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











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 323(6): C1728-C1739, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36280392

RESUMO

Stretch activation is defined as a delayed increase in force after rapid stretches. Although there is considerable evidence for stretch activation in isolated cardiac myofibrillar preparations, few studies have measured mechanisms of stretch activation in mammalian skeletal muscle fibers. We measured stretch activation following rapid step stretches [∼1%-4% sarcomere length (SL)] during submaximal Ca2+ activations of rat permeabilized slow-twitch skeletal muscle fibers before and after protein kinase A (PKA), which phosphorylates slow myosin binding protein-C. PKA significantly increased stretch activation during low (∼25%) Ca2+ activation and accelerated rates of delayed force development (kef) during both low and half-maximal Ca2+ activation. Following the step stretches and subsequent force development, fibers were rapidly shortened to original sarcomere length, which often elicited a shortening-induced transient force overshoot. After PKA, step shortening-induced transient force overshoot increased ∼10-fold following an ∼4% SL shortening during low Ca2+ activation levels. kdf following step shortening also increased after PKA during low and half-maximal Ca2+ activations. We next investigated thin filament regulation of stretch activation. We tested the interplay between cardiac troponin I (cTnI) phosphorylation at the canonical PKA and novel tyrosine kinase sites on stretch activation. Native slow-skeletal Tn complexes were exchanged with recombinant human cTn complex with different human cTnI N-terminal pseudo-phosphorylation molecules: 1) nonphosphorylated wild type (WT), 2) the canonical S22/23D PKA sites, 3) the tyrosine kinase Y26E site, and 4) the combinatorial S22/23D + Y26E cTnI. All three pseudo-phosphorylated cTnIs elicited greater stretch activation than WT. Following stretch activation, a new, elevated stretch-induced steady-state force was reached with pseudo-phosphorylated cTnI. Combinatorial S22/23D + Y26E pseudo-phosphorylated cTnI increased kdf. These results suggest that slow-skeletal myosin binding protein-C (sMyBP-C) phosphorylation modulates stretch activation by a combination of cross-bridge recruitment and faster cycling kinetics, whereas cTnI phosphorylation regulates stretch activation by both redundant and synergistic mechanisms; and, taken together, these sarcomere phosphoproteins offer precision targets for enhanced contractility.


Assuntos
Cálcio , Miofibrilas , Ratos , Humanos , Animais , Miofibrilas/metabolismo , Cálcio/metabolismo , Sarcômeros/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Troponina I/química , Fosforilação , Miosinas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Miocárdio/metabolismo , Contração Miocárdica/fisiologia , Mamíferos/metabolismo
2.
Elife ; 112022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35502901

RESUMO

Phosphorylation and acetylation of sarcomeric proteins are important for fine-tuning myocardial contractility. Here, we used bottom-up proteomics and label-free quantification to identify novel post-translational modifications (PTMs) on ß-myosin heavy chain (ß-MHC) in normal and failing human heart tissues. We report six acetylated lysines and two phosphorylated residues: K34-Ac, K58-Ac, S210-P, K213-Ac, T215-P, K429-Ac, K951-Ac, and K1195-Ac. K951-Ac was significantly reduced in both ischemic and nonischemic failing hearts compared to nondiseased hearts. Molecular dynamics (MD) simulations show that K951-Ac may impact stability of thick filament tail interactions and ultimately myosin head positioning. K58-Ac altered the solvent-exposed SH3 domain surface - known for protein-protein interactions - but did not appreciably change motor domain conformation or dynamics under conditions studied. Together, K213-Ac/T215-P altered loop 1's structure and dynamics - known to regulate ADP-release, ATPase activity, and sliding velocity. Our study suggests that ß-MHC acetylation levels may be influenced more by the PTM location than the type of heart disease since less protected acetylation sites are reduced in both heart failure groups. Additionally, these PTMs have potential to modulate interactions between ß-MHC and other regulatory sarcomeric proteins, ADP-release rate of myosin, flexibility of the S2 region, and cardiac myofilament contractility in normal and failing hearts.


Assuntos
Cadeias Pesadas de Miosina , Sarcômeros , Difosfato de Adenosina/metabolismo , Humanos , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosinas/metabolismo , Processamento de Proteína Pós-Traducional , Sarcômeros/metabolismo , Fatores de Transcrição/metabolismo
3.
J Mol Cell Cardiol ; 139: 135-147, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31981571

RESUMO

OBJECTIVE: Cardiac troponin I (cTnI) is an essential physiological and pathological regulator of cardiac relaxation. Significant to this regulation, the post-translational modification of cTnI through phosphorylation functions as a key mechanism to accelerate myofibril relaxation. Similar to phosphorylation, post-translational modification by acetylation alters amino acid charge and protein function. Recent studies have demonstrated that the acetylation of cardiac myofibril proteins accelerates relaxation and that cTnI is acetylated in the heart. These findings highlight the potential significance of myofilament acetylation; however, it is not known if site-specific acetylation of cTnI can lead to changes in myofilament, myofibril, and/or cellular mechanics. The objective of this study was to determine the effects of mimicking acetylation at a single site of cTnI (lysine-132; K132) on myofilament, myofibril, and cellular mechanics and elucidate its influence on molecular function. METHODS: To determine if pseudo-acetylation of cTnI at 132 modulates thin filament regulation of the acto-myosin interaction, we reconstituted thin filaments containing WT or K132Q (to mimic acetylation) cTnI and assessed in vitro motility. To test if mimicking acetylation at K132 alters cellular relaxation, adult rat ventricular cardiomyocytes were infected with adenoviral constructs expressing either cTnI K132Q or K132 replaced with arginine (K132R; to prevent acetylation) and cell shortening and isolated myofibril mechanics were measured. Finally, to confirm that changes in cell shortening and myofibril mechanics were directly due to pseudo-acetylation of cTnI at K132, we exchanged troponin containing WT or K132Q cTnI into isolated myofibrils and measured myofibril mechanical properties. RESULTS: Reconstituted thin filaments containing K132Q cTnI exhibited decreased calcium sensitivity compared to thin filaments reconstituted with WT cTnI. Cardiomyocytes expressing K132Q cTnI had faster relengthening and myofibrils isolated from these cells had faster relaxation along with decreased calcium sensitivity compared to cardiomyocytes expressing WT or K132R cTnI. Myofibrils exchanged with K132Q cTnI ex vivo demonstrated faster relaxation and decreased calcium sensitivity. CONCLUSIONS: Our results indicate for the first time that mimicking acetylation of a specific cTnI lysine accelerates myofilament, myofibril, and myocyte relaxation. This work underscores the importance of understanding how acetylation of specific sarcomeric proteins affects cardiac homeostasis and disease and suggests that modulation of myofilament lysine acetylation may represent a novel therapeutic target to alter cardiac relaxation.


Assuntos
Cálcio/metabolismo , Miocárdio/metabolismo , Miofibrilas/metabolismo , Troponina I/metabolismo , Acetilação , Animais , Feminino , Ventrículos do Coração/citologia , Lisina/metabolismo , Miócitos Cardíacos/metabolismo , Ratos Endogâmicos Dahl , Ratos Sprague-Dawley
4.
Life Sci ; 215: 119-127, 2018 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-30399377

RESUMO

AIMS: Heart failure (HF) is a prevalent disease that is considered the foremost reason for hospitalization in the United States. Most protein kinases (PK) are activated in heart disease and their inhibition has been shown to improve cardiac function in both animal and human studies. However, little is known about the direct impact of PKA and PKC inhibitors on human cardiac contractile function. MATERIAL AND METHODS: We investigated the ex vivo effect of such inhibitors on force as well as on kinetics of left ventricular (LV) trabeculae dissected from non-failing and failing human hearts. In these experiments, we applied 0.5 µM of H-89 and GF109203X, which are PKA and PKC inhibitors, respectively, in comparison to their vehicle DMSO (0.05%). KEY FINDINGS AND CONCLUSION: Statistical analyses revealed no significant effect for H-89 and GF109203X on either contractile force or kinetics parameters of both non-failing and failing muscles even though they were used at a concentration higher than the reported IC50s and Kis. Therefore, several factors such as selectivity, concentration, and treatment time, which are related to these PK inhibitors according to previous studies require further exploration.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Insuficiência Cardíaca/tratamento farmacológico , Miocárdio/patologia , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Adulto , Idoso , Feminino , Coração/efeitos dos fármacos , Coração/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/metabolismo , Humanos , Indóis/administração & dosagem , Indóis/farmacologia , Concentração Inibidora 50 , Isoquinolinas/administração & dosagem , Isoquinolinas/farmacologia , Masculino , Maleimidas/administração & dosagem , Maleimidas/farmacologia , Pessoa de Meia-Idade , Contração Miocárdica/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Adulto Jovem
5.
J Biol Chem ; 293(22): 8588-8599, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669813

RESUMO

The acceleration of myocardial relaxation produced by ß-adrenoreceptor stimulation is mediated in part by protein kinase A (PKA)-mediated phosphorylation of cardiac troponin-I (cTnI), which decreases myofibrillar Ca2+ sensitivity. Previous evidence suggests that phosphorylation of both Ser-23 and Ser-24 in cTnI is required for this Ca2+ desensitization. PKA-mediated phosphorylation also partially protects cTnI from proteolysis by calpain. Here we report that protein kinase D (PKD) phosphorylates only one serine of cTnI Ser-23/24. To explore the functional consequences of this monophosphorylation, we examined the Ca2+ sensitivity of force production and susceptibility of cTnI to calpain-mediated proteolysis when Ser-23/24 of cTnI in mouse cardiac myofibrils was nonphosphorylated, mono-phosphorylated, or bisphosphorylated (using sequential incubations in λ-phosphatase, PKD, and PKA, respectively). Phos-tag gels, Western blotting, and high-resolution MS revealed that PKD produced >90% monophosphorylation of cTnI, primarily at Ser-24, whereas PKA led to cTnI bisphosphorylation exclusively. PKD markedly decreased the Ca2+ sensitivity of force production in detergent-permeabilized ventricular trabeculae, whereas subsequent incubation with PKA produced only a small further fall of Ca2+ sensitivity. Unlike PKD, PKA also substantially phosphorylated myosin-binding protein-C and significantly accelerated cross-bridge kinetics (ktr). After phosphorylation by PKD or PKA, cTnI in isolated myofibrils was partially protected from calpain-mediated degradation. We conclude that cTnI monophosphorylation at Ser-23/24 decreases myofibrillar Ca2+ sensitivity and partially protects cTnI from calpain-induced proteolysis. In healthy cardiomyocytes, the basal monophosphorylation of cTnI may help tonically regulate myofibrillar Ca2+ sensitivity.


Assuntos
Cálcio/metabolismo , Calpaína/farmacologia , Miócitos Cardíacos/fisiologia , Miofibrilas/fisiologia , Proteólise/efeitos dos fármacos , Serina/metabolismo , Troponina I/metabolismo , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miofibrilas/efeitos dos fármacos , Fosforilação , Proteína Quinase C/metabolismo , Ratos , Serina/química
7.
Sci Rep ; 7(1): 15424, 2017 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-29133913

RESUMO

Myeloid derived suppressor cells (MDSC) produce nitric oxide (NO) and inhibit dendritic cell (DC) immune responses in cancer. DCs present cancer cell antigens to CD4+ T cells through Jak-STAT signal transduction. In this study, NO donors (SNAP and DETA-NONOate) inhibited DC antigen presentation. As expected, MDSC isolated from peripheral blood mononuclear cells (PBMC) from cancer patients produced high NO levels. We hypothesized that NO producing MDSC in tumor-bearing hosts would inhibit DC antigen presentation. Antigen presentation from DCs to CD4+ T cells (T cell receptor transgenic OT-II) was measured via a [3H]-thymidine incorporation proliferation assay. MDSC from melanoma tumor models decreased the levels of proliferation more than pancreatic cancer derived MDSC. T cell proliferation was restored when MDSC were treated with inhibitors of inducible nitric oxide synthase (L-NAME and NCX-4016). A NO donor inhibited OT II T cell receptor recognition of OT II specific tetramers, thus serving as a direct measure of NO inhibition of antigen presentation. Our group has previously demonstrated that STAT1 nitration also mediates MDSC inhibitory effects on immune cells. Therefore, a novel liquid chromatography-tandem mass spectrometry assay demonstrated that nitration of the STAT1-Tyr701 occurs in PBMC derived from both pancreatic cancer and melanoma patients.


Assuntos
Melanoma Experimental/imunologia , Células Supressoras Mieloides/metabolismo , Óxido Nítrico/metabolismo , Neoplasias Pancreáticas/imunologia , Fator de Transcrição STAT1/metabolismo , Animais , Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Humanos , Melanoma Experimental/patologia , Camundongos , Camundongos Transgênicos , Células Supressoras Mieloides/imunologia , Óxido Nítrico/imunologia , Doadores de Óxido Nítrico/metabolismo , Neoplasias Pancreáticas/sangue , Fator de Transcrição STAT1/análise , Espectrometria de Massas em Tandem
8.
Sci Transl Med ; 9(400)2017 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-28747516

RESUMO

The human sinoatrial node (SAN) efficiently maintains heart rhythm even under adverse conditions. However, the specific mechanisms involved in the human SAN's ability to prevent rhythm failure, also referred to as its robustness, are unknown. Challenges exist because the three-dimensional (3D) intramural structure of the human SAN differs from well-studied animal models, and clinical electrode recordings are limited to only surface atrial activation. Hence, to innovate the translational study of human SAN structural and functional robustness, we integrated intramural optical mapping, 3D histology reconstruction, and molecular mapping of the ex vivo human heart. When challenged with adenosine or atrial pacing, redundant intranodal pacemakers within the human SAN maintained automaticity and delivered electrical impulses to the atria through sinoatrial conduction pathways (SACPs), thereby ensuring a fail-safe mechanism for robust maintenance of sinus rhythm. During adenosine perturbation, the primary central SAN pacemaker was suppressed, whereas previously inactive superior or inferior intranodal pacemakers took over automaticity maintenance. Sinus rhythm was also rescued by activation of another SACP when the preferential SACP was suppressed, suggesting two independent fail-safe mechanisms for automaticity and conduction. The fail-safe mechanism in response to adenosine challenge is orchestrated by heterogeneous differences in adenosine A1 receptors and downstream GIRK4 channel protein expressions across the SAN complex. Only failure of all pacemakers and/or SACPs resulted in SAN arrest or conduction block. Our results unmasked reserve mechanisms that protect the human SAN pacemaker and conduction complex from rhythm failure, which may contribute to treatment of SAN arrhythmias.


Assuntos
Arritmias Cardíacas/fisiopatologia , Nó Sinoatrial/metabolismo , Nó Sinoatrial/fisiologia , Potenciais de Ação/efeitos dos fármacos , Adenosina/farmacologia , Adulto , Idoso , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/prevenção & controle , Eletrocardiografia , Feminino , Átrios do Coração/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Humanos , Técnicas In Vitro , Pessoa de Meia-Idade , Nó Sinoatrial/efeitos dos fármacos
9.
Circulation ; 134(6): 486-98, 2016 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-27462069

RESUMO

BACKGROUND: Adenosine provokes atrial fibrillation (AF) with a higher activation frequency in right atria (RA) versus left atria (LA) in patients, but the underlying molecular and functional substrates are unclear. We tested the hypothesis that adenosine-induced AF is driven by localized reentry in RA areas with highest expression of adenosine A1 receptor and its downstream GIRK (G protein-coupled inwardly rectifying potassium channels) channels (IK,Ado). METHODS: We applied biatrial optical mapping and immunoblot mapping of various atrial regions to reveal the mechanism of adenosine-induced AF in explanted failing and nonfailing human hearts (n=37). RESULTS: Optical mapping of coronary-perfused atria (n=24) revealed that adenosine perfusion (10-100 µmol/L) produced more significant shortening of action potential durations in RA (from 290±45 to 239±41 ms, 17.3±10.4%; P<0.01) than LA (from 307±24 to 286±23 ms, 6.7±6.6%; P<0.01). In 10 hearts, adenosine induced AF (317±116 s) that, when sustained (≥2 minutes), was primarily maintained by 1 to 2 localized reentrant drivers in lateral RA. Tertiapin (10-100 nmol/L), a selective GIRK channel blocker, counteracted adenosine-induced action potential duration shortening and prevented AF induction. Immunoblotting showed that the superior/middle lateral RA had significantly higher adenosine A1 receptor (2.7±1.7-fold; P<0.01) and GIRK4 (1.7±0.8-fold; P<0.05) protein expression than lateral/posterior LA. CONCLUSIONS: This study revealed a 3-fold RA-to-LA adenosine A1 receptor protein expression gradient in the human heart, leading to significantly greater RA versus LA repolarization sensitivity in response to adenosine. Sustained adenosine-induced AF is maintained by reentrant drivers localized in lateral RA regions with the highest adenosine A1 receptor/GIRK4 expression. Selective atrial GIRK channel blockade may effectively treat AF during conditions with increased endogenous adenosine.


Assuntos
Adenosina/toxicidade , Fibrilação Atrial/induzido quimicamente , Fibrilação Atrial/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/biossíntese , Átrios do Coração/metabolismo , Receptor A1 de Adenosina/biossíntese , Adulto , Idoso , Feminino , Regulação da Expressão Gênica , Coração/diagnóstico por imagem , Coração/efeitos dos fármacos , Átrios do Coração/diagnóstico por imagem , Átrios do Coração/efeitos dos fármacos , Sistema de Condução Cardíaco/diagnóstico por imagem , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada
10.
Circ Arrhythm Electrophysiol ; 8(5): 1219-27, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26304511

RESUMO

BACKGROUND: The hyperpolarization-activated current, If, plays an important role in sinoatrial node (SAN) pacemaking. Surprisingly, the distribution of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in human SAN has only been investigated at the mRNA level. Our aim was to define the expression pattern of HCN proteins in human SAN and different atrial regions. METHODS AND RESULTS: Entire SAN complexes were isolated from failing (n=5) and nonfailing (n=9) human hearts cardioplegically arrested in the operating room. Three-dimensional intramural SAN structure was identified as the fibrotic compact region around the SAN artery with Connexin 43-negative pacemaker cardiomyocytes visualized in Masson's trichrome and immunostained cryosections. SAN protein was precisely isolated from the adjacent frozen SAN tissue blocks using a 16G biopsy needle. The purity of the SAN protein was confirmed by Connexin 43 immunoblot. All 3 HCN isoform proteins were detected in SAN. HCN1 was predominantly distributed in the human SAN with a 125.1±40.2 (n=12) expression ratio of SAN to right atrium. HCN2 and HCN4 expression levels were higher in SAN than in atria, with SAN to right atrium ratios of 6.1±0.9 and 4.6±0.6 (n=12), respectively. CONCLUSIONS: This is the first study to conduct precise 3D molecular mapping of the human SAN by isolating pure pacemaker SAN tissue. All 3 cardiac HCN isoforms had higher expression in the SAN than in the atria. HCN1 was almost exclusively expressed in SAN, emphasizing its utility as a new specific molecular marker of the human SAN and as a potential target of specific treatments intended to modify sinus rhythm.


Assuntos
Insuficiência Cardíaca/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Miócitos Cardíacos/metabolismo , Nó Sinoatrial/metabolismo , Adulto , Idoso , Feminino , Humanos , Immunoblotting , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/metabolismo , Canais de Potássio/metabolismo , Isoformas de Proteínas/metabolismo
11.
Circulation ; 130(4): 315-24, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-24838362

RESUMO

BACKGROUND: Although sinoatrial node (SAN) dysfunction is a hallmark of human heart failure (HF), the underlying mechanisms remain poorly understood. We aimed to examine the role of adenosine in SAN dysfunction and tachy-brady arrhythmias in chronic HF. METHODS AND RESULTS: We applied multiple approaches to characterize SAN structure, SAN function, and adenosine A1 receptor expression in control (n=17) and 4-month tachypacing-induced chronic HF (n=18) dogs. Novel intramural optical mapping of coronary-perfused right atrial preparations revealed that adenosine (10 µmol/L) markedly prolonged postpacing SAN conduction time in HF by 206 ± 99 milliseconds (versus 66 ± 21 milliseconds in controls; P=0.02). Adenosine induced SAN intranodal conduction block or microreentry in 6 of 8 dogs with HF versus 0 of 7 controls (P=0.007). Adenosine-induced SAN conduction abnormalities and automaticity depression caused postpacing atrial pauses in HF versus control dogs (17.1 ± 28.9 versus 1.5 ± 1.3 seconds; P<0.001). Furthermore, 10 µmol/L adenosine shortened atrial repolarization and led to pacing-induced atrial fibrillation in 6 of 7 HF versus 0 of 7 control dogs (P=0.002). Adenosine-induced SAN dysfunction and atrial fibrillation were abolished or prevented by adenosine A1 receptor antagonists (50 µmol/L theophylline/1 µmol/L 8-cyclopentyl-1,3-dipropylxanthine). Adenosine A1 receptor protein expression was significantly upregulated during HF in the SAN (by 47 ± 19%) and surrounding atrial myocardium (by 90 ± 40%). Interstitial fibrosis was significantly increased within the SAN in HF versus control dogs (38 ± 4% versus 23 ± 4%; P<0.001). CONCLUSIONS: In chronic HF, adenosine A1 receptor upregulation in SAN pacemaker and atrial cardiomyocytes may increase cardiac sensitivity to adenosine. This effect may exacerbate conduction abnormalities in the structurally impaired SAN, leading to SAN dysfunction, and potentiate atrial repolarization shortening, thereby facilitating atrial fibrillation. Atrial fibrillation may further depress SAN function and lead to tachy-brady arrhythmias in HF.


Assuntos
Bradicardia/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Receptor A1 de Adenosina/biossíntese , Nó Sinoatrial/fisiopatologia , Taquicardia/fisiopatologia , Imagens com Corantes Sensíveis à Voltagem/métodos , Potenciais de Ação/efeitos dos fármacos , Adenosina/administração & dosagem , Adenosina/farmacologia , Adenosina/toxicidade , Antagonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina/uso terapêutico , Animais , Fibrilação Atrial/etiologia , Fibrilação Atrial/fisiopatologia , Bradicardia/etiologia , Estimulação Cardíaca Artificial/efeitos adversos , Cães , Relação Dose-Resposta a Droga , Fibrose , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiopatologia , Insuficiência Cardíaca/genética , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/fisiologia , Nó Sinoatrial/efeitos dos fármacos , Nó Sinoatrial/patologia , Taquicardia/etiologia , Teofilina/farmacologia , Teofilina/uso terapêutico , Regulação para Cima , Xantinas/farmacologia , Xantinas/uso terapêutico
12.
J Mol Cell Cardiol ; 72: 177-85, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24657721

RESUMO

The binding of Ca(2+) to troponin C (TnC) in the troponin complex is a critical step regulating the thin filament, the actin-myosin interaction and cardiac contraction. Phosphorylation of the troponin complex is a key regulatory mechanism to match cardiac contraction to demand. Here we demonstrate that phosphorylation of the troponin I (TnI) subunit is simultaneously increased at Ser-150 and Ser-23/24 during in vivo myocardial ischemia. Myocardial ischemia decreases intracellular pH resulting in depressed binding of Ca(2+) to TnC and impaired contraction. To determine the pathological relevance of these simultaneous TnI phosphorylations we measured individual TnI Ser-150 (S150D), Ser-23/24 (S23/24D) and combined (S23/24/150D) pseudo-phosphorylation effects on thin filament regulation at acidic pH similar to that in myocardial ischemia. Results demonstrate that while acidic pH decreased thin filament Ca(2+) binding to TnC regardless of TnI composition, TnI S150D attenuated this decrease rendering it similar to non-phosphorylated TnI at normal pH. The dissociation of Ca(2+) from TnC was unaltered by pH such that TnI S150D remained slow, S23/24D remained accelerated and the combined S23/24/150D remained accelerated. This effect of the combined TnI Ser-150 and Ser-23/24 pseudo-phosphorylations to maintain Ca(2+) binding while accelerating Ca(2+) dissociation represents the first post-translational modification of troponin by phosphorylation to both accelerate thin filament deactivation and maintain Ca(2+) sensitive activation. These data suggest that TnI Ser-150 phosphorylation induced attenuation of the pH-dependent decrease in Ca(2+) sensitivity and its combination with Ser-23/24 phosphorylation to maintain accelerated thin filament deactivation may impart an adaptive role to preserve contraction during acidic ischemia pH without slowing relaxation.


Assuntos
Citoesqueleto de Actina/metabolismo , Cálcio/metabolismo , Ventrículos do Coração/metabolismo , Infarto do Miocárdio/metabolismo , Processamento de Proteína Pós-Traducional , Troponina I/metabolismo , Actinas/metabolismo , Adaptação Fisiológica , Animais , Ventrículos do Coração/patologia , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica , Infarto do Miocárdio/patologia , Miosinas/metabolismo , Fosforilação , Ligação Proteica , Troponina C/metabolismo
13.
J Appl Physiol (1985) ; 116(9): 1165-74, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24651988

RESUMO

Repeated, intense contractile activity compromises the ability of skeletal muscle to generate force and velocity, resulting in fatigue. The decrease in velocity is thought to be due, in part, to the intracellular build-up of acidosis inhibiting the function of the contractile proteins myosin and troponin; however, the underlying molecular basis of this process remains poorly understood. We sought to gain novel insight into the decrease in velocity by determining whether the depressive effect of acidosis could be altered by 1) introducing Ca(++)-sensitizing mutations into troponin (Tn) or 2) by agents that directly affect myosin function, including inorganic phosphate (Pi) and 2-deoxy-ATP (dATP) in an in vitro motility assay. Acidosis reduced regulated thin-filament velocity (VRTF) at both maximal and submaximal Ca(++) levels in a pH-dependent manner. A truncated construct of the inhibitory subunit of Tn (TnI) and a Ca(++)-sensitizing mutation in the Ca(++)-binding subunit of Tn (TnC) increased VRTF at submaximal Ca(++) under acidic conditions but had no effect on VRTF at maximal Ca(++) levels. In contrast, both Pi and replacement of ATP with dATP reversed much of the acidosis-induced depression of VRTF at saturating Ca(++). Interestingly, despite producing similar magnitude increases in VRTF, the combined effects of Pi and dATP were additive, suggesting different underlying mechanisms of action. These findings suggest that acidosis depresses velocity by slowing the detachment rate from actin but also by possibly slowing the attachment rate.


Assuntos
Acidose/genética , Cálcio/metabolismo , Nucleotídeos de Desoxiadenina/genética , Mutação/genética , Fosfatos/fisiologia , Troponina/genética , Acidose/metabolismo , Actinas/química , Actinas/genética , Sequência de Aminoácidos , Animais , Galinhas , Nucleotídeos de Desoxiadenina/química , Humanos , Dados de Sequência Molecular , Miosinas/química , Miosinas/genética , Estrutura Secundária de Proteína , Coelhos , Troponina/química
14.
J Physiol ; 591(18): 4535-47, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23836688

RESUMO

According to the Frank-Starling relationship, greater end-diastolic volume increases ventricular output. The Frank-Starling relationship is based, in part, on the length-tension relationship in cardiac myocytes. Recently, we identified a dichotomy in the steepness of length-tension relationships in mammalian cardiac myocytes that was dependent upon protein kinase A (PKA)-induced myofibrillar phosphorylation. Because PKA has multiple myofibrillar substrates including titin, myosin-binding protein-C and cardiac troponin I (cTnI), we sought to define if phosphorylation of one of these molecules could control length-tension relationships. We focused on cTnI as troponin can be exchanged in permeabilized striated muscle cell preparations, and tested the hypothesis that phosphorylation of cTnI modulates length dependence of force generation. For these experiments, we exchanged unphosphorylated recombinant cTn into either a rat cardiac myocyte preparation or a skinned slow-twitch skeletal muscle fibre. In all cases unphosphorylated cTn yielded a shallow length-tension relationship, which was shifted to a steep relationship after PKA treatment. Furthermore, exchange with cTn having cTnI serines 23/24 mutated to aspartic acids to mimic phosphorylation always shifted a shallow length-tension relationship to a steep relationship. Overall, these results indicate that phosphorylation of cTnI serines 23/24 is a key regulator of length dependence of force generation in striated muscle.


Assuntos
Força Muscular , Músculo Estriado/metabolismo , Troponina I/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Masculino , Contração Muscular , Fibras Musculares de Contração Lenta/metabolismo , Fibras Musculares de Contração Lenta/fisiologia , Músculo Estriado/citologia , Músculo Estriado/fisiologia , Mutação , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Serina/genética , Serina/metabolismo , Troponina I/genética
15.
J Biol Chem ; 287(33): 27930-40, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22718768

RESUMO

The rate-limiting step of cardiac muscle relaxation has been proposed to reside in the myofilament. Both the rates of cross-bridge detachment and Ca(2+) dissociation from troponin C (TnC) have been hypothesized to rate-limit myofilament inactivation. In this study we used a fluorescent TnC to measure both the rate of Ca(2+) dissociation from TnC and the rate of cross-bridge detachment from several different species of ventricular myofibrils. The fluorescently labeled TnC was sensitive to both Ca(2+) dissociation and cross-bridge detachment at low Ca(2+) (presence of EGTA), allowing for a direct comparison between the two proposed rates of myofilament inactivation. Unlike Ca(2+) dissociation from TnC, cross-bridge detachment varied in myofibrils from different species and was rate-limited by ADP release. At subphysiological temperatures (<20 °C), the rate of Ca(2+) dissociation from TnC was faster than the rate of cross-bridge detachment in the presence of ADP. These results support the hypothesis that cross-bridge detachment rate-limits relaxation. However, Ca(2+) dissociation from TnC was not as temperature-sensitive as cross-bridge detachment. At a near physiological temperature (35 °C) and ADP, the rate of cross-bridge detachment may actually be faster than the rate of Ca(2+) dissociation. This provides evidence that there may not be a simple, single rate-limiting step of myofilament inactivation.


Assuntos
Cálcio/química , Corantes Fluorescentes/química , Ventrículos do Coração/química , Miocárdio/química , Miofibrilas/química , Troponina C/química , Difosfato de Adenosina/química , Difosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Ácido Egtázico/química , Corantes Fluorescentes/metabolismo , Ventrículos do Coração/metabolismo , Temperatura Alta , Miocárdio/metabolismo , Miofibrilas/metabolismo , Coelhos , Troponina C/metabolismo
16.
J Biol Chem ; 285(25): 19688-98, 2010 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-20410305

RESUMO

The cardiac troponin I (cTnI) isoform contains a unique N-terminal extension that functions to modulate activation of cardiac myofilaments. During cardiac remodeling restricted proteolysis of cTnI removes this cardiac specific N-terminal modulatory extension to alter myofilament regulation. We have demonstrated expression of the N-terminal-deleted cTnI (cTnI-ND) in the heart decreased the development of the cardiomyopathy like phenotype in a beta-adrenergic-deficient transgenic mouse model. To investigate the potential beneficial effects of cTnI-ND on the development of naturally occurring cardiac dysfunction, we measured the hemodynamic and biochemical effects of cTnI-ND transgenic expression in the aged heart. Echocardiographic measurements demonstrate cTnI-ND transgenic mice exhibit increased systolic and diastolic functions at 16 months of age compared with age-matched controls. This improvement likely results from decreased Ca(2+) sensitivity and increased cross-bridge kinetics as observed in skinned papillary bundles from young transgenic mice prior to the effects of aging. Hearts of cTnI-ND transgenic mice further exhibited decreased beta myosin heavy chain expression compared to age matched non-transgenic mice as well as altered cTnI phosphorylation. Finally, we demonstrated cTnI-ND expressed in the heart is not phosphorylated indicating the cTnI N-terminal is necessary for the higher level phosphorylation of cTnI. Taken together, our data suggest the regulated proteolysis of cTnI during cardiac stress to remove the unique cardiac N-terminal extension functions to improve cardiac contractility at the myofilament level and improve overall cardiac function.


Assuntos
Envelhecimento , Coração/fisiologia , Troponina I/química , Citoesqueleto de Actina/metabolismo , Adenosina Trifosfatases/química , Animais , Cálcio/química , Ecocardiografia/métodos , Camundongos , Camundongos Transgênicos , Fenótipo , Fosforilação , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Transgenes
17.
J Biol Chem ; 283(22): 15114-21, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18378675

RESUMO

The precise mechanism of cardiac troponin I (cTnI) proteolysis in myocardial stunning is not fully understood. Accordingly, we determined the effect of cTnI C terminus truncation on chemo-mechanical transduction in isolated skinned rat trabeculae. Recombinant troponin complex (cTn), containing either mouse cTnI-(1-193) or human cTnI-(1-192) was exchanged into skinned cardiac trabeculae; Western blot analysis confirmed that 60-70% of the endogenous cTn was replaced by recombinant Tn. Incorporation of truncated cTnI induced significant reductions ( approximately 50%) in maximum force and cooperative activation as well as increases ( approximately 50%) in myofilament Ca(2+) sensitivity and tension cost. Similar results were obtained with either mouse or human truncated cTn. Presence of truncated cTnI increased maximum actin-activated S1 ATPase activity as well as its Ca(2+) sensitivity in vitro. Partial exchange (50%) for truncated cTnI resulted in similar reductions in maximum force and cooperativity; tension cost was increased in proportion to truncated cTnI content. In vitro, to determine the molecular mechanism responsible for the enhanced myofilament Ca(2+) sensitivity, we measured Ca(2+) binding to cTn as reported using a fluorescent probe. Incorporation of truncated cTnI did not affect Ca(2+) binding affinity to cTn alone. However, when cTn was incorporated into thin filaments, cTnI truncation induced a significant increase in Ca(2+) binding affinity to cTn. We conclude that cTnI truncation induces depressed myofilament function. Decreased cardiac function after ischemia/reperfusion injury may directly result, in part, from proteolytic degradation of cTnI, resulting in alterations in cross-bridge cycling kinetics.


Assuntos
Mecanotransdução Celular , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio Atordoado/metabolismo , Miocárdio/metabolismo , Troponina I/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patologia , Animais , Cálcio/metabolismo , Humanos , Cinética , Masculino , Mecanotransdução Celular/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio Atordoado/genética , Miocárdio Atordoado/patologia , Miocárdio/patologia , Miosinas/metabolismo , Ratos , Ratos Endogâmicos Lew , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Troponina I/genética , Troponina I/farmacologia
18.
Circ Res ; 100(10): 1486-93, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17446435

RESUMO

Striated muscle contraction is regulated by the binding of Ca(2+) to the N-terminal regulatory lobe of the cardiac troponin C (cTnC) subunit in the troponin complex. In the heart, beta-adrenergic stimulation induces protein kinase A phosphorylation of cardiac troponin I (cTnI) at Ser23/24 to alter the interaction of cTnI with cTnC in the troponin complex and is critical to the regulation of cardiac contractility. We investigated the effect of the dilated cardiomyopathy linked cTnC Gly159 to Asp (cTnC-G159D) mutation on the development of Ca(2+)-dependent tension and ATPase rate in whole troponin-exchanged skinned rat trabeculae. Even though this mutation is located in the C-terminal lobe of cTnC, the G159D mutation was demonstrated to depress ATPase activation and filament sliding in vitro. The effects of this mutation within the cardiac myofilament are unknown. Our results demonstrate that the cTnC-G159D mutation by itself does not alter the myofilament response to Ca(2+) in the cardiac muscle lattice. However, in the presence of cTnI phosphorylated at Ser23/24, which reduced Ca(2+) sensitivity and enhanced cross-bridge cycling in controls, cTnC-G159D specifically blunted the phosphorylation induced decrease in Ca(2+)-sensitive tension development without altering cross-bridge cycling. Measurements in purified troponin confirmed that this cTnC-G159D blunting of myofilament desensitization results from altered Ca(2+)-binding to cTnC. Our results provide novel evidence that modification of the cTnC-cTnI interaction has distinct effects on troponin Ca(2+)-binding and cross-bridge kinetics to suggest a novel role for thin filament mutations in the modulation of myofilament function through beta-adrenergic signaling as well as the development of cardiomyopathy.


Assuntos
Citoesqueleto de Actina/fisiologia , Mutação , Troponina C/genética , Troponina I/metabolismo , Cálcio/metabolismo , Cardiomiopatias/etiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Humanos , Fosforilação , Receptores Adrenérgicos beta/fisiologia , Serina
19.
Am J Physiol Cell Physiol ; 290(2): C567-76, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16192301

RESUMO

Striated muscle contraction is powered by actin-activated myosin ATPase. This process is regulated by Ca(2+) via the troponin complex. Slow- and fast-twitch fibers of vertebrate skeletal muscle express type I and type II myosin, respectively, and these myosin isoenzymes confer different ATPase activities, contractile velocities, and force. Skeletal muscle troponin has also diverged into fast and slow isoforms, but their functional significance is not fully understood. To investigate the expression of troponin isoforms in mammalian skeletal muscle and their functional relationship to that of the myosin isoforms, we concomitantly studied myosin, troponin T (TnT), and troponin I (TnI) isoform contents and isometric contractile properties in single fibers of rat skeletal muscle. We characterized a large number of Triton X-100-skinned single fibers from soleus, diaphragm, gastrocnemius, and extensor digitorum longus muscles and selected fibers with combinations of a single myosin isoform and a single class (slow or fast) of the TnT and TnI isoforms to investigate their role in determining contractility. Types IIa, IIx, and IIb myosin fibers produced higher isometric force than that of type I fibers. Despite the polyploidy of adult skeletal muscle fibers, the expression of fast or slow isoforms of TnT and TnI is tightly coupled. Fibers containing slow troponin had higher Ca(2+) sensitivity than that of the fast troponin fibers, whereas fibers containing fast troponin showed a higher cooperativity of Ca(2+) activation than that of the slow troponin fibers. These results demonstrate distinct but coordinated regulation of troponin and myosin isoform expression in skeletal muscle and their contribution to the contractile properties of muscle.


Assuntos
Contração Muscular/fisiologia , Fibras Musculares Esqueléticas , Músculo Esquelético , Isoformas de Proteínas/metabolismo , Troponina I/metabolismo , Troponina T/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Cálcio/metabolismo , Masculino , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Isoformas de Proteínas/genética , Ratos , Ratos Sprague-Dawley , Troponina I/genética , Troponina T/genética
20.
J Biol Chem ; 279(14): 13825-32, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-14736877

RESUMO

Cardiac muscle contraction is regulated by Ca(2+) through the troponin complex consisting of three subunits: troponin C (TnC), troponin T (TnT), and troponin I (TnI). We reported previously that the abnormal splicing of cardiac TnT in turkeys with dilated cardiomyopathy resulted in a greater binding affinity to TnI. In the present study, we characterized a polymorphism of cardiac TnI in the heart of wild turkeys. cDNA cloning and sequencing of the novel turkey cardiac TnI revealed a single amino acid substitution, R111C. Arg(111) in avian cardiac TnI corresponds to a Lys in mammals. This residue is conserved in cardiac and skeletal muscle TnIs across the vertebrate phylum, implying a functional importance. In the partial crystal structure of cardiac troponin, this amino acid resides in an alpha-helix that directly contacts with TnT. Structural modeling indicates that the substitution of Cys for Arg or Lys at this position would not disrupt the global structure of troponin. To evaluate the functional significance of the different size and charge between the Arg and Cys side chains, protein-binding assays using purified turkey cardiac TnI expressed in Escherichia coli were performed. The results show that the R111C substitution lowered binding affinity to TnT, which is potentially compensatory to the increased TnI-binding affinity of the cardiomyopathy-related cardiac TnT splicing variant. Therefore, the fixation of the cardiac TnI Cys(111) allele in the wild turkey population and the corresponding functional effect reflect an increased fitness value, suggesting a novel target for the treatment of TnT myopathies.


Assuntos
Cardiomiopatia Dilatada/genética , Polimorfismo Genético , Troponina I/genética , Troponina T/genética , Perus/genética , Processamento Alternativo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Animais Selvagens , Arginina/genética , Galinhas , Clonagem Molecular , Cistina/genética , DNA Complementar , Expressão Gênica , Dados de Sequência Molecular , Ligação Proteica , Troponina I/química , Troponina I/metabolismo , Troponina T/química , Troponina T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA