Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Vet Pharmacol Ther ; 46(1): 52-61, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36382714

RESUMO

Hypertrophic cardiomyopathy (HCM) is the most prevalent cardiac disease in cats and lacks efficacious preclinical pharmacologic intervention, prompting investigation of novel therapies. Genetic mutations encoding sarcomeric proteins are implicated in the development of HCM and small molecule myosin inhibitors are an emerging class of therapeutics designed to target the interaction of actin and myosin to alleviate the detrimental effects of inappropriate contractile protein interactions. The purpose of this study was to characterize the pharmacodynamic effects of a single oral dose of the novel cardiac myosin inhibitor aficamten (CK-274) on cardiac function in purpose bred cats with naturally occurring A31P MYBPC3 mutation and a clinical diagnosis of HCM with left ventricular outflow tract obstruction (LVOTO). Five purpose bred cats were treated with aficamten (2 mg/kg) or vehicle and echocardiographic evaluations were performed at 0, 6, 24, and 48 h post-dosing. High dose aficamten (2 mg/kg) reduced left ventricular fractional shortening (LVFS%) by increasing the LV systolic internal dimension (LVIDs) and reduced isovolumic relaxation time (IVRT) compared with baseline without significant adverse effects. The marked reduction in systolic function and reduced IVRT coupled with an increased heart rate in treated cats, suggest a lower dose may be optimal. Further studies to determine optimal dosing of aficamten are indicated.


Assuntos
Cardiomiopatia Hipertrófica , Doenças do Gato , Gatos , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Cardiomiopatia Hipertrófica/tratamento farmacológico , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/veterinária , Mutação , Contração Miocárdica , Ecocardiografia/veterinária , Doenças do Gato/tratamento farmacológico
2.
J Mol Cell Cardiol ; 162: 1-9, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34487755

RESUMO

Diabetes doubles the risk of developing heart failure (HF). As the prevalence of diabetes grows, so will HF unless the mechanisms connecting these diseases can be identified. Methylglyoxal (MG) is a glycolysis by-product that forms irreversible modifications on lysine and arginine, called glycation. We previously found that myofilament MG glycation causes sarcomere contractile dysfunction and is increased in patients with diabetes and HF. The aim of this study was to discover the molecular mechanisms by which MG glycation of myofilament proteins cause sarcomere dysfunction and to identify therapeutic avenues to compensate. In humans with type 2 diabetes without HF, we found increased glycation of sarcomeric actin compared to non-diabetics and it correlated with decreased calcium sensitivity. Depressed calcium sensitivity is pathogenic for HF, therefore myofilament glycation represents a promising therapeutic target to inhibit the development of HF in diabetics. To identify possible therapeutic targets, we further defined the molecular actions of myofilament glycation. Skinned myocytes exposed to 100 µM MG exhibited decreased calcium sensitivity, maximal calcium-activated force, and crossbridge kinetics. Replicating MG's functional affects using a computer simulation of sarcomere function predicted simultaneous decreases in tropomyosin's blocked-to-closed rate transition and crossbridge duty cycle were consistent with all experimental findings. Stopped-flow experiments and ATPase activity confirmed MG decreased the blocked-to-closed transition rate. Currently, no therapeutics target tropomyosin, so as proof-of-principal, we used a n-terminal peptide of myosin-binding protein C, previously shown to alter tropomyosin's position on actin. C0C2 completely rescued MG-induced calcium desensitization, suggesting a possible treatment for diabetic HF.


Assuntos
Diabetes Mellitus Tipo 2 , Tropomiosina , Citoesqueleto de Actina/metabolismo , Cálcio/metabolismo , Simulação por Computador , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Miofibrilas/metabolismo , Tropomiosina/metabolismo
3.
Circ Res ; 126(6): 737-749, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32078438

RESUMO

RATIONALE: cMyBP-C (cardiac myosin-binding protein-C) is a critical regulator of heart contraction, but the mechanisms by which cMyBP-C affects actin and myosin are only partly understood. A primary obstacle is that cMyBP-C localization on thick filaments may be a key factor defining its interactions, but most in vitro studies cannot duplicate the unique spatial arrangement of cMyBP-C within the sarcomere. OBJECTIVE: The goal of this study was to validate a novel hybrid genetic/protein engineering approach for rapid manipulation of cMyBP-C in sarcomeres in situ. METHODS AND RESULTS: We designed a novel cut and paste approach for removal and replacement of cMyBP-C N'-terminal domains (C0-C7) in detergent-permeabilized cardiomyocytes from gene-edited Spy-C mice. Spy-C mice express a TEVp (tobacco etch virus protease) cleavage site and a SpyTag (st) between cMyBP-C domains C7 and C8. A cut is achieved using TEVp which cleaves cMyBP-C to create a soluble N'-terminal γC0C7 (endogenous [genetically encoded] N'-terminal domains C0 to C7 of cardiac myosin binding protein-C) fragment and an insoluble C'-terminal SpyTag-C8-C10 fragment that remains associated with thick filaments. Paste of new recombinant (r)C0C7 domains is achieved by a covalent bond formed between SpyCatcher (-sc; encoded at the C'-termini of recombinant proteins) and SpyTag. Results show that loss of γC0C7 reduced myofilament Ca2+ sensitivity and increased cross-bridge cycling (ktr) at submaximal [Ca2+]. Acute loss of γC0C7 also induced auto-oscillatory contractions at submaximal [Ca2+]. Ligation of rC0C7 (exogenous [recombinant] N'-terminal domains C0 to C7 of cardiac myosin binding protein-C)-sc returned pCa50 and ktr to control values and abolished oscillations, but phosphorylated (p)-rC0C7-sc did not completely rescue these effects. CONCLUSIONS: We describe a robust new approach for acute removal and replacement of cMyBP-C in situ. The method revealed a novel role for cMyBP-C N'-terminal domains to damp sarcomere-driven contractile waves (so-called spontaneous oscillatory contractions). Because phosphorylated (p)-rC0C7-sc was less effective at damping contractile oscillations, results suggest that spontaneous oscillatory contractions may contribute to enhanced contractility in response to inotropic stimuli.


Assuntos
Sinalização do Cálcio , Proteínas de Transporte/genética , Edição de Genes/métodos , Contração Miocárdica , Engenharia de Proteínas/métodos , Sarcômeros/metabolismo , Animais , Sistemas CRISPR-Cas , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Endopeptidases/genética , Endopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Domínios Proteicos , Sarcômeros/fisiologia
4.
Proc Natl Acad Sci U S A ; 113(6): 1558-63, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26831109

RESUMO

Mutations in genes encoding myosin, the molecular motor that powers cardiac muscle contraction, and its accessory protein, cardiac myosin binding protein C (cMyBP-C), are the two most common causes of hypertrophic cardiomyopathy (HCM). Recent studies established that the N-terminal domains (NTDs) of cMyBP-C (e.g., C0, C1, M, and C2) can bind to and activate or inhibit the thin filament (TF). However, the molecular mechanism(s) by which NTDs modulate interaction of myosin with the TF remains unknown and the contribution of each individual NTD to TF activation/inhibition is unclear. Here we used an integrated structure-function approach using cryoelectron microscopy, biochemical kinetics, and force measurements to reveal how the first two Ig-like domains of cMyPB-C (C0 and C1) interact with the TF. Results demonstrate that despite being structural homologs, C0 and C1 exhibit different patterns of binding on the surface of F-actin. Importantly, C1 but not C0 binds in a position to activate the TF by shifting tropomyosin (Tm) to the "open" structural state. We further show that C1 directly interacts with Tm and traps Tm in the open position on the surface of F-actin. Both C0 and C1 compete with myosin subfragment 1 for binding to F-actin and effectively inhibit actomyosin interactions when present at high ratios of NTDs to F-actin. Finally, we show that in contracting sarcomeres, the activating effect of C1 is apparent only once low levels of Ca(2+) have been achieved. We suggest that Ca(2+) modulates the interaction of cMyBP-C with the TF in the sarcomere.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Actinas/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Humanos , Hidrólise , Modelos Moleculares , Células Musculares/efeitos dos fármacos , Células Musculares/metabolismo , Contração Muscular/efeitos dos fármacos , Subfragmentos de Miosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Coelhos , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Relação Estrutura-Atividade , Sus scrofa
5.
J Mol Cell Cardiol ; 119: 116-124, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29729251

RESUMO

Cardiac myosin binding protein-C (cMyBP-C) is an essential regulatory protein required for proper systolic contraction and diastolic relaxation. We previously showed that N'-terminal domains of cMyBP-C stimulate contraction by binding to actin and activating the thin filament in vitro. In principle, thin filament activating effects of cMyBP-C could influence contraction and relaxation rates, or augment force amplitude in vivo. cMyBP-C binding to actin could also contribute to an internal load that slows muscle shortening velocity as previously hypothesized. However, the functional significance of cMyBP-C binding to actin has not yet been established in vivo. We previously identified an actin binding site in the regulatory M-domain of cMyBP-C and described two missense mutations that either increased (L348P) or decreased (E330K) binding affinity of recombinant cMyBP-C N'-terminal domains for actin in vitro. Here we created transgenic mice with either the L348P or E330K mutations to determine the functional significance of cMyBP-C binding to actin in vivo. Results showed that enhanced binding of cMyBP-C to actin in L348P-Tg mice prolonged the time to end-systole and slowed relaxation rates. Reduced interactions between cMyBP-C and actin in E330K-Tg mice had the opposite effect and significantly shortened the duration of ejection. Neither mouse model displayed overt systolic dysfunction, but L348P-Tg mice showed diastolic dysfunction presumably resulting from delayed relaxation. We conclude that cMyBP-C binding to actin contributes to sustained thin filament activation at the end of systole and during isovolumetric relaxation. These results provide the first functional evidence that cMyBP-C interactions with actin influence cardiac function in vivo.


Assuntos
Citoesqueleto de Actina/genética , Proteínas de Transporte/genética , Sarcômeros/metabolismo , Sístole/fisiologia , Citoesqueleto de Actina/metabolismo , Actinas/genética , Sequência de Aminoácidos/genética , Animais , Sítios de Ligação , Diástole/genética , Diástole/fisiologia , Feminino , Humanos , Masculino , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação , Mutação Puntual/genética , Ligação Proteica , Domínios Proteicos/genética , Sarcômeros/patologia , Sístole/genética
6.
J Mol Cell Cardiol ; 91: 141-7, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26718724

RESUMO

Mutations in cardiac myosin binding protein C (cMyBP-C), a thick filament protein that modulates contraction of the heart, are a leading cause of hypertrophic cardiomyopathy (HCM). Electron microscopy and 3D reconstruction of thin filaments decorated with cMyBP-C N-terminal fragments suggest that one mechanism of this modulation involves the interaction of cMyBP-C's N-terminal domains with thin filaments to enhance their Ca(2+)-sensitivity by displacement of tropomyosin from its blocked (low Ca(2+)) to its closed (high Ca(2+)) position. The extent of this tropomyosin shift is reduced when cMyBP-C N-terminal domains are phosphorylated. In the current study, we have examined L348P, a sequence variant of cMyBP-C first identified in a screen of patients with HCM. In L348P, leucine 348 is replaced by proline in cMyBP-C's regulatory M-domain, resulting in an increase in cMyBP-C's ability to enhance thin filament Ca(2+)-sensitization. Our goal here was to determine the structural basis for this enhancement by carrying out 3D reconstruction of thin filaments decorated with L348P-mutant cMyBP-C. When thin filaments were decorated with wild type N-terminal domains at low Ca(2+), tropomyosin moved from the blocked to the closed position, as found previously. In contrast, the L348P mutant caused a significantly larger tropomyosin shift, to approximately the open position, consistent with its enhancement of Ca(2+)-sensitization. Phosphorylated wild type fragments showed a smaller shift than unphosphorylated fragments, whereas the shift induced by the L348P mutant was not affected by phosphorylation. We conclude that the L348P mutation causes a gain of function by enhancing tropomyosin displacement on the thin filament in a phosphorylation-independent way.


Assuntos
Actinas/química , Proteínas de Transporte/química , Citoesqueleto/química , Sarcômeros/química , Tropomiosina/química , Actinas/genética , Actinas/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Cálcio/metabolismo , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Bovinos , Galinhas , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Regulação da Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Miocárdio/química , Miocárdio/metabolismo , Fosforilação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Transdução de Sinais , Tropomiosina/genética , Tropomiosina/metabolismo
7.
J Mol Cell Cardiol ; 97: 286-94, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27139341

RESUMO

Thin filament length (TFL) is an important determinant of the force-sarcomere length (SL) relation of cardiac muscle. However, the various mechanisms that control TFL are not well understood. Here we tested the previously proposed hypothesis that the actin-binding protein nebulin contributes to TFL regulation in the heart by using a cardiac-specific nebulin cKO mouse model (αMHC Cre Neb cKO). Atrial myocytes were studied because nebulin expression has been reported to be most prominent in this cell type. TFL was measured in right and left atrial myocytes using deconvolution optical microscopy and staining for filamentous actin with phalloidin and for the thin filament pointed-end with an antibody to the capping protein Tropomodulin-1 (Tmod1). Results showed that TFLs in Neb cKO and littermate control mice were not different. Thus, deletion of nebulin in the heart does not alter TFL. However, TFL was found to be ~0.05µm longer in the right than in the left atrium and Tmod1 expression was increased in the right atrium. We also tested the hypothesis that the length of titin's spring region is a factor controlling TFL by studying the Rbm20(ΔRRM) mouse which expresses titins that are ~500kDa (heterozygous mice) and ~1000kDa (homozygous mice) longer than in control mice. Results revealed that TFL was not different in Rbm20(ΔRRM) mice. An unexpected finding in all genotypes studied was that TFL increased as sarcomeres were stretched (~0.1µm per 0.35µm of SL increase). This apparent increase in TFL reached a maximum at a SL of ~3.0µm where TFL was ~1.05µm. The SL dependence of TFL was independent of chemical fixation or the presence of cardiac myosin-binding protein C (cMyBP-C). In summary, we found that in cardiac myocytes TFL varies with SL in a manner that is independent of the size of titin or the presence of nebulin.


Assuntos
Conectina/metabolismo , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Sarcômeros/fisiologia , Animais , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos , Microscopia , Proteínas Musculares/deficiência , Proteínas Musculares/genética , Miofibrilas , Cadeias Pesadas de Miosina/deficiência , Cadeias Pesadas de Miosina/genética
8.
J Mol Cell Cardiol ; 94: 65-71, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27021517

RESUMO

Cardiac myosin binding protein-C (cMyBP-C) is a structural and regulatory component of cardiac thick filaments. It is observed in electron micrographs as seven to nine transverse stripes in the central portion of each half of the A band. Its C-terminus binds tightly to the myosin rod and contributes to thick filament structure, while the N-terminus can bind both myosin S2 and actin, influencing their structure and function. Mutations in the MYBPC3 gene (encoding cMyBP-C) are commonly associated with hypertrophic cardiomyopathy (HCM). In cardiac cells there exists a population of myosin heads in the super-relaxed (SRX) state, which are bound to the thick filament core with a highly inhibited ATPase activity. This report examines the role cMyBP-C plays in regulating the population of the SRX state of cardiac myosin by using an assay that measures single ATP turnover of myosin. We report a significant decrease in the proportion of myosin heads in the SRX state in homozygous cMyBP-C knockout mice, however heterozygous cMyBP-C knockout mice do not significantly differ from the wild type. A smaller, non-significant decrease is observed when thoracic aortic constriction is used to induce cardiac hypertrophy in mutation negative mice. These results support the proposal that cMyBP-C stabilises the thick filament and that the loss of cMyBP-C results in an untethering of myosin heads. This results in an increased myosin ATP turnover, further consolidating the relationship between thick filament structure and the myosin ATPase.


Assuntos
Miosinas Cardíacas/metabolismo , Proteínas de Transporte/genética , Miócitos Cardíacos/metabolismo , Animais , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/fisiopatologia , Genótipo , Camundongos , Camundongos Knockout , Fosforilação , Sarcômeros/metabolismo
9.
Arch Biochem Biophys ; 601: 133-40, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26777460

RESUMO

Mutations in MYBPC3, the gene encoding cardiac myosin binding protein C (cMyBP-C), are a major cause of hypertrophic cardiomyopathy (HCM). While most mutations encode premature stop codons, missense mutations causing single amino acid substitutions are also common. Here we investigated effects of a single proline for alanine substitution at amino acid 31 (A31P) in the C0 domain of cMyBP-C, which was identified as a natural cause of HCM in cats. Results using recombinant proteins showed that the mutation disrupted C0 structure, altered sensitivity to trypsin digestion, and reduced recognition by an antibody that preferentially recognizes N-terminal domains of cMyBP-C. Western blots detecting A31P cMyBP-C in myocardium of cats heterozygous for the mutation showed a reduced amount of A31P mutant protein relative to wild-type cMyBP-C, but the total amount of cMyBP-C was not different in myocardium from cats with or without the A31P mutation indicating altered rates of synthesis/degradation of A31P cMyBP-C. Also, the mutant A31P cMyBP-C was properly localized in cardiac sarcomeres. These results indicate that reduced protein expression (haploinsufficiency) cannot account for effects of the A31P cMyBP-C mutation and instead suggest that the A31P mutation causes HCM through a poison polypeptide mechanism that disrupts cMyBP-C or myocyte function.


Assuntos
Cardiomiopatia Hipertrófica/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Haploinsuficiência , Mutação de Sentido Incorreto , Alanina/química , Animais , Gatos , Dicroísmo Circular , Códon de Terminação , Coração/fisiopatologia , Imuno-Histoquímica , Células Musculares/citologia , Mutação , Miocárdio/metabolismo , Prolina/química , Conformação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sarcômeros/metabolismo
10.
Biophys J ; 108(1): 10-3, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25564844

RESUMO

Although mutations in cardiac myosin binding protein-C (cMyBP-C) cause heart disease, its role in muscle contraction is not well understood. A mechanism remains elusive partly because the protein can have multiple effects, such as dual biphasic activation and inhibition observed in actin motility assays. Here we develop a mathematical model for the interaction of cMyBP-C with the contractile proteins actin and myosin and the regulatory protein tropomyosin. We use this model to show that a drag-activation-competition mechanism accurately describes actin motility measurements, while models lacking either drag or competition do not. These results suggest that complex effects can arise simply from cMyBP-C binding to actin.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Modelos Moleculares , Miosinas/metabolismo , Tropomiosina/metabolismo , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Movimento (Física)
11.
J Mol Cell Cardiol ; 88: 124-32, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26455481

RESUMO

Cardiac myosin binding protein C (cMyBP-C) is an essential regulator of cross bridge cycling. Through mechanisms that are incompletely understood the N-terminal domains (NTDs) of cMyBP-C can activate contraction even in the absence of calcium and can also inhibit cross bridge kinetics in the presence of calcium. In vitro studies indicated that the proline-alanine rich (p/a) region and C1 domain are involved in these processes, although effects were greater using human proteins compared to murine proteins (Shaffer et al. J Biomed Biotechnol 2010, 2010: 789798). We hypothesized that the p/a and C1 region are critical for the timing of contraction. In this study we tested this hypothesis using a mouse model lacking the p/a and C1 region (p/a-C1(-/-) mice) to investigate the in vivo relevance of these regions on cardiac performance. Surprisingly, hearts of adult p/a-C1(-/-) mice functioned normally both on a cellular and whole organ level. Force measurements in permeabilized cardiomyocytes from adult p/a-C1(-/-) mice and wild type (Wt) littermate controls demonstrated similar rates of force redevelopment both at submaximal and maximal activation. Maximal and passive force and calcium sensitivity of force were comparable between groups as well. Echocardiograms showed normal isovolumetric contraction times, fractional shortening and ejection fraction, indicating proper systolic function in p/a-C1(-/-) mouse hearts. p/a-C1(-/-) mice showed a slight but significant reduction in isovolumetric relaxation time compared to Wt littermates, yet this difference disappeared in older mice (7-8months of age). Moreover, stroke volume was preserved in p/a-C1(-/-) mice, corroborating sufficient time for normal filling of the heart. Overall, the hearts of p/a-C1(-/-) mice showed no signs of dysfunction even after chronic stress with an adrenergic agonist. Together, these results indicate that the p/a region and the C1 domain of cMyBP-C are not critical for normal cardiac contraction in mice and that these domains have little if any impact on cross bridge kinetics in mice. These results thus contrast with in vitro studies utilizing proteins encoding the human p/a region and C1 domain. More detailed insight in how individual domains of cMyBP-C function and interact, across species and over the wide spectrum of conditions in which the heart has to function, will be essential to a better understanding of how cMyBP-C tunes cardiac contraction.


Assuntos
Proteínas de Transporte/metabolismo , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Volume Sistólico/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Alanina/genética , Alanina/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Proteínas de Transporte/genética , Ecocardiografia , Expressão Gênica , Humanos , Isoproterenol/farmacologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Miocárdio/ultraestrutura , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Prolina/genética , Prolina/metabolismo , Estrutura Terciária de Proteína , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Deleção de Sequência , Sístole/fisiologia
12.
Surg Radiol Anat ; 37(3): 267-72, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25189812

RESUMO

PURPOSE: An understanding of root anatomy is an important foundation for providing successful endodontic treatment. The aim of this study was to use micro-computed tomography (micro-CT) to investigate the root anatomy of the mandibular second molar. METHODS: Eighteen mandibular second molars were scanned using micro-CT. Images were reconstructed, and measurements and observations were recorded regarding pulpal floor anatomy, canal configuration, root wall thickness along the root, presence of calcifications in the pulp chamber and in canals, and apical anatomy. RESULTS/CONCLUSIONS: The most frequently found mesial root canal configuration was Vertucci Type 7 (1-2-1-2), which was seen in 33.3% of samples. Distal canals were most frequently Vertucci Type 1 (one canal), with 61.1% of samples showing this configuration. 11.1% of samples had two canals, 44.4% of samples had three canals, 33.3% of samples had four canals, and 11.1% of samples had five canals at some point along the length of the roots. Average root wall thickness between the mesiobuccal canal and the furcation was 1.23 mm. Mesiolingual canal root wall thickness was on average 1.29 mm, and the distal root furcation wall thickness averaged 1.41 mm. 77.8% of samples had calcifications present in both the pulp chamber and within the canals.


Assuntos
Cavidade Pulpar/anatomia & histologia , Mandíbula/anatomia & histologia , Dente Molar/anatomia & histologia , Dente Molar/diagnóstico por imagem , Microtomografia por Raio-X/métodos , Cavidade Pulpar/diagnóstico por imagem , Humanos , Mandíbula/diagnóstico por imagem , Estudos de Amostragem , Raiz Dentária/anatomia & histologia , Raiz Dentária/diagnóstico por imagem
13.
Biochemistry ; 53(42): 6717-24, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25265574

RESUMO

We have used enzyme kinetics to investigate the molecular mechanism by which the N-terminal domains of human and mouse cardiac MyBP-C (C0C1, C1C2, and C0C2) affect the activation of myosin ATP hydrolysis by F-actin and by native porcine thin filaments. N-Terminal domains of cMyBP-C inhibit the activation of myosin-S1 ATPase by F-actin. However, mouse and human C1C2 and C0C2 produce biphasic activating and inhibitory effects on the activation of myosin ATP hydrolysis by native cardiac thin filaments. Low ratios of MyBP-C N-terminal domains to thin filaments activate myosin-S1 ATP hydrolysis, but higher ratios inhibit ATP hydrolysis, as is observed with F-actin alone. These data suggest that low concentrations of C1C2 and C0C2 activate thin filaments by a mechanism similar to that of rigor myosin-S1, whereas higher concentrations inhibit the ATPase rate by competing with myosin-S1-ADP-Pi for binding to actin and thin filaments. In contrast to C0C2 and C1C2, the activating effects of the C0C1 domain are species-dependent: human C0C1 activates actomyosin-S1 ATPase rates, but mouse C0C1 does not produce significant activation or inhibition. Phosphorylation of serine residues in the m-linker between the C1 and C2 domains by protein kinase-A decreases the activation of thin filaments by huC0C2 at pCa > 8 but has little effect on the activation mechanism at pCa = 4. In sarcomeres, the low ratio of cMyBP-C to actin is expected to favor the activating effects of cMyBP-C while minimizing inhibition produced by competition with myosin heads.


Assuntos
Citoesqueleto de Actina/química , Trifosfato de Adenosina/química , Proteínas de Transporte/química , Miocárdio/química , Miosinas/química , Actinas/química , Animais , Cálcio/química , Miosinas Cardíacas/química , Humanos , Hidrólise , Cinética , Camundongos , Fosforilação , Estrutura Terciária de Proteína , Coelhos , Proteínas Recombinantes/química , Especificidade da Espécie , Suínos
14.
J Biol Chem ; 288(30): 21496-505, 2013 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-23782699

RESUMO

The M-domain is the major regulatory subunit of cardiac myosin-binding protein-C (cMyBP-C) that modulates actin and myosin interactions to influence muscle contraction. However, the precise mechanism(s) and the specific residues involved in mediating the functional effects of the M-domain are not fully understood. Positively charged residues adjacent to phosphorylation sites in the M-domain are thought to be critical for effects of cMyBP-C on cross-bridge interactions by mediating electrostatic binding with myosin S2 and/or actin. However, recent structural studies revealed that highly conserved sequences downstream of the phosphorylation sites form a compact tri-helix bundle. Here we used site-directed mutagenesis to probe the functional significance of charged residues adjacent to the phosphorylation sites and conserved residues within the tri-helix bundle. Results confirm that charged residues adjacent to phosphorylation sites and residues within the tri-helix bundle are important for mediating effects of the M-domain on contraction. In addition, four missense variants within the tri-helix bundle that are associated with human hypertrophic cardiomyopathy caused either loss-of-function or gain-of-function effects on force. Importantly, the effects of the gain-of-function variant, L348P, increased the affinity of the M-domain for actin. Together, results demonstrate that functional effects of the M-domain are not due solely to interactions with charged residues near phosphorylatable serines and provide the first demonstration that the tri-helix bundle contributes to the functional effects of the M-domain, most likely by binding to actin.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Mutação , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Cálcio/metabolismo , Cálcio/farmacologia , Cardiomiopatia Hipertrófica/genética , Proteínas de Transporte/química , Humanos , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Contração Muscular/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Fosforilação , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
16.
Pflugers Arch ; 466(3): 445-50, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24442149

RESUMO

Myosin binding protein-C (MyBP-C) was first discovered as an impurity during the purification of myosin from skeletal muscle. However, soon after its discovery, MyBP-C was also shown to bind actin. While the unique functional implications for a protein that could cross-link thick and thin filaments together were immediately recognized, most early research nonetheless focused on interactions of MyBP-C with the thick filament. This was in part because interactions of MyBP-C with the thick filament could adequately explain most (but not all) effects of MyBP-C on actomyosin interactions and in part because the specificity of actin binding was uncertain. However, numerous recent studies have now established that MyBP-C can indeed bind to actin through multiple binding sites, some of which are highly specific. Many of these interactions involve critical regulatory domains of MyBP-C that are also reported to interact with myosin. Here we review current evidence supporting MyBP-C interactions with actin and discuss these findings in terms of their ability to account for the functional effects of MyBP-C. We conclude that the influence of MyBP-C on muscle contraction can be explained equally well by interactions with actin as by interactions with myosin. However, because data showing that MyBP-C binds to either myosin or actin has come almost exclusively from in vitro biochemical studies, the challenge for future studies is to define which binding partner(s) MyBP-C interacts with in vivo.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Sarcômeros/metabolismo , Animais , Sítios de Ligação , Proteínas de Transporte/química , Humanos , Ligação Proteica , Sarcômeros/ultraestrutura
17.
Arch Biochem Biophys ; 550-551: 28-32, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24736382

RESUMO

The two genes most commonly associated with mutations linked to hypertrophic or dilated cardiomyopathies are ß-myosin and cardiac myosin binding protein-C (cMyBP-C). Both of these proteins interact with cardiac actin (ACTC). Currently there are 16 ACTC variants that have been found in patients with HCM or DCM. While some of these ACTC variants exhibit protein instability or polymerization-deficiencies that might contribute to the development of disease, other changes could cause changes in protein-protein interactions between sarcomere proteins and ACTC. To test the hypothesis that changes in ACTC disrupt interactions with cMyBP-C, we examined the interactions between seven ACTC variants and the N-terminal C0C2 fragment of cMyBP-C. We found there was a significant decrease in binding affinity (increase in Kd values) for the A331P and Y166C variants of ACTC. These results suggest that a change in the ability of cMyBP-C to bind actin filaments containing these ACTC protein variants might contribute to the development of disease. These results also provide clues regarding the binding site of the C0C2 fragment of cMyBP-C on F-actin.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas de Transporte/metabolismo , Miosinas Ventriculares/metabolismo , Citoesqueleto de Actina/patologia , Actinas/genética , Sítios de Ligação , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/patologia , Proteínas de Transporte/genética , Expressão Gênica , Humanos , Cinética , Miocárdio/metabolismo , Miocárdio/patologia , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sarcômeros/metabolismo , Sarcômeros/patologia , Miosinas Ventriculares/genética
18.
Nat Commun ; 15(1): 2628, 2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38521794

RESUMO

Muscle contraction is produced via the interaction of myofilaments and is regulated so that muscle performance matches demand. Myosin-binding protein C (MyBP-C) is a long and flexible protein that is tightly bound to the thick filament at its C-terminal end (MyBP-CC8C10), but may be loosely bound at its middle- and N-terminal end (MyBP-CC1C7) to myosin heads and/or the thin filament. MyBP-C is thought to control muscle contraction via the regulation of myosin motors, as mutations lead to debilitating disease. We use a combination of mechanics and small-angle X-ray diffraction to study the immediate and selective removal of the MyBP-CC1C7 domains of fast MyBP-C in permeabilized skeletal muscle. We show that cleavage leads to alterations in crossbridge kinetics and passive structural signatures of myofilaments that are indicative of a shift of myosin heads towards the ON state, highlighting the importance of MyBP-CC1C7 to myofilament force production and regulation.


Assuntos
Proteínas de Transporte , Sarcômeros , Sarcômeros/metabolismo , Proteínas de Transporte/metabolismo , Contração Muscular/fisiologia , Músculo Esquelético/metabolismo , Miosinas/metabolismo
19.
Sci Rep ; 14(1): 12038, 2024 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-38802475

RESUMO

Hypertrophic cardiomyopathy (HCM) remains the most common cardiomyopathy in humans and cats with few preclinical pharmacologic interventional studies. Small-molecule sarcomere inhibitors are promising novel therapeutics for the management of obstructive HCM (oHCM) patients and have shown efficacy in left ventricular outflow tract obstruction (LVOTO) relief. The objective of this study was to explore the 6-, 24-, and 48-hour (h) pharmacodynamic effects of the cardiac myosin inhibitor, CK-586, in six purpose-bred cats with naturally occurring oHCM. A blinded, randomized, five-treatment group, crossover preclinical trial was conducted to assess the pharmacodynamic effects of CK-586 in this oHCM model. Dose assessments and select echocardiographic variables were assessed five times over a 48-h period. Treatment with oral CK-586 safely ameliorated LVOTO in oHCM cats. CK-586 treatment dose-dependently eliminated obstruction (reduced LVOTOmaxPG), increased measures of systolic chamber size (LVIDs Sx), and decreased select measures of heart function (LV FS% and LV EF%) in the absence of impact on heart rate. At all tested doses, a single oral CK-586 dose resulted in improved or resolved LVOTO with well-tolerated, dose-dependent, reductions in LV systolic function. The results from this study pave the way for the potential use of CK-586 in both the veterinary and human clinical setting.


Assuntos
Miosinas Cardíacas , Cardiomiopatia Hipertrófica , Animais , Gatos , Cardiomiopatia Hipertrófica/tratamento farmacológico , Miosinas Cardíacas/metabolismo , Doenças do Gato/tratamento farmacológico , Masculino , Feminino , Obstrução do Fluxo Ventricular Externo/tratamento farmacológico , Sístole/efeitos dos fármacos , Ecocardiografia , Estudos Cross-Over
20.
Biophys J ; 104(11): 2465-75, 2013 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-23746519

RESUMO

Cardiac myosin binding protein-C (cMyBP-C) is a member of the immunoglobulin (Ig) superfamily of proteins and consists of 8 Ig- and 3 fibronectin III (FNIII)-like domains along with a unique regulatory sequence referred to as the MyBP-C motif or M-domain. We previously used atomic force microscopy to investigate the mechanical properties of murine cMyBP-C expressed using a baculovirus/insect cell expression system. Here, we investigate whether the mechanical properties of cMyBP-C are conserved across species by using atomic force microscopy to manipulate recombinant human cMyBP-C and native cMyBP-C purified from bovine heart. Force versus extension data obtained in velocity-clamp experiments showed that the mechanical response of the human recombinant protein was remarkably similar to that of the bovine native cMyBP-C. Ig/Fn-like domain unfolding events occurred in a hierarchical fashion across a threefold range of forces starting at relatively low forces of ~50 pN and ending with the unfolding of the highest stability domains at ~180 pN. Force-extension traces were also frequently marked by the appearance of anomalous force drops suggestive of additional mechanical complexity such as structural coupling among domains. Both recombinant and native cMyBP-C exhibited a prominent segment ~100 nm-long that could be stretched by forces <50 pN before the unfolding of Ig- and FN-like domains. Combined with our previous observations of mouse cMyBP-C, these results establish that although the response of cMyBP-C to mechanical load displays a complex pattern, it is highly conserved across species.


Assuntos
Proteínas de Transporte/metabolismo , Fenômenos Mecânicos , Animais , Proteínas de Transporte/química , Bovinos , Humanos , Camundongos , Modelos Moleculares , Método de Monte Carlo , Estrutura Terciária de Proteína , Desdobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA