Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Trends Pharmacol Sci ; 45(3): 191-192, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38302402

RESUMEN

Muscle contraction is orchestrated by the well-understood thin filaments and the markedly complex thick filaments. Studies by Dutta et al. and Tamborrini et al., discussed here, have unravelled the structure of the mammalian heart thick filament in exquisite near-atomic detail and pave the way for understanding physiological modulation pathways and mutation-induced dysfunction and for designing potential drugs to modify defects.


Asunto(s)
Miocardio , Sarcómeros , Humanos , Animales , Miocardio/metabolismo , Sarcómeros/metabolismo , Mamíferos
3.
J Muscle Res Cell Motil ; 44(3): 153-163, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37173591

RESUMEN

Early x-ray diffraction studies of muscle revealed spacings larger than the basic thick filament lattice spacing and led to a number of speculations on the mutual rotations of the filaments in the myosin lattice. The nature of the arrangements of the filaments was resolved by John Squire and Pradeep Luther using careful electron microscopy and image analysis. The intriguing disorder in the rotations, that they termed the myosin superlattice, remained a curiosity, until work with Rick Millane and colleagues showed a connection to "geometric frustration," a well-known phenomenon in statistical and condensed matter physics. In this review, we describe how this connection gives a satisfying physical basis for the myosin superlattice, and how recent work has shown relationships to muscle mechanical behaviour.


Asunto(s)
Frustación , Vertebrados , Animales , Miosinas , Citoesqueleto , Sarcómeros
4.
J Muscle Res Cell Motil ; 44(3): 165-178, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37115473

RESUMEN

Myosin binding protein C (MyBP-C) is an accessory protein of the thick filament in vertebrate cardiac muscle arranged over 9 stripes of intervals of 430 Å in each half of the A-band in the region called the C-zone. Mutations in cardiac MyBP-C are a leading cause of hypertrophic cardiomyopathy the mechanism of which is unknown. It is a rod-shaped protein composed of 10 or 11 immunoglobulin- or fibronectin-like domains labelled C0 to C10 which binds to the thick filament via its C-terminal region. MyBP-C regulates contraction in a phosphorylation dependent fashion that may be through binding of its N-terminal domains with myosin or actin. Understanding the 3D organisation of MyBP-C in the sarcomere environment may provide new light on its function. We report here the fine structure of MyBP-C in relaxed rat cardiac muscle by cryo-electron tomography and subtomogram averaging of refrozen Tokuyasu cryosections. We find that on average MyBP-C connects via its distal end to actin across a disc perpendicular to the thick filament. The path of MyBP-C suggests that the central domains may interact with myosin heads. Surprisingly MyBP-C at Stripe 4 is different; it has weaker density than the other stripes which could result from a mainly axial or wavy path. Given that the same feature at Stripe 4 can also be found in several mammalian cardiac muscles and in some skeletal muscles, our finding may have broader implication and significance. In the D-zone, we show the first demonstration of myosin crowns arranged on a uniform 143 Å repeat.


Asunto(s)
Actinas , Tomografía con Microscopio Electrónico , Ratas , Animales , Actinas/metabolismo , Miocardio/metabolismo , Miosinas/metabolismo , Citoesqueleto de Actina/metabolismo , Mamíferos/metabolismo
5.
J R Soc Interface ; 18(185): 20210585, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34905966

RESUMEN

Geometric frustration results from an incompatibility between minimum energy arrangements and the geometry of a system, and gives rise to interesting and novel phenomena. Here, we report geometric frustration in a native biological macromolecular system---vertebrate muscle. We analyse the disorder in the myosin filament rotations in the myofibrils of vertebrate striated (skeletal and cardiac) muscle, as seen in thin-section electron micrographs, and show that the distribution of rotations corresponds to an archetypical geometrically frustrated system---the triangular Ising antiferromagnet. Spatial correlations are evident out to at least six lattice spacings. The results demonstrate that geometric frustration can drive the development of structure in complex biological systems, and may have implications for the nature of the actin--myosin interactions involved in muscle contraction. Identification of the distribution of myosin filament rotations with an Ising model allows the extensive results on the latter to be applied to this system. It shows how local interactions (between adjacent myosin filaments) can determine long-range order and, conversely, how observations of long-range order (such as patterns seen in electron micrographs) can be used to estimate the energetics of these local interactions. Furthermore, since diffraction by a disordered system is a function of the second-order statistics, the derived correlations allow more accurate diffraction calculations, which can aid in interpretation of X-ray diffraction data from muscle specimens for structural analysis.


Asunto(s)
Frustación , Miosinas , Animales , Microscopía Electrónica , Contracción Muscular , Músculos , Vertebrados , Difracción de Rayos X
6.
J Cachexia Sarcopenia Muscle ; 11(1): 169-194, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31647200

RESUMEN

BACKGROUND: Myopalladin (MYPN) is a striated muscle-specific, immunoglobulin-containing protein located in the Z-line and I-band of the sarcomere as well as the nucleus. Heterozygous MYPN gene mutations are associated with hypertrophic, dilated, and restrictive cardiomyopathy, and homozygous loss-of-function truncating mutations have recently been identified in patients with cap myopathy, nemaline myopathy, and congenital myopathy with hanging big toe. METHODS: Constitutive MYPN knockout (MKO) mice were generated, and the role of MYPN in skeletal muscle was studied through molecular, cellular, biochemical, structural, biomechanical, and physiological studies in vivo and in vitro. RESULTS: MKO mice were 13% smaller compared with wild-type controls and exhibited a 48% reduction in myofibre cross-sectional area (CSA) and significantly increased fibre number. Similarly, reduced myotube width was observed in MKO primary myoblast cultures. Biomechanical studies showed reduced isometric force and power output in MKO mice as a result of the reduced CSA, whereas the force developed by each myosin molecular motor was unaffected. While the performance by treadmill running was similar in MKO and wild-type mice, MKO mice showed progressively decreased exercise capability, Z-line damage, and signs of muscle regeneration following consecutive days of downhill running. Additionally, MKO muscle exhibited progressive Z-line widening starting from 8 months of age. RNA-sequencing analysis revealed down-regulation of serum response factor (SRF)-target genes in muscles from postnatal MKO mice, important for muscle growth and differentiation. The SRF pathway is regulated by actin dynamics as binding of globular actin to the SRF-cofactor myocardin-related transcription factor A (MRTF-A) prevents its translocation to the nucleus where it binds and activates SRF. MYPN was found to bind and bundle filamentous actin as well as interact with MRTF-A. In particular, while MYPN reduced actin polymerization, it strongly inhibited actin depolymerization and consequently increased MRTF-A-mediated activation of SRF signalling in myogenic cells. Reduced myotube width in MKO primary myoblast cultures was rescued by transduction with constitutive active SRF, demonstrating that MYPN promotes skeletal muscle growth through activation of the SRF pathway. CONCLUSIONS: Myopalladin plays a critical role in the control of skeletal muscle growth through its effect on actin dynamics and consequently the SRF pathway. In addition, MYPN is important for the maintenance of Z-line integrity during exercise and aging. These results suggest that muscle weakness in patients with biallelic MYPN mutations may be associated with reduced myofibre CSA and SRF signalling and that the disease phenotype may be aggravated by exercise.


Asunto(s)
Proteínas Musculares/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Factor de Respuesta Sérica/efectos de los fármacos , Animales , Femenino , Humanos , Ratones , Ratones Noqueados , Proteínas Musculares/farmacología
8.
Proc Natl Acad Sci U S A ; 116(31): 15534-15539, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31320587

RESUMEN

Striated muscle enables movement in all animals by the contraction of myriads of sarcomeres joined end to end by the Z-bands. The contraction is due to tension generated in each sarcomere between overlapping arrays of actin and myosin filaments. At the Z-band, actin filaments from adjoining sarcomeres overlap and are cross-linked in a regular pattern mainly by the protein α-actinin. The Z-band is dynamic, reflected by the 2 regular patterns seen in transverse section electron micrographs; the so-called small-square and basketweave forms. Although these forms are attributed, respectively, to relaxed and actively contracting muscles, the basketweave form occurs in certain relaxed muscles as in the muscle studied here. We used electron tomography and subtomogram averaging to derive the 3D structure of the Z-band in the swimbladder sonic muscle of type I male plainfin midshipman fish (Porichthys notatus), into which we docked the crystallographic structures of actin and α-actinin. The α-actinin links run diagonally between connected pairs of antiparallel actin filaments and are oriented at an angle of about 25° away from the actin filament axes. The slightly curved and flattened structure of the α-actinin rod has a distinct fit into the map. The Z-band model provides a detailed understanding of the role of α-actinin in transmitting tension between actin filaments in adjoining sarcomeres.


Asunto(s)
Actinina/metabolismo , Sacos Aéreos/metabolismo , Proteínas de Peces/metabolismo , Peces/metabolismo , Contracción Muscular , Sarcómeros/metabolismo , Animales , Masculino
9.
J Mol Cell Cardiol ; 129: 236-246, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30862451

RESUMEN

Cardiac myosin binding protein-C (cMyBP-C) phosphorylation is essential for normal heart function and protects the heart from ischemia-reperfusion (I/R) injury. It is known that protein kinase-A (PKA)-mediated phosphorylation of cMyBP-C prevents I/R-dependent proteolysis, whereas dephosphorylation of cMyBP-C at PKA sites correlates with its degradation. While sites on cMyBP-C associated with phosphorylation and proteolysis co-localize, the mechanisms that link cMyBP-C phosphorylation and proteolysis during cardioprotection are not well understood. Therefore, we aimed to determine if abrogation of cMyBP-C proteolysis in association with calpain, a calcium-activated protease, confers cardioprotection during I/R injury. Calpain is activated in both human ischemic heart samples and ischemic mouse myocardium where cMyBP-C is dephosphorylated and undergoes proteolysis. Moreover, cMyBP-C is a substrate for calpain proteolysis and cleaved by calpain at residues 272-TSLAGAGRR-280, a domain termed as the calpain-target site (CTS). Cardiac-specific transgenic (Tg) mice in which the CTS motif was ablated were bred into a cMyBP-C null background. These Tg mice were conclusively shown to possess a normal basal structure and function by analysis of histology, electron microscopy, immunofluorescence microscopy, Q-space MRI of tissue architecture, echocardiography, and hemodynamics. However, the genetic ablation of the CTS motif conferred resistance to calpain-mediated proteolysis of cMyBP-C. Following I/R injury, the loss of the CTS reduced infarct size compared to non-transgenic controls. Collectively, these findings demonstrate the physiological significance of calpain-targeted cMyBP-C proteolysis and provide a rationale for studying inhibition of calpain-mediated proteolysis of cMyBP-C as a therapeutic target for cardioprotection.


Asunto(s)
Calpaína/metabolismo , Cardiotónicos/metabolismo , Proteínas Portadoras/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Animales , Femenino , Pruebas de Función Cardíaca , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Fosforilación , Proteolisis
11.
Am J Physiol Heart Circ Physiol ; 311(2): H465-75, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27233767

RESUMEN

Myocardial remodeling in response to chronic myocardial infarction (CMI) progresses through two phases, hypertrophic "compensation" and congestive "decompensation." Nothing is known about the ability of uninfarcted myocardium to produce force, velocity, and power during these clinical phases, even though adaptation in these regions likely drives progression of compensation. We hypothesized that enhanced cross-bridge-level contractility underlies mechanical compensation and is controlled in part by changes in the phosphorylation states of myosin regulatory proteins. We induced CMI in rats by left anterior descending coronary artery ligation. We then measured mechanical performance in permeabilized ventricular trabecula taken distant from the infarct zone and assayed myosin regulatory protein phosphorylation in each individual trabecula. During full activation, the compensated myocardium produced twice as much power and 31% greater isometric force compared with noninfarcted controls. Isometric force during submaximal activations was raised >2.4-fold, while power was 2-fold greater. Electron and confocal microscopy demonstrated that these mechanical changes were not a result of increased density of contractile protein and therefore not an effect of tissue hypertrophy. Hence, sarcomere-level contractile adaptations are key determinants of enhanced trabecular mechanics and of the overall cardiac compensatory response. Phosphorylation of myosin regulatory light chain (RLC) increased and remained elevated post-MI, while phosphorylation of myosin binding protein-C (MyBP-C) was initially depressed but then increased as the hearts became decompensated. These sensitivities to CMI are in accordance with phosphorylation-dependent regulatory roles for RLC and MyBP-C in crossbridge function and with compensatory adaptation in force and power that we observed in post-CMI trabeculae.


Asunto(s)
Proteínas Portadoras/metabolismo , Contracción Miocárdica/fisiología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Sarcómeros/metabolismo , Adaptación Fisiológica , Animales , Vasos Coronarios/cirugía , Ligadura , Masculino , Microscopía Confocal , Microscopía Electrónica , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/ultraestructura , Fosforilación , Ratas , Ratas Sprague-Dawley , Sarcómeros/fisiología , Sarcómeros/ultraestructura
12.
PLoS One ; 11(2): e0145937, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26840448

RESUMEN

INTRODUCTION: Left ventricular dysfunction is a frequent and potentially severe side effect of many tyrosine kinase inhibitors (TKI). The mode of toxicity is not identified, but may include impairment of mitochondrial or sarcomeric function, autophagy or angiogenesis, either as an on-target or off-target mechanism. METHODS AND RESULTS: We studied concentration-response curves and time courses for nine TKIs in three-dimensional, force generating engineered heart tissue (EHT) from neonatal rat heart cells. We detected a concentration- and time-dependent decline in contractile force for gefitinib, lapatinib, sunitinib, imatinib, sorafenib, vandetanib and lestaurtinib and no decline in contractile force for erlotinib and dasatinib after 96 hours of incubation. The decline in contractile force was associated with an impairment of autophagy (LC3 Western blot) and appearance of autophagolysosomes (transmission electron microscopy). CONCLUSION: This study demonstrates the feasibility to study TKI-mediated force effects in EHTs and identifies an association between a decline in contractility and inhibition of autophagic flux.


Asunto(s)
Cardiotoxinas/farmacología , Contracción Miocárdica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Ingeniería de Tejidos , Animales , Autofagia/efectos de los fármacos , Estudios de Factibilidad , Miocitos Cardíacos/citología , Miocitos Cardíacos/diagnóstico por imagen , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Endogámicas Lew , Ratas Wistar , Ingeniería de Tejidos/métodos , Ultrasonografía
13.
J Mol Biol ; 427(22): 3527-3537, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26362007

RESUMEN

The Z-band in vertebrate striated muscle crosslinks actin filaments of opposite polarity from adjoining sarcomeres and transmits tension along myofibrils during muscular contraction. It is also the location of a number of proteins involved in signalling and myofibrillogenesis; mutations in these proteins lead to myopathies. Understanding the high-resolution structure of the Z-band will help us understand its role in muscle contraction and the role of these proteins in the function of muscle. The appearance of the Z-band in transverse-section electron micrographs typically resembles a small-square lattice or a basketweave appearance. In longitudinal sections, the Z-band width varies more with muscle type than species: slow skeletal and cardiac muscles have wider Z-bands than fast skeletal muscles. As the Z-band is periodic, Fourier methods have previously been used for three-dimensional structural analysis. To cope with variations in the periodic structure of the Z-band, we have used subtomogram averaging of tomograms of rat cardiac muscle in which subtomograms are extracted and compared and similar ones are averaged. We show that the Z-band comprises four to six layers of links, presumably α-actinin, linking antiparallel overlapping ends of the actin filaments from the adjoining sarcomeres. The reconstruction shows that the terminal 5-7nm of the actin filaments within the Z-band is devoid of any α-actinin links and is likely to be the location of capping protein CapZ.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Citoesqueleto/metabolismo , Miocardio/ultraestructura , Sarcómeros/ultraestructura , Actinina/metabolismo , Animales , Conectina/metabolismo , Microscopía Electrónica , Modelos Moleculares , Proteínas Musculares/metabolismo , Miocardio/química , Miocardio/metabolismo , Ratas , Sarcómeros/química , Sarcómeros/metabolismo
14.
Nanotoxicology ; 9(3): 356-64, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25030098

RESUMEN

Nanoparticles (NPs) may come into contact with circulating blood elements including platelets following inhalation and translocation from the airways to the bloodstream or during proposed medical applications. Studies with model polystyrene latex nanoparticles (PLNPs) have shown that NPs are able to induce platelet aggregation in vitro suggesting a poorly defined potential mechanism of increased cardiovascular risk upon NP exposure. We aimed to provide insight into the mechanisms by which NPs may increase cardiovascular risk by determining the impact of a range of concentrations of PLNPs on platelet activation in vitro and in vivo and identifying the signaling events driving NP-induced aggregation. Model PLNPs of varying nano-size (50 and 100 nm) and surface chemistry [unmodified (uPLNP), amine-modified (aPLNP) and carboxyl-modified (cPLNP)] were therefore examined using in vitro platelet aggregometry and an established mouse model of platelet thromboembolism. Most PLNPs tested induced GPIIb/IIIa-mediated platelet aggregation with potencies that varied with both surface chemistry and nano-size. Aggregation was associated with signaling events, such as granule secretion and release of secondary agonists, indicative of conventional agonist-mediated aggregation. Platelet aggregation was associated with the physical interaction of PLNPs with the platelet membrane or internalization. 50 nm aPLNPs acted through a distinct mechanism involving the physical bridging of adjacent non-activated platelets leading to enhanced agonist-induced aggregation in vitro and in vivo. Our study suggests that should they translocate the pulmonary epithelium, or be introduced into the blood, NPs may increase the risk of platelet-driven events by inducing or enhancing platelet aggregation via mechanisms that are determined by their distinct combination of nano-size and surface chemistry.


Asunto(s)
Nanopartículas/química , Agregación Plaquetaria/efectos de los fármacos , Poliestirenos/química , Animales , Técnicas In Vitro , Ratones
15.
Biology (Basel) ; 3(4): 846-65, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25478994

RESUMEN

Myosin filaments in vertebrate striated muscle have a long roughly cylindrical backbone with cross-bridge projections on the surfaces of both halves except for a short central bare zone. In the middle of this central region the filaments are cross-linked by the M-band which holds them in a well-defined hexagonal lattice in the muscle A-band. During muscular contraction the M-band-defined rotation of the myosin filaments around their long axes influences the interactions that the cross-bridges can make with the neighbouring actin filaments. We can visualise this filament rotation by electron microscopy of thin cross-sections in the bare-region immediately adjacent to the M-band where the filament profiles are distinctly triangular. In the muscles of teleost fishes, the thick filament triangular profiles have a single orientation giving what we call the simple lattice. In other vertebrates, for example all the tetrapods, the thick filaments have one of two orientations where the triangles point in opposite directions (they are rotated by 60° or 180°) according to set rules. Such a distribution cannot be developed in an ordered fashion across a large 2D lattice, but there are small domains of superlattice such that the next-nearest neighbouring thick filaments often have the same orientation. We believe that this difference in the lattice forms can lead to different contractile behaviours. Here we provide a historical review, and when appropriate cite recent work related to the emergence of the simple and superlattice forms by examining the muscles of several species ranging back to primitive vertebrates and we discuss the functional differences that the two lattice forms may have.

16.
J Mol Cell Cardiol ; 74: 151-61, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24852842

RESUMEN

Spontaneously beating engineered heart tissue (EHT) represents an advanced in vitro model for drug testing and disease modeling, but cardiomyocytes in EHTs are less mature and generate lower forces than in the adult heart. We devised a novel pacing system integrated in a setup for videooptical recording of EHT contractile function over time and investigated whether sustained electrical field stimulation improved EHT properties. EHTs were generated from neonatal rat heart cells (rEHT, n=96) or human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (hEHT, n=19). Pacing with biphasic pulses was initiated on day 4 of culture. REHT continuously paced for 16-18 days at 0.5Hz developed 2.2× higher forces than nonstimulated rEHT. This was reflected by higher cardiomyocyte density in the center of EHTs, increased connexin-43 abundance as investigated by two-photon microscopy and remarkably improved sarcomere ultrastructure including regular M-bands. Further signs of tissue maturation include a rightward shift (to more physiological values) of the Ca(2+)-response curve, increased force response to isoprenaline and decreased spontaneous beating activity. Human EHTs stimulated at 2Hz in the first week and 1.5Hz thereafter developed 1.5× higher forces than nonstimulated hEHT on day 14, an ameliorated muscular network of longitudinally oriented cardiomyocytes and a higher cytoplasm-to-nucleus ratio. Taken together, continuous pacing improved structural and functional properties of rEHTs and hEHTs to an unprecedented level. Electrical stimulation appears to be an important step toward the generation of fully mature EHT.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Miocardio/citología , Miocitos Cardíacos/citología , Técnicas de Cultivo de Tejidos/métodos , Ingeniería de Tejidos/métodos , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Calcio/metabolismo , Diferenciación Celular , Núcleo Celular/fisiología , Núcleo Celular/ultraestructura , Conexina 43/metabolismo , Citoplasma/fisiología , Citoplasma/ultraestructura , Estimulación Eléctrica , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Isoproterenol/farmacología , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Sarcómeros/fisiología , Sarcómeros/ultraestructura
17.
Cytoskeleton (Hoboken) ; 71(5): 294-301, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24616277

RESUMEN

Primary ciliary dyskinesia is an autosomal recessive disorder affecting the motility of cilia. There are a range of ultrastructural ciliary defects that lead to associated clinical symptoms including ineffective mucus clearance, reduced lung function, infertility, and left-right isomerism. Mutations in radial spoke head proteins are a known cause of primary ciliary dyskinesia. Ultrastructually these defects are identified by a portion of cilia lacking a central pair and transposed outer microtubular doublets. We have repeatedly observed an intermittent loss of the central pair in patients with a transposition defect. To further understand the central pair changes in these radial spoke head mutations we employ electron tomography, a high resolution electron microscope technique, to elucidate in three dimensions the ultrastructural arrangements caused by mutation of the RSPH4A gene. We thereby provide an explanation of the structures observed by conventional electron microscopy studies. We demonstrate that the central pair can be present within the cilium. In some cilia, the central pair rotates at the base of the axoneme. We propose that it is this rotation that gives rise to an intermittent appearance of the central pair when viewed under conventional electron microscopy. We discuss the potential causes and consequences of these findings. © 2014 Wiley Periodicals, Inc.


Asunto(s)
Cilios/ultraestructura , Síndrome de Kartagener/patología , Microtúbulos/ultraestructura , Adolescente , Niño , Preescolar , Tomografía con Microscopio Electrónico , Femenino , Humanos , Interpretación de Imagen Asistida por Computador , Masculino , Adulto Joven
18.
Pflugers Arch ; 466(3): 425-31, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24413886

RESUMEN

Myosin-binding protein-C (MyBP-C) is an accessory protein of the myosin filaments of vertebrate striated muscle. In the heart, it plays a key role in modulating contractility in response to ß-adrenergic stimulation. Mutations in the cardiac isoform (cMyBP-C) are a leading cause of inherited hypertrophic cardiomyopathy. Understanding cMyBP-C function and its role in disease requires knowledge of the structure of the molecule, its organization in the sarcomere, and its interactions with other sarcomeric proteins. Here we review the main structural features of this modular, elongated molecule and the properties of some of its key domains. We describe observations suggesting that the bulk of the molecule extends perpendicular to the thick filament, enabling it to reach neighboring thin filaments in the sarcomere. We review structural and functional evidence for interaction of its N-terminal domains with actin and how this may modulate thin filament activation. We also discuss the effects that phosphorylation of cMyBP-C has on some of these structural features and how this might relate to cMyBP-C function in the beating heart.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Microfilamentos/metabolismo , Sarcómeros/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas Portadoras/química , Humanos , Datos de Secuencia Molecular , Unión Proteica , Sarcómeros/ultraestructura
19.
J Cell Sci ; 126(Pt 23): 5477-89, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24046450

RESUMEN

Nemaline myopathy (NM) is a congenital myopathy with an estimated incidence of 150,000 live births. It is caused by mutations in thin filament components, including nebulin, which accounts for about 50% of the cases. The identification of NM cases with nonsense mutations resulting in loss of the extreme C-terminal SH3 domain of nebulin suggests an important role of the nebulin SH3 domain, which is further supported by the recent demonstration of its role in IGF-1-induced sarcomeric actin filament formation through targeting of N-WASP to the Z-line. To provide further insights into the functional significance of the nebulin SH3 domain in the Z-disk and to understand the mechanisms by which truncations of nebulin lead to NM, we took two approaches: (1) an affinity-based proteomic screening to identify novel interaction partners of the nebulin SH3 domain; and (2) generation and characterization of a novel knockin mouse model with a premature stop codon in the nebulin gene, eliminating its C-terminal SH3 domain (NebΔSH3 mouse). Surprisingly, detailed analyses of NebΔSH3 mice revealed no structural or histological skeletal muscle abnormalities and no changes in gene expression or localization of interaction partners of the nebulin SH3 domain, including myopalladin, palladin, zyxin and N-WASP. Also, no significant effect on peak isometric stress production, passive tensile stress or Young's modulus was found. However, NebΔSH3 muscle displayed a slightly altered force-frequency relationship and was significantly more susceptible to eccentric contraction-induced injury, suggesting that the nebulin SH3 domain protects against eccentric contraction-induced injury and possibly plays a role in fine-tuning the excitation-contraction coupling mechanism.


Asunto(s)
Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Animales , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Módulo de Elasticidad/fisiología , Acoplamiento Excitación-Contracción/fisiología , Femenino , Expresión Génica , Humanos , Contracción Isométrica/fisiología , Masculino , Ratones , Proteínas Musculares/química , Proteínas Musculares/deficiencia , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Miopatías Nemalínicas/genética , Miopatías Nemalínicas/metabolismo , Miopatías Nemalínicas/patología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Resistencia a la Tracción/fisiología , Soporte de Peso/fisiología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Zixina/genética , Zixina/metabolismo
20.
Hum Mol Genet ; 22(24): 4967-77, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23873045

RESUMEN

Myosin-binding protein C1 (MYBPC1) is an abundant skeletal muscle protein that is expressed predominantly in slow-twitch muscle fibers. Human MYBPC1 mutations are associated with distal arthrogryposis type 1 and lethal congenital contracture syndrome type 4. As MYBPC1 function is incompletely understood, the mechanism by which human mutations result in contractures is unknown. Here, we demonstrate using antisense morpholino knockdown, that mybpc1 is required for embryonic motor activity and survival in a zebrafish model of arthrogryposis. Mybpc1 morphant embryos have severe body curvature, cardiac edema, impaired motor excitation and are delayed in hatching. Myofibril organization is selectively impaired in slow skeletal muscle and sarcomere numbers are greatly reduced in mybpc1 knockdown embryos, although electron microscopy reveals normal sarcomere structure. To evaluate the effects of human distal arthrogryposis mutations, mybpc1 mRNAs containing the corresponding human W236R and Y856H MYBPC1 mutations were injected into embryos. Dominant-negative effects of these mutations were suggested by the resultant mild bent body curvature, decreased motor activity, as well as impaired overall survival compared with overexpression of wild-type RNA. These results demonstrate a critical role for mybpc1 in slow skeletal muscle development and establish zebrafish as a tractable model of human distal arthrogryposis.


Asunto(s)
Artrogriposis/genética , Artrogriposis/metabolismo , Proteínas Portadoras/genética , Músculo Esquelético/metabolismo , Mutación , Pez Cebra/genética , Pez Cebra/metabolismo , Animales , Tipificación del Cuerpo/genética , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Corazón/embriología , Actividad Motora/genética , Desarrollo de Músculos/genética , Fibras Musculares de Contracción Lenta/metabolismo , Transporte de Proteínas , Sarcómeros/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...