency impairs autophagy, leading to cardiac dysfunction and life-threatening arrhythmias.


Asunto(s)
Arritmias Cardíacas/metabolismo , Autofagia , Mortalidad Prematura , Proteínas Represoras/deficiencia , Adulto , Aldosterona/farmacología , Animales , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Biomarcadores/metabolismo , Presión Sanguínea , Cardiomegalia/complicaciones , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Catepsina D/metabolismo , Colágeno/metabolismo , Fenómenos Electrofisiológicos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Hemodinámica , Humanos , Lisosomas/metabolismo , Lisosomas/ultraestructura , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Fosforilación/efectos de los fármacos , Fosfotreonina/metabolismo , Proteínas Represoras/metabolismo , Vacuolas/metabolismo , Vacuolas/ultraestructura
7.
Circulation ; 135(19): 1832-1847, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28167635

RESUMEN

BACKGROUND: Advancing structural and functional maturation of stem cell-derived cardiomyocytes remains a key challenge for applications in disease modeling, drug screening, and heart repair. Here, we sought to advance cardiomyocyte maturation in engineered human myocardium (EHM) toward an adult phenotype under defined conditions. METHODS: We systematically investigated cell composition, matrix, and media conditions to generate EHM from embryonic and induced pluripotent stem cell-derived cardiomyocytes and fibroblasts with organotypic functionality under serum-free conditions. We used morphological, functional, and transcriptome analyses to benchmark maturation of EHM. RESULTS: EHM demonstrated important structural and functional properties of postnatal myocardium, including: (1) rod-shaped cardiomyocytes with M bands assembled as a functional syncytium; (2) systolic twitch forces at a similar level as observed in bona fide postnatal myocardium; (3) a positive force-frequency response; (4) inotropic responses to ß-adrenergic stimulation mediated via canonical ß1- and ß2-adrenoceptor signaling pathways; and (5) evidence for advanced molecular maturation by transcriptome profiling. EHM responded to chronic catecholamine toxicity with contractile dysfunction, cardiomyocyte hypertrophy, cardiomyocyte death, and N-terminal pro B-type natriuretic peptide release; all are classical hallmarks of heart failure. In addition, we demonstrate the scalability of EHM according to anticipated clinical demands for cardiac repair. CONCLUSIONS: We provide proof-of-concept for a universally applicable technology for the engineering of macroscale human myocardium for disease modeling and heart repair from embryonic and induced pluripotent stem cell-derived cardiomyocytes under defined, serum-free conditions.


Asunto(s)
Células Madre Embrionarias/trasplante , Insuficiencia Cardíaca/terapia , Células Madre Pluripotentes Inducidas/trasplante , Miocitos Cardíacos/trasplante , Ingeniería de Tejidos/métodos , Remodelación Ventricular/fisiología , Animales , Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Insuficiencia Cardíaca/patología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/fisiología , Impresión Tridimensional , Ratas , Ratas Desnudas
9.
Cell Microbiol ; 19(1)2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27341322

RESUMEN

Intoxication of eukaryotic cells by Photorhabdus luminescens toxin TccC3 induces cell rounding and detachment from the substratum within a few hours and compromises a number of cell functions like phagocytosis. Here, we used morphological and biochemical procedures to analyse the mechanism of TccC3 intoxication. Life imaging of TccC3-intoxicated HeLa cells transfected with AcGFP-actin shows condensation of F-actin into large aggregates. Life cell total internal reflection fluorescence (TIRF) microscopy of identically treated HeLa cells confirmed the formation of actin aggregates but also disassembly of F-actin stress fibres. Recombinant TccC3 toxin ADP-ribosylates purified skeletal and non-muscle actin at threonine148 leading to a strong propensity to polymerize and F-actin bundle formation as shown by TIRF and electron microscopy. Native gel electrophoresis shows strongly reduced binding of Thr148-ADP-ribosylated actin to the severing proteins gelsolin and its fragments G1 and G1-3, and to ADF/cofilin. Complexation of actin with these proteins inhibits its ADP-ribosylation. TIRF microscopy demonstrates rapid polymerization of Thr148-ADP-ribosylated actin to curled F-actin bundles even in the presence of thymosin ß4, gelsolin or G1-3. Thr148-ADP-ribosylated F-actin cannot be depolymerized by gelsolin or G1-3 as verified by TIRF, co-sedimentation and electron microscopy and shows reduced treadmilling as indicated by a lack of stimulation of its ATPase activity after addition of cofilin-1.


Asunto(s)
Actinas/metabolismo , Adenosina Difosfato/metabolismo , Toxinas Bacterianas/metabolismo , Photorhabdus/metabolismo , Agregación Patológica de Proteínas , Células Epiteliales/efectos de los fármacos , Células HeLa , Humanos , Microscopía Electrónica , Microscopía Fluorescente
10.
Cereb Cortex ; 27(3): 2318-2334, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27073217

RESUMEN

Cholecystokinin-expressing interneurons (CCK-INs) mediate behavior state-dependent inhibition in cortical circuits and themselves receive strong GABAergic input. However, it remains unclear to what extent GABAB receptors (GABABRs) contribute to their inhibitory control. Using immunoelectron microscopy, we found that CCK-INs in the rat hippocampus possessed high levels of dendritic GABABRs and KCTD12 auxiliary proteins, whereas postsynaptic effector Kir3 channels were present at lower levels. Consistently, whole-cell recordings revealed slow GABABR-mediated inhibitory postsynaptic currents (IPSCs) in most CCK-INs. In spite of the higher surface density of GABABRs in CCK-INs than in CA1 principal cells, the amplitudes of IPSCs were comparable, suggesting that the expression of Kir3 channels is the limiting factor for the GABABR currents in these INs. Morphological analysis showed that CCK-INs were diverse, comprising perisomatic-targeting basket cells (BCs), as well as dendrite-targeting (DT) interneurons, including a previously undescribed DT type. GABABR-mediated IPSCs in CCK-INs were large in BCs, but small in DT subtypes. In response to prolonged activation, GABABR-mediated currents displayed strong desensitization, which was absent in KCTD12-deficient mice. This study highlights that GABABRs differentially control CCK-IN subtypes, and the kinetics and desensitization of GABABR-mediated currents are modulated by KCTD12 proteins.


Asunto(s)
Colecistoquinina/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Potenciales Postsinápticos Inhibidores/fisiología , Interneuronas/metabolismo , Canales de Potasio/metabolismo , Receptores de GABA-A/metabolismo , Animales , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/ultraestructura , Dendritas/metabolismo , Dendritas/ultraestructura , Inmunohistoquímica , Interneuronas/ultraestructura , Masculino , Microscopía Inmunoelectrónica , Técnicas de Placa-Clamp , Ratas Wistar , Técnicas de Cultivo de Tejidos
11.
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
12.
J Mol Cell Cardiol ; 91: 207-14, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26724190

RESUMEN

BACKGROUND: Dilated cardiomyopathy (DCM) could be caused by mutations in more than 40 different genes. However, the pathogenic impact of specific mutations is in most cases unknown complicating the genetic counseling of affected families. Therefore, functional studies could contribute to distinguish pathogenic mutations and benign variants. Here, we present a novel heterozygous DES missense variant (c.407C>T; p.L136P) identified by next generation sequencing in a DCM patient. DES encodes the cardiac intermediate filament protein desmin, which has important functions in mechanical stabilization and linkage of the cell structures in cardiomyocytes. METHODS AND RESULTS: Cell transfection experiments and assembly assays of recombinant desmin in combination with atomic force microscopy were used to investigate the impact of this novel DES variant on filament formation. Desmin-p.L136P forms cytoplasmic aggregates indicating a severe intrinsic filament assembly defect of this mutant. Co-transfection experiments of wild-type and mutant desmin conjugated to different fluorescence proteins revealed a dominant affect of this mutant on filament assembly. These experiments were complemented by apertureless scanning near-field optical microscopy. CONCLUSION: In vitro analysis demonstrated that desmin-p.L136P is unable to form regular filaments and accumulate instead within the cytoplasm. Therefore, we classified DES-p.L136P as a likely pathogenic mutation. In conclusion, the functional characterization of DES-p.L136P might have relevance for the genetic counseling of affected families with similar DES mutations and could contribute to distinguish pathogenic mutations from benign rare variants.


Asunto(s)
Cardiomiopatía Dilatada/genética , Desmina/genética , Filamentos Intermedios/metabolismo , Mutación Missense , Proteínas Recombinantes de Fusión/genética , Secuencia de Aminoácidos , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Desmina/química , Desmina/metabolismo , Desmosomas/metabolismo , Desmosomas/ultraestructura , Femenino , Expresión Génica , Genes Dominantes , Asesoramiento Genético , Células HEK293 , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Filamentos Intermedios/ultraestructura , Masculino , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Linaje , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia
13.
Nature ; 465(7295): 231-5, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20400944

RESUMEN

GABA(B) receptors are the G-protein-coupled receptors for gamma-aminobutyric acid (GABA), the main inhibitory neurotransmitter in the brain. They are expressed in almost all neurons of the brain, where they regulate synaptic transmission and signal propagation by controlling the activity of voltage-gated calcium (Ca(v)) and inward-rectifier potassium (K(ir)) channels. Molecular cloning revealed that functional GABA(B) receptors are formed by the heteromeric assembly of GABA(B1) with GABA(B2) subunits. However, cloned GABA(B(1,2)) receptors failed to reproduce the functional diversity observed with native GABA(B) receptors. Here we show by functional proteomics that GABA(B) receptors in the brain are high-molecular-mass complexes of GABA(B1), GABA(B2) and members of a subfamily of the KCTD (potassium channel tetramerization domain-containing) proteins. KCTD proteins 8, 12, 12b and 16 show distinct expression profiles in the brain and associate tightly with the carboxy terminus of GABA(B2) as tetramers. This co-assembly changes the properties of the GABA(B(1,2)) core receptor: the KCTD proteins increase agonist potency and markedly alter the G-protein signalling of the receptors by accelerating onset and promoting desensitization in a KCTD-subtype-specific manner. Taken together, our results establish the KCTD proteins as auxiliary subunits of GABA(B) receptors that determine the pharmacology and kinetics of the receptor response.


Asunto(s)
Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Receptores de GABA-B/química , Receptores de GABA-B/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Conductividad Eléctrica , Agonistas de Receptores GABA-B , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Cinética , Ratones , Neuronas/metabolismo , Oocitos/metabolismo , Potasio/metabolismo , Canales de Potasio/metabolismo , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Transducción de Señal , Xenopus
14.
Biochim Biophys Acta ; 1833(4): 812-22, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23047121

RESUMEN

Protein lysine methylation controls gene expression and repair of deoxyribonucleic acid in the nucleus but also occurs in the cytoplasm, where the role of this posttranslational modification is less understood. Members of the Smyd protein family of lysine methyltransferases are particularly abundant in the cytoplasm, with Smyd1 and Smyd2 being most highly expressed in the heart and in skeletal muscles. Smyd1 is a crucial myogenic regulator with histone methyltransferase activity but also associates with myosin, which promotes sarcomere assembly. Smyd2 methylates histones and non-histone proteins, such as the tumor suppressors, p53 and retinoblastoma protein, RB. Smyd2 has an intriguing function in the cytoplasm of skeletal myocytes, where it methylates the chaperone Hsp90, thus promoting the interaction of a Smyd2-methyl-Hsp90 complex with the N2A-domain of titin. This complex protects the sarcomeric I-band region and myocyte organization. We briefly summarize some novel functions of Smyd family members, with a focus on Smyd2, and highlight their role in striated muscles and cytoplasmic actions. We then provide experimental evidence that Smyd2 is also important for cardiac function. In the cytoplasm of cardiomyocytes, Smyd2 was found to associate with the sarcomeric I-band region at the titin N2A-domain. Binding to N2A occurred in vitro and in yeast via N-terminal and extreme C-terminal regions of Smyd2. Smyd2-knockdown in zebrafish using an antisense oligonucleotide morpholino approach strongly impaired cardiac performance. We conclude that Smyd2 and presumably several other Smyd family members are lysine methyltransferases which have, next to their nuclear activity, specific regulatory functions in the cytoplasm of heart and skeletal muscle cells. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.


Asunto(s)
Citoplasma/enzimología , N-Metiltransferasa de Histona-Lisina/genética , Proteínas Musculares/metabolismo , Miocardio/enzimología , Miocitos Cardíacos/enzimología , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Sarcómeros/enzimología , Actinina/genética , Actinina/metabolismo , Animales , Animales Recién Nacidos , Núcleo Celular/enzimología , Embrión de Pollo , Conectina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Lisina/genética , Lisina/metabolismo , Ratones , Modelos Moleculares , Proteínas Musculares/genética , Miocardio/citología , Miocitos Cardíacos/citología , Unión Proteica , Proteínas Quinasas/genética , Estructura Terciaria de Proteína , Ratas , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Basic Res Cardiol ; 109(6): 451, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25358972

RESUMEN

Based on evidence that FHL2 (four and a half LIM domains protein 2) negatively regulates cardiac hypertrophy we tested whether FHL2 altered expression or variants could be associated with hypertrophic cardiomyopathy (HCM). HCM is a myocardial disease characterized by left ventricular hypertrophy, diastolic dysfunction and increased interstitial fibrosis and is mainly caused by mutations in genes coding for sarcomeric proteins. FHL2 mRNA level, FHL2 protein level and I-band-binding density were lower in HCM patients than control individuals. Screening of 121 HCM patients without mutations in established disease genes identified 2 novel (T171M, V187L) and 4 known (R177Q, N226N, D268D, P273P) FHL2 variants in unrelated HCM families. We assessed the structural and functional consequences of the nonsynonymous substitutions after adeno-associated viral-mediated gene transfer in cardiac myocytes and in 3D-engineered heart tissue (EHT). Overexpression of FHL2 wild type or nonsynonymous substitutions in cardiac myocytes markedly down-regulated α-skeletal actin and partially blunted hypertrophy induced by phenylephrine or endothelin-1. After gene transfer in EHTs, force and velocity of both contraction and relaxation were higher with T171M and V187L FHL2 variants than wild type under basal conditions. Finally, chronic phenylephrine stimulation depressed EHT function in all groups, but to a lower extent in T171M-transduced EHTs. These data suggest that (1) FHL2 is down-regulated in HCM, (2) both FHL2 wild type and variants partially protected phenylephrine- or endothelin-1-induced hypertrophy in cardiac myocytes, and (3) FHL2 T171M and V187L nonsynonymous variants induced altered EHT contractility. These findings provide evidence that the 2 novel FHL2 variants could increase cardiac function in HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Proteínas con Homeodominio LIM/genética , Proteínas Musculares/genética , Factores de Transcripción/genética , Adolescente , Adulto , Anciano , Animales , Preescolar , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Persona de Mediana Edad , Linaje
16.
Arch Biochem Biophys ; 536(1): 38-45, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23707758

RESUMEN

Two distinct isoforms of the Ca-dependent actin filament severing protein gelsolin were identified in cross-striated muscles of the American lobster. The variants (termed LG1 and LG2) differ by an extension of 18 AA at the C-terminus of LG1, and by two substitutions at AA735 and AA736, the two C-terminal amino acids of LG2. Functional comparison of the isolated and purified proteins revealed gelsolin-typical properties for both with differences in Ca(2+)-sensitivity, LG2 being activated at significant lower Ca-concentration than LG1: Half maximal activation for both filament severing and G-actin binding was ∼4×10(-7)M Ca(2+) for LG2 vs. ∼2×10(-6)M Ca(2+) for LG1. This indicates a differential activation for the two isoproteins in vivo where they are present in almost equal amounts in the muscle cell. Structure prediction modeling on the basis of the known structure of mammalian gelsolin shows that LG2 lacks the C-terminal alpha-helix which is involved in contact formation between domains G6 and G2. In both mammalian gelsolin and LG1, this "latch bridge" is assumed to play a critical role in Ca(2+)-activation by keeping gelsolin in a closed, inactive conformation at low [Ca(2+)]. In LG2, the reduced contact between G6 and G2 may be responsible for its activation at low Ca(2+)-concentration.


Asunto(s)
Proteínas de Artrópodos/análisis , Proteínas de Artrópodos/metabolismo , Calcio/metabolismo , Gelsolina/análisis , Gelsolina/metabolismo , Nephropidae/metabolismo , Actinas/análisis , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/genética , Gelsolina/genética , Modelos Moleculares , Datos de Secuencia Molecular , Músculo Estriado/química , Músculo Estriado/metabolismo , Nephropidae/química , Nephropidae/genética , Conformación Proteica , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína , ARN Mensajero/genética
17.
Cell Commun Signal ; 11: 56, 2013 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-23937664

RESUMEN

BACKGROUND: In the heart, cytoplasmic actin networks are thought to have important roles in mechanical support, myofibrillogenesis, and ion channel function. However, subcellular localization of cytoplasmic actin isoforms and proteins involved in the modulation of the cytoplasmic actin networks are elusive. Mena and VASP are important regulators of actin dynamics. Due to the lethal phenotype of mice with combined deficiency in Mena and VASP, however, distinct cardiac roles of the proteins remain speculative. In the present study, we analyzed the physiological functions of Mena and VASP in the heart and also investigated the role of the proteins in the organization of cytoplasmic actin networks. RESULTS: We generated a mouse model, which simultaneously lacks Mena and VASP in the heart. Mena/VASP double-deficiency induced dilated cardiomyopathy and conduction abnormalities. In wild-type mice, Mena and VASP specifically interacted with a distinct αII-Spectrin splice variant (SH3i), which is in cardiomyocytes exclusively localized at Z- and intercalated discs. At Z- and intercalated discs, Mena and ß-actin localized to the edges of the sarcomeres, where the thin filaments are anchored. In Mena/VASP double-deficient mice, ß-actin networks were disrupted and the integrity of Z- and intercalated discs was markedly impaired. CONCLUSIONS: Together, our data suggest that Mena, VASP, and αII-Spectrin assemble cardiac multi-protein complexes, which regulate cytoplasmic actin networks. Conversely, Mena/VASP deficiency results in disrupted ß-actin assembly, Z- and intercalated disc malformation, and induces dilated cardiomyopathy and conduction abnormalities.


Asunto(s)
Actinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Microfilamentos/metabolismo , Miocitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , Espectrina/metabolismo , Animales , Cardiomiopatía Dilatada/metabolismo , Moléculas de Adhesión Celular/genética , Citoplasma/metabolismo , Proteínas del Citoesqueleto/genética , Sistema de Conducción Cardíaco/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Fosfoproteínas/genética , Espectrina/química , Dominios Homologos src
18.
Int Orthop ; 37(1): 77-82, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23142861

RESUMEN

PURPOSE: We present a new technique of arthroscopic-assisted AC-hook plate fixation for acromioclavicular joint dislocation with all the advantages of minimally invasive surgery and the possibility to treat concomitant pathologies. METHODS: Initially a glenohumeral arthroscopy is performed to address concomitant intra-articular injuries. Under subacromial visualisation the drill hole for the hook of the plate can be exactly positioned in the acromion. The hook plate is put in place under visual control. RESULTS: The initial results (n = 3) are promising with good to excellent results in the Constant score [90.5 (range 82-100)] in all cases studied. The cross-body test was slightly positive in one case. The median follow-up time after the index procedure was seven months (range five to ten). CONCLUSIONS: In conclusion, arthroscopic-assisted reconstruction of acromioclavicular joint separation is feasible and may provide patients with all the benefits of AC-hook fixation with decreased risks related to open surgery. The described technique is recommended for all surgeons familiar with arthroscopic surgery.


Asunto(s)
Articulación Acromioclavicular/cirugía , Artroplastia de Reemplazo/métodos , Artroscopía/métodos , Placas Óseas , Prótesis Articulares , Luxación del Hombro/cirugía , Articulación Acromioclavicular/lesiones , Actividades Cotidianas , Artroplastia de Reemplazo/instrumentación , Humanos , Resultado del Tratamiento
19.
Int Orthop ; 37(4): 667-72, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23359099

RESUMEN

PURPOSE: A biomechanical study was performed to test the hypothesis that a new anatomically preformed, thinner, soft-tissue protecting plate system for distal humeral fractures (Tifix®-hybridplate [HP]) would show comparable results in the quasi-static and dynamic testings compared to two conventional implants: The 3.5-mm reconstruction plate (RP) providing primary stability with normal bone mineral density (BMD), and a multidirectional locking plate (Tifix(®)-plate [P]) which can be used with poor bone quality. METHODS: The Tifix(®)-HP was developed by the working group. The biomechanical testing was performed on a C2-fracture-model in 24 synthetic humeri. Three groups, each with eight bone-implant-constructs, were analysed in quasi-static and dynamic tests. RESULTS: The quasi-static measurements showed that under extension loading both locking plates (Tifix(®)-P, Tifix(®)-HP) were significantly stiffer than the reconstruction plate, and that the Tifix(®)-HP had a significantly lower stiffness than the two other implants under flexion loading. In the dynamic tests the Tifix(®)-P allowed significantly less fracture motion compared to the Tifix(®)-HP and the reconstruction plate. In an osteopaenic bone model locking plates failed only under much higher dynamic force than the reconstruction plate. The reconstruction plate and the Tifix(®)-P always failed through screw loosening, whereas the newly developed Tifix(®)-HP showed screw loosening in only one third of cases. CONCLUSION: The hypothesis that the newly designed plate system showed comparable results in the quasi-static and dynamic tests compared to the conventional implants with a significantly lower implant volume and thickness was confirmed.


Asunto(s)
Placas Óseas , Fijación Interna de Fracturas/instrumentación , Fijación Interna de Fracturas/métodos , Fracturas del Húmero/cirugía , Fijadores Internos , Fenómenos Biomecánicos , Tornillos Óseos , Humanos , Húmero/lesiones , Húmero/cirugía , Ensayo de Materiales , Estrés Mecánico
20.
Cells ; 12(9)2023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-37174721

RESUMEN

Myofibrillar myopathies (MFM) are a group of chronic muscle diseases pathophysiologically characterized by accumulation of protein aggregates and structural failure of muscle fibers. A subtype of MFM is caused by heterozygous mutations in the filamin C (FLNC) gene, exhibiting progressive muscle weakness, muscle structural alterations and intracellular protein accumulations. Here, we characterize in depth the pathogenicity of two novel truncating FLNc variants (p.Q1662X and p.Y2704X) and assess their distinct effect on FLNc stability and distribution as well as their impact on protein quality system (PQS) pathways. Both variants cause a slowly progressive myopathy with disease onset in adulthood, chronic myopathic alterations in muscle biopsy including the presence of intracellular protein aggregates. Our analyses revealed that p.Q1662X results in FLNc haploinsufficiency and p.Y2704X in a dominant-negative FLNc accumulation. Moreover, both protein-truncating variants cause different PQS alterations: p.Q1662X leads to an increase in expression of several genes involved in the ubiquitin-proteasome system (UPS) and the chaperone-assisted selective autophagy (CASA) system, whereas p.Y2704X results in increased abundance of proteins involved in UPS activation and autophagic buildup. We conclude that truncating FLNC variants might have different pathogenetic consequences and impair PQS function by diverse mechanisms and to varying extents. Further studies on a larger number of patients are necessary to confirm our observations.


Asunto(s)
Miopatías Estructurales Congénitas , Agregado de Proteínas , Humanos , Filaminas/genética , Filaminas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA