Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.666
Filtrar
1.
Physiol Rep ; 12(19): e70036, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39362825

RESUMEN

The age-related loss of muscle mass is partly accounted for by the loss of sarcomeres in series, contributing to declines in muscle mechanical performance. Resistance training biased to eccentric contractions increases serial sarcomere number (SSN) in young muscle, however, maximal eccentric training in old rats previously did not alter SSN and worsened performance. A submaximal eccentric training stimulus may be more conducive to adaptation for aged muscle. The purpose of this study was to assess whether submaximal eccentric training can increase SSN and improve mechanical function in old rats. Twelve 32-month-old male F344/BN rats completed 4 weeks of submaximal (60% maximum) eccentric plantar-flexion training 3 days/week. Pre- and post-training, we assessed in-vivo maximum isometric torque at a stretched and neutral ankle angle, the passive torque-angle relationship, and the isotonic torque-velocity-power relationship. The soleus and medial gastrocnemius (MG) were harvested for SSN measurements via laser diffraction, with the untrained leg as a control. SSN increased 11% and 8% in the soleus and MG, respectively. Training also shifted optimal torque production towards longer muscle lengths, reduced passive torque 42%, and increased peak isotonic power 23%. Submaximal eccentric training was beneficial for aged muscle adaptations, increasing SSN, reducing muscle passive tension, and improving dynamic contractile performance.


Asunto(s)
Músculo Esquelético , Condicionamiento Físico Animal , Ratas Endogámicas F344 , Entrenamiento de Fuerza , Sarcómeros , Animales , Masculino , Músculo Esquelético/fisiología , Sarcómeros/fisiología , Ratas , Condicionamiento Físico Animal/fisiología , Condicionamiento Físico Animal/métodos , Entrenamiento de Fuerza/métodos , Envejecimiento/fisiología , Torque , Contracción Muscular/fisiología , Ratas Endogámicas BN , Contracción Isométrica/fisiología , Fuerza Muscular/fisiología
2.
J Gen Physiol ; 156(12)2024 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-39373654

RESUMEN

Myosin-binding protein H (MyBP-H) is a component of the vertebrate skeletal muscle sarcomere with sequence and domain homology to myosin-binding protein C (MyBP-C). Whereas skeletal muscle isoforms of MyBP-C (fMyBP-C, sMyBP-C) modulate muscle contractility via interactions with actin thin filaments and myosin motors within the muscle sarcomere "C-zone," MyBP-H has no known function. This is in part due to MyBP-H having limited expression in adult fast-twitch muscle and no known involvement in muscle disease. Quantitative proteomics reported here reveal that MyBP-H is highly expressed in prenatal rat fast-twitch muscles and larval zebrafish, suggesting a conserved role in muscle development and prompting studies to define its function. We take advantage of the genetic control of the zebrafish model and a combination of structural, functional, and biophysical techniques to interrogate the role of MyBP-H. Transgenic, FLAG-tagged MyBP-H or fMyBP-C both localize to the C-zones in larval myofibers, whereas genetic depletion of endogenous MyBP-H or fMyBP-C leads to increased accumulation of the other, suggesting competition for C-zone binding sites. Does MyBP-H modulate contractility in the C-zone? Globular domains critical to MyBP-C's modulatory functions are absent from MyBP-H, suggesting that MyBP-H may be functionally silent. However, our results suggest an active role. In vitro motility experiments indicate MyBP-H shares MyBP-C's capacity as a molecular "brake." These results provide new insights and raise questions about the role of the C-zone during muscle development.


Asunto(s)
Proteínas Portadoras , Fibras Musculares de Contracción Rápida , Pez Cebra , Animales , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Rápida/fisiología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Sarcómeros/metabolismo , Contracción Muscular/fisiología , Desarrollo de Músculos/fisiología , Ratas , Citoesqueleto de Actina/metabolismo
3.
J Gen Physiol ; 156(11)2024 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-39302315

RESUMEN

Sarcomere activation in striated muscle requires both thin filament-based and thick filament-based activation mechanisms. Recent studies have shown that myosin heads on the thick filaments undergo OFF to ON structural transitions in response to calcium (Ca2+) in permeabilized porcine myocardium in the presence of a small molecule inhibitor that eliminated active force. The changes in X-ray diffraction signatures of OFF to ON transitions were interpreted as Ca2+ acting to activate the thick filaments. Alternatively, Ca2+ binding to troponin could initiate a Ca2+-dependent crosstalk from the thin filament to the thick filament via interfilament connections such as the myosin binding protein-C. Here, we exchanged native troponin in permeabilized porcine myocardium for troponin containing the cTnC D65A mutation, which disallows the activation of troponin through Ca2+ binding to determine if Ca2+-dependent thick filament activation persists in the absence of thin filament activation. After the exchange protocol, over 95% of the Ca2+-activated force was eliminated. Equatorial intensity ratio increased significantly in both WT and D65A exchanged myocardium with increasing Ca2+ concentration. The degree of helical ordering of the myosin heads decreased by the same amount in WT and D65A myocardium when Ca2+ concentration increased. These results are consistent with a direct effect of Ca2+ in activating the thick filament rather than an indirect effect due to Ca2+-mediated crosstalk between the thick and thin filaments.


Asunto(s)
Calcio , Miocardio , Animales , Calcio/metabolismo , Porcinos , Miocardio/metabolismo , Sarcómeros/metabolismo , Miosinas/metabolismo , Troponina/metabolismo , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología
4.
Elife ; 122024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39254193

RESUMEN

The force developed by actively lengthened muscle depends on different structures across different scales of lengthening. For small perturbations, the active response of muscle is well captured by a linear-time-invariant (LTI) system: a stiff spring in parallel with a light damper. The force response of muscle to longer stretches is better represented by a compliant spring that can fix its end when activated. Experimental work has shown that the stiffness and damping (impedance) of muscle in response to small perturbations is of fundamental importance to motor learning and mechanical stability, while the huge forces developed during long active stretches are critical for simulating and predicting injury. Outside of motor learning and injury, muscle is actively lengthened as a part of nearly all terrestrial locomotion. Despite the functional importance of impedance and active lengthening, no single muscle model has all these mechanical properties. In this work, we present the viscoelastic-crossbridge active-titin (VEXAT) model that can replicate the response of muscle to length changes great and small. To evaluate the VEXAT model, we compare its response to biological muscle by simulating experiments that measure the impedance of muscle, and the forces developed during long active stretches. In addition, we have also compared the responses of the VEXAT model to a popular Hill-type muscle model. The VEXAT model more accurately captures the impedance of biological muscle and its responses to long active stretches than a Hill-type model and can still reproduce the force-velocity and force-length relations of muscle. While the comparison between the VEXAT model and biological muscle is favorable, there are some phenomena that can be improved: the low frequency phase response of the model, and a mechanism to support passive force enhancement.


Asunto(s)
Modelos Biológicos , Músculo Esquelético/fisiología , Fenómenos Biomecánicos , Humanos , Contracción Muscular/fisiología , Animales , Sarcómeros/fisiología , Impedancia Eléctrica
5.
Mol Biol Cell ; 35(11): ar137, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39259762

RESUMEN

Formin HOmology Domain 2-containing (FHOD) proteins are a subfamily of actin-organizing formins important for striated muscle development in many animals. We showed previously that absence of the sole FHOD protein, FHOD-1, from Caenorhabditis elegans results in thin body wall muscles with misshapen dense bodies that serve as sarcomere Z-lines. We demonstrate here that mutations predicted to specifically disrupt actin polymerization by FHOD-1 similarly disrupt muscle development, and that FHOD-1 cooperates with profilin PFN-3 for dense body morphogenesis, and with profilins PFN-2 and PFN-3 to promote body wall muscle growth. We further demonstrate that dense bodies in worms lacking FHOD-1 or PFN-2/PFN-3 are less stable than in wild-type animals, having a higher proportion of dynamic protein, and becoming distorted by prolonged muscle contraction. We also observe accumulation of actin and actin depolymerization factor/cofilin homologue UNC-60B in body wall muscle of these mutants. Such accumulations may indicate targeted disassembly of thin filaments dislodged from unstable dense bodies, possibly accounting for the abnormally slow growth and reduced body wall muscle strength in fhod-1 mutants. Overall, these results implicate FHOD protein-mediated actin assembly in forming stable sarcomere Z-lines, and identify profilin as a new contributor to FHOD activity in striated muscle development.


Asunto(s)
Actinas , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Forminas , Contracción Muscular , Profilinas , Sarcómeros , Animales , Caenorhabditis elegans/metabolismo , Profilinas/metabolismo , Profilinas/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Sarcómeros/metabolismo , Contracción Muscular/fisiología , Forminas/metabolismo , Actinas/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/genética , Mutación/genética , Desarrollo de Músculos/fisiología , Citoesqueleto de Actina/metabolismo , Músculo Estriado/metabolismo , Músculos/metabolismo , Factores Despolimerizantes de la Actina/metabolismo
6.
J Physiol ; 602(19): 4941-4958, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39283968

RESUMEN

Precise regulation of sarcomeric contraction is essential for normal cardiac function. The heart must generate sufficient force to pump blood throughout the body, but either inadequate or excessive force can lead to dysregulation and disease. Myosin regulatory light chain (RLC) is a thick-filament protein that binds to the neck of the myosin heavy chain. Post-translational phosphorylation of RLC (RLC-P) by myosin light chain kinase is known to influence acto-myosin interactions, thereby increasing force production and Ca2+-sensitivity of contraction. Here, we investigated the role of RLC-P on cardiac structure and function as sarcomere length and [Ca2+] were altered. We found that at low, non-activating levels of Ca2+, RLC-P contributed to myosin head disorder, though there were no effects on isometric stress production and viscoelastic stiffness. With increases in sarcomere length and Ca2+-activation, the structural changes due to RLC-P become greater, which translates into greater force production, greater viscoelastic stiffness, slowed myosin detachment rates and altered nucleotide handling. Altogether, these data suggest that RLC-P may alter thick-filament structure by releasing ordered, off-state myosin. These more disordered myosin heads are available to bind actin, which could result in greater force production as Ca2+ levels increase. However, prolonged cross-bridge attachment duration due to slower ADP release could delay relaxation long enough to enable cross-bridge rebinding. Together, this work further elucidates the effects of RLC-P in regulating muscle function, thereby promoting a better understanding of thick-filament regulatory contributions to cardiac function in health and disease. KEY POINTS: Myosin regulatory light chain (RLC) is a thick-filament protein in the cardiac sarcomere that can be phosphorylated (RLC-P), and changes in RLC-P are associated with cardiac dysfunction and disease. This study assesses how RLC-P alters cardiac muscle structure and function at different sarcomere lengths and calcium concentrations. At low, non-activating levels of Ca2+, RLC-P contributed to myofilament disorder, though there were no effects on isometric stress production and viscoelastic stiffness. With increases in sarcomere length and Ca2+-activation, the structural changes due to RLC-P become greater, which translates into greater force production, greater viscoelastic stiffness, slower myosin detachment rate and altered cross-bridge nucleotide handling rates. This work elucidates the role of RLC-P in regulating muscle function and facilitates understanding of thick-filament regulatory protein contributions to cardiac function in health and disease.


Asunto(s)
Calcio , Cadenas Ligeras de Miosina , Sarcómeros , Animales , Sarcómeros/fisiología , Sarcómeros/metabolismo , Fosforilación , Cadenas Ligeras de Miosina/metabolismo , Calcio/metabolismo , Masculino , Contracción Miocárdica/fisiología , Ratas Sprague-Dawley
7.
Proc Natl Acad Sci U S A ; 121(35): e2322077121, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39172779

RESUMEN

2'-deoxy-ATP (dATP) improves cardiac function by increasing the rate of crossbridge cycling and Ca[Formula: see text] transient decay. However, the mechanisms of these effects and how therapeutic responses to dATP are achieved when dATP is only a small fraction of the total ATP pool remain poorly understood. Here, we used a multiscale computational modeling approach to analyze the mechanisms by which dATP improves ventricular function. We integrated atomistic simulations of prepowerstroke myosin and actomyosin association, filament-scale Markov state modeling of sarcomere mechanics, cell-scale analysis of myocyte Ca[Formula: see text] dynamics and contraction, organ-scale modeling of biventricular mechanoenergetics, and systems level modeling of circulatory dynamics. Molecular and Brownian dynamics simulations showed that dATP increases the actomyosin association rate by 1.9 fold. Markov state models predicted that dATP increases the pool of myosin heads available for crossbridge cycling, increasing steady-state force development at low dATP fractions by 1.3 fold due to mechanosensing and nearest-neighbor cooperativity. This was found to be the dominant mechanism by which small amounts of dATP can improve contractile function at myofilament to organ scales. Together with faster myocyte Ca[Formula: see text] handling, this led to improved ventricular contractility, especially in a failing heart model in which dATP increased ejection fraction by 16% and the energy efficiency of cardiac contraction by 1%. This work represents a complete multiscale model analysis of a small molecule myosin modulator from single molecule to organ system biophysics and elucidates how the molecular mechanisms of dATP may improve cardiovascular function in heart failure with reduced ejection fraction.


Asunto(s)
Nucleótidos de Desoxiadenina , Insuficiencia Cardíaca , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Nucleótidos de Desoxiadenina/metabolismo , Animales , Humanos , Función Ventricular , Modelos Cardiovasculares , Contracción Miocárdica/efectos de los fármacos , Miosinas/metabolismo , Sarcómeros/metabolismo , Actomiosina/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Calcio/metabolismo , Cadenas de Markov
8.
Cells ; 13(16)2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39195263

RESUMEN

Synaptopodin 2-like protein (SYNPO2L) is localized in the sarcomere of cardiomyocytes and is involved in heart morphogenesis. However, the molecular function of SYNPO2L in the heart is not fully understood. We investigated the interaction of SYNPO2L with sarcomeric α-actinin and actin filaments in cultured mouse cardiomyocytes. Immunofluorescence studies showed that SYNPO2L colocalized with α-actinin and actin filaments at the Z-discs of the sarcomere. Recombinant SYNPO2La or SYNPO2Lb caused a bundling of the actin filaments in the absence of α-actinin and enhanced the α-actinin-dependent formation of actin bundles. In addition, high-speed atomic force microscopy revealed that SYNPO2La directly bound to α-actinin via its globular ends. The interaction between α-actinin and SYNPO2La fixed the movements of the two proteins on the actin filaments. These results strongly suggest that SYNPO2L cooperates with α-actinin during actin bundle formation to facilitate sarcomere formation and maintenance.


Asunto(s)
Actinina , Proteínas de Microfilamentos , Proteínas Musculares , Miocitos Cardíacos , Unión Proteica , Sarcómeros , Animales , Ratones , Citoesqueleto de Actina/metabolismo , Actinina/metabolismo , Actinas/metabolismo , Proteínas de Microfilamentos/metabolismo , Miocitos Cardíacos/metabolismo , Sarcómeros/metabolismo , Proteínas Musculares/metabolismo
9.
Curr Biol ; 34(18): 4143-4159.e6, 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39163855

RESUMEN

Muscle morphogenesis is a multi-step program, starting with myoblast fusion, followed by myotube-tendon attachment and sarcomere assembly, with subsequent sarcomere maturation, mitochondrial amplification, and specialization. The correct chronological order of these steps requires precise control of the transcriptional regulators and their effectors. How this regulation is achieved during muscle development is not well understood. In a genome-wide RNAi screen in Drosophila, we identified the BTB-zinc-finger protein Tono (CG32121) as a muscle-specific transcriptional regulator. tono mutant flight muscles display severe deficits in mitochondria and sarcomere maturation, resulting in uncontrolled contractile forces causing muscle rupture and degeneration during development. Tono protein is expressed during sarcomere maturation and localizes in distinct condensates in flight muscle nuclei. Interestingly, internal pressure exerted by the maturing sarcomeres deforms the muscle nuclei into elongated shapes and changes the Tono condensates, suggesting that Tono senses the mechanical status of the muscle cells. Indeed, external mechanical pressure on the muscles triggers rapid liquid-liquid phase separation of Tono utilizing its BTB domain. Thus, we propose that Tono senses high mechanical pressure to adapt muscle transcription, specifically at the sarcomere maturation stages. Consistently, tono mutant muscles display specific defects in a transcriptional switch that represses early muscle differentiation genes and boosts late ones. We hypothesize that a similar mechano-responsive regulation mechanism may control the activity of related BTB-zinc-finger proteins that, if mutated, can result in uncontrolled force production in human muscle.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Desarrollo de Músculos , Animales , Desarrollo de Músculos/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Sarcómeros/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Regulación del Desarrollo de la Expresión Génica
10.
Nat Cardiovasc Res ; 3(8): 907-914, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39196036

RESUMEN

Over half of patients with heart failure have a preserved ejection fraction (>50%, called HFpEF), a syndrome with substantial morbidity/mortality and few effective therapies1. Its dominant comorbidity is now obesity, which worsens disease and prognosis1-3. Myocardial data from patients with morbid obesity and HFpEF show depressed myocyte calcium-stimulated tension4 and disrupted gene expression of mitochondrial and lipid metabolic pathways5,6, abnormalities shared by human HF with a reduced EF but less so in HFpEF without severe obesity. The impact of severe obesity on human HFpEF myocardial ultrastructure remains unexplored. Here we assessed the myocardial ultrastructure in septal biopsies from patients with HFpEF using transmission electron microscopy. We observed sarcomere disruption and sarcolysis, mitochondrial swelling with cristae separation and dissolution and lipid droplet accumulation that was more prominent in the most obese patients with HFpEF and not dependent on comorbid diabetes. Myocardial proteomics revealed associated reduction in fatty acid uptake, processing and oxidation and mitochondrial respiration proteins, particularly in very obese patients with HFpEF.


Asunto(s)
Insuficiencia Cardíaca , Mitocondrias Cardíacas , Miocardio , Volumen Sistólico , Humanos , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/metabolismo , Masculino , Femenino , Anciano , Persona de Mediana Edad , Miocardio/patología , Miocardio/metabolismo , Miocardio/ultraestructura , Mitocondrias Cardíacas/ultraestructura , Mitocondrias Cardíacas/patología , Mitocondrias Cardíacas/metabolismo , Microscopía Electrónica de Transmisión , Función Ventricular Izquierda/fisiología , Sarcómeros/ultraestructura , Sarcómeros/metabolismo , Sarcómeros/patología , Biopsia , Proteómica , Obesidad/patología , Obesidad/metabolismo , Gotas Lipídicas/metabolismo , Comorbilidad
11.
Nat Cardiovasc Res ; 3(8): 1003-1016, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39196032

RESUMEN

Hypertrophic cardiomyopathy (HCM) is an inherited disease of the sarcomere resulting in excessive cardiac contractility. The first-in-class cardiac myosin inhibitor, mavacamten, improves symptoms in obstructive HCM. Here we present aficamten, a selective small-molecule inhibitor of cardiac myosin that diminishes ATPase activity by strongly slowing phosphate release, stabilizing a weak actin-binding state. Binding to an allosteric site on the myosin catalytic domain distinct from mavacamten, aficamten prevents the conformational changes necessary to enter the strongly actin-bound force-generating state. In doing so, aficamten reduces the number of functional myosin heads driving sarcomere shortening. The crystal structure of aficamten bound to cardiac myosin in the pre-powerstroke state provides a basis for understanding its selectivity over smooth and fast skeletal muscle. Furthermore, in cardiac myocytes and in mice bearing the hypertrophic R403Q cardiac myosin mutation, aficamten reduces cardiac contractility. Our findings suggest aficamten holds promise as a therapy for HCM.


Asunto(s)
Miosinas Cardíacas , Cardiomiopatía Hipertrófica , Contracción Miocárdica , Animales , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/metabolismo , Humanos , Contracción Miocárdica/efectos de los fármacos , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/genética , Modelos Animales de Enfermedad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratones , Cristalografía por Rayos X , Mutación , Sarcómeros/metabolismo , Sarcómeros/efectos de los fármacos , Actinas/metabolismo , Modelos Moleculares , Ratones Transgénicos , Conformación Proteica
12.
Skelet Muscle ; 14(1): 18, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095894

RESUMEN

BACKGROUND: Older adults exhibit a slower recovery of muscle mass following disuse atrophy than young adults. At a smaller scale, muscle fibre cross-sectional area (i.e., sarcomeres in parallel) exhibits this same pattern. Less is known, however, about age-related differences in the recovery of muscle fibre length, driven by increases in serial sarcomere number (SSN), following disuse. The purpose of this study was to investigate age-related differences in SSN adaptations and muscle mechanical function during and following muscle immobilization. We hypothesized that older adult rats would experience a similar magnitude of SSN loss during immobilization, however, take longer to recover SSN than young following cast removal, which would limit the recovery of muscle mechanical function. METHODS: We casted the plantar flexors of young (8 months) and old (32 months) male rats in a shortened position for 2 weeks, and assessed recovery during 4 weeks of voluntary ambulation. Following sacrifice, legs were fixed in formalin for measurement of soleus SSN and physiological cross-sectional area (PCSA) with the un-casted soleus acting as a control. Ultrasonographic measurements of pennation angle (PA) and muscle thickness (MT) were conducted weekly. In-vivo active and passive torque-angle relationships were constructed pre-cast, post-cast, and following 4 weeks of recovery. RESULTS: From pre- to post-cast, young and older adult rats experienced similar decreases in SSN (-20%, P < 0.001), muscle wet weight (-25%, P < 0.001), MT (-30%), PA (-15%, P < 0.001), and maximum isometric torque (-40%, P < 0.001), but there was a greater increase in passive torque in older (+ 180%, P < 0.001) compared to young adult rats (+ 68%, P = 0.006). Following cast removal, young exhibited quicker recovery of SSN and MT than old, but SSN recovered sooner than PA and MT in both young and old. PCSA nearly recovered and active torque fully recovered in young adult rats, whereas in older adult rats these remained unrecovered at ∼ 75%. CONCLUSIONS: This study showed that older adult rats retain a better ability to recover longitudinal compared to parallel muscle morphology following cast removal, making SSN a highly adaptable target for improving muscle function in elderly populations early on during rehabilitation.


Asunto(s)
Envejecimiento , Músculo Esquelético , Sarcómeros , Animales , Masculino , Sarcómeros/metabolismo , Sarcómeros/patología , Músculo Esquelético/fisiopatología , Músculo Esquelético/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/diagnóstico por imagen , Ratas , Ratas Endogámicas F344 , Trastornos Musculares Atróficos/fisiopatología , Trastornos Musculares Atróficos/patología , Trastornos Musculares Atróficos/diagnóstico por imagen , Trastornos Musculares Atróficos/etiología , Recuperación de la Función , Suspensión Trasera/efectos adversos , Adaptación Fisiológica
13.
PLoS Comput Biol ; 20(8): e1012321, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39102392

RESUMEN

Understanding muscle contraction mechanisms is a standing challenge, and one of the approaches has been to create models of the sarcomere-the basic contractile unit of striated muscle. While these models have been successful in elucidating many aspects of muscle contraction, they fall short in explaining the energetics of functional phenomena, such as rigor, and in particular, their dependence on the concentrations of the biomolecules involved in the cross-bridge cycle. Our hypothesis posits that the stochastic time delay between ATP adsorption and ADP/Pi release in the cross-bridge cycle necessitates a modeling approach where the rates of these two reaction steps are controlled by two independent parts of the total free energy change of the hydrolysis reaction. To test this hypothesis, we built a two-filament, stochastic-mechanical half-sarcomere model that separates the energetic roles of ATP and ADP/Pi in the cross-bridge cycle's free energy landscape. Our results clearly demonstrate that there is a nontrivial dependence of the cross-bridge cycle's kinetics on the independent concentrations of ATP, ADP, and Pi. The simplicity of the proposed model allows for analytical solutions of the more basic systems, which provide novel insight into the dominant mechanisms driving some of the experimentally observed contractile phenomena.


Asunto(s)
Adenosina Difosfato , Adenosina Trifosfato , Modelos Biológicos , Sarcómeros , Adenosina Difosfato/metabolismo , Sarcómeros/fisiología , Sarcómeros/metabolismo , Adenosina Trifosfato/metabolismo , Cinética , Contracción Muscular/fisiología , Biología Computacional , Animales
14.
Theranostics ; 14(11): 4462-4480, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39113806

RESUMEN

Rationale: Cardiomyocytes (CMs) undergo dramatic structural and functional changes in postnatal maturation; however, the regulatory mechanisms remain greatly unclear. Cypher/Z-band alternatively spliced PDZ-motif protein (ZASP) is an essential sarcomere component maintaining Z-disc stability. Deletion of mouse Cypher and mutation in human ZASP result in dilated cardiomyopathy (DCM). Whether Cypher/ZASP participates in CM maturation and thereby affects cardiac function has not been answered. Methods: Immunofluorescence, transmission electron microscopy, real-time quantitative PCR, and Western blot were utilized to identify the role of Cypher in CM maturation. Subsequently, RNA sequencing and bioinformatics analysis predicted serum response factor (SRF) as the key regulator. Rescue experiments were conducted using adenovirus or adeno-associated viruses encoding SRF, both in vitro and in vivo. The molecular mechanisms were elucidated through G-actin/F-actin fractionation, nuclear-cytoplasmic extraction, actin disassembly assays, and co-sedimentation assays. Results: Cypher deletion led to impaired sarcomere isoform switch and morphological abnormalities in mitochondria, transverse-tubules, and intercalated discs. RNA-sequencing analysis revealed significant dysregulation of crucial genes related to sarcomere assembly, mitochondrial metabolism, and electrophysiology in the absence of Cypher. Furthermore, SRF was predicted as key transcription factor mediating the transcriptional differences. Subsequent rescue experiments showed that SRF re-expression during the critical postnatal period effectively rectified CM maturation defects and notably improved cardiac function in Cypher-depleted mice. Mechanistically, Cypher deficiency resulted in the destabilization of F-actin and a notable increase in G-actin levels, thereby impeding the nuclear localisation of myocardin-related transcription factor A (MRTFA) and subsequently initiating SRF transcription. Conclusion: Cypher/ZASP plays a crucial role in CM maturation through actin-mediated MRTFA-SRF signalling. The linkage between CM maturation abnormalities and the late-onset of DCM is suggested, providing further insights into the pathogenesis of DCM and potential treatment strategies.


Asunto(s)
Actinas , Cardiomiopatía Dilatada , Miocitos Cardíacos , Factor de Respuesta Sérica , Transducción de Señal , Transactivadores , Animales , Miocitos Cardíacos/metabolismo , Factor de Respuesta Sérica/metabolismo , Factor de Respuesta Sérica/genética , Ratones , Actinas/metabolismo , Transactivadores/metabolismo , Transactivadores/genética , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Sarcómeros/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Humanos , Ratones Noqueados
15.
Biophys J ; 123(18): 3024-3037, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-38956875

RESUMEN

Cardiac function relies on the autonomous molecular contraction mechanisms in the ventricular wall. Contraction is driven by ordered motor proteins acting in parallel to generate a macroscopic force. The averaged structure can be investigated by diffraction from model tissues such as trabecular and papillary cardiac muscle using collimated synchrotron beams, offering high resolution in reciprocal space. In the ventricular wall, however, the muscle tissue is compartmentalized into smaller branched cardiomyocytes, with a higher degree of disorder. We show that X-ray diffraction is now also capable of resolving the structural organization of actomyosin in single isolated cardiomyocytes of the ventricular wall. In addition to the hexagonal arrangement of thick and thin filaments, the diffraction signal of the hydrated and fixated cardiomyocytes was sufficient to reveal the myosin motor repeat (M3), the troponin complex repeat (Tn), and the sarcomere length. The sarcomere length signal comprised up to 13 diffraction orders, which were used to compute the sarcomere density profile based on Fourier synthesis. The Tn and M3 spacings were found in the same range as previously reported for other muscle types. The approach opens up a pathway to record the structural dynamics of living cells during the contraction cycle, toward a more complete understanding of cardiac muscle function.


Asunto(s)
Miocitos Cardíacos , Miosinas , Sarcómeros , Troponina , Difracción de Rayos X , Animales , Miocitos Cardíacos/metabolismo , Miosinas/metabolismo , Miosinas/química , Sarcómeros/metabolismo , Análisis de la Célula Individual , Troponina/metabolismo , Troponina/química , Ratones
16.
J Mol Cell Cardiol ; 195: 14-23, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39059462

RESUMEN

Missense mutations in cardiac myosin binding protein C (cMyBP-C) are known to cause hypertrophic cardiomyopathy (HCM). The W792R mutation in the C6 domain of cMyBP-C causes severe, early onset HCM in humans, yet its impact on the function of cMyBP-C and the mechanism through which it causes disease remain unknown. To fully characterize the effect of the W792R mutation on cardiac morphology and function in vivo, we generated a murine knock-in model. We crossed heterozygous W792RWR mice to produce homozygous mutant W792RRR, heterozygous W792RWR, and control W792RWW mice. W792RRR mice present with cardiac hypertrophy, myofibrillar disarray and fibrosis by postnatal day 10 (PND10), and do not survive past PND21. Full-length cMyBP-C is present at similar levels in W792RWW, W792RWR and W792RRR mice and is properly incorporated into the sarcomere. Heterozygous W792RWR mice displayed normal heart morphology and contractility. Permeabilized myocardium from PND10 W792RRR mice showed increased Ca2+ sensitivity, accelerated cross-bridge cycling kinetics, decreased cooperativity in the activation of force, and increased expression of hypertrophy-related genes. In silico modeling suggests that the W792R mutation destabilizes the fold of the C6 domain and increases torsion in the C5-C7 region, possibly impacting regulatory interactions of cMyBP-C with myosin and actin. Based on the data presented here, we propose a model in which mutant W792R cMyBP-C preferentially forms Ca2+ sensitizing interactions with actin, rather than inhibitory interactions with myosin. The W792R-cMyBP-C mouse model provides mechanistic insights into the pathology of this mutation and may provide a mechanism by which other central domain missense mutations in cMyBP-C may alter contractility, leading to HCM.


Asunto(s)
Animales Recién Nacidos , Cardiomiopatía Hipertrófica , Proteínas Portadoras , Mutación Missense , Contracción Miocárdica , Miocardio , Animales , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/fisiopatología , Cardiomiopatía Hipertrófica/patología , Contracción Miocárdica/genética , Ratones , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Miocardio/metabolismo , Dominios Proteicos , Sarcómeros/metabolismo , Calcio/metabolismo , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen
19.
Meat Sci ; 217: 109613, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39084123

RESUMEN

Medium voltage electrical stimulation and wet ageing periods effects on the carcasses and meat quality of Australian rangeland goats were investigated. 64 rangeland goats were slaughtered as 2 equal consignments, wherein half were exposed to post-dressing electrical stimulation (300 V, 500 ms pulse width, 45 s) and half were not (control). Carcass and pH decline parameters and glycogen concentration were recorded. At 24 h post-mortem, fresh colour was measured and longissimus lumborum muscles (LL) removed. Within carcass, paired LL were halved and allocated to each of 4 ageing periods (1, 2, 4, or 21 d). There were no significant effects from electric stimulation on LL pH at 18 °C, LL temperature at pH 6, LL pH at 24 h post-mortem, semitendinosus pH at 24 h post-mortem, and LL glycogen concentration. There were no significant interactions between stimulation and ageing period on cooking loss, particle size, purge loss, sarcomere length, shear force, and ultimate pH. Independent to stimulation, shear force, particle size, and cooking values declined as ageing period increased. Purge loss was highest after 21 d of ageing. Colour stability was unaffected by ageing, although all CIE colour coordinates varied across the 74 h total retail display period. There were negative linear relationships found between LL glycogen concentrations and LL temperature at pH 6, ultimate pH, and sarcomere length. These findings suggest there to be little benefit to post-dressing electrical stimulation of rangeland goat carcasses, when applied independent to or in combination with ageing.


Asunto(s)
Color , Estimulación Eléctrica , Manipulación de Alimentos , Glucógeno , Cabras , Músculo Esquelético , Animales , Concentración de Iones de Hidrógeno , Músculo Esquelético/química , Glucógeno/análisis , Glucógeno/metabolismo , Australia , Manipulación de Alimentos/métodos , Carne Roja/análisis , Culinaria , Masculino , Sarcómeros
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA