Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Eur Heart J ; 43(20): 1973-1989, 2022 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-35190817

RESUMO

AIMS: Cereblon (CRBN) is a substrate receptor of the E3 ubiquitin ligase complex that was reported to target ion channel proteins. L-type voltage-dependent Ca2+ channel (LTCC) density and dysfunction is a critical player in heart failure with reduced ejection fraction (HFrEF). However, the underlying cellular mechanisms by which CRBN regulates LTCC subtype Cav1.2α during cardiac dysfunction remain unclear. Here, we explored the role of CRBN in HFrEF by investigating the direct regulatory role of CRBN in Cav1.2α activity and examining how it can serve as a target to address myocardial dysfunction. METHODS AND RESULTS: Cardiac tissues from HFrEF patients exhibited increased levels of CRBN compared with controls. In vivo and ex vivo studies demonstrated that whole-body CRBN knockout (CRBN-/-) and cardiac-specific knockout mice (Crbnfl/fl/Myh6Cre+) exhibited enhanced cardiac contractility with increased LTCC current (ICaL) compared with their respective controls, which was modulated by the direct interaction of CRBN with Cav1.2α. Mechanistically, the Lon domain of CRBN directly interacted with the N-terminal of Cav1.2α. Increasing CRBN levels enhanced the ubiquitination and proteasomal degradation of Cav1.2α and decreased ICaL. In contrast, genetic or pharmacological depletion of CRBN via TD-165, a novel PROTAC-based CRBN degrader, increased surface expression of Cav1.2α and enhanced ICaL. Low CRBN levels protected the heart against cardiomyopathy in vivo. CONCLUSION: Cereblon selectively degrades Cav1.2α, which in turn facilitates cardiac dysfunction. A targeted approach or an efficient method of reducing CRBN levels could serve as a promising strategy for HFrEF therapeutics.


Assuntos
Insuficiência Cardíaca , Ubiquitina-Proteína Ligases , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Humanos , Camundongos , Volume Sistólico , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
2.
Pflugers Arch ; 472(2): 259-269, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32025886

RESUMO

All living beings on earth are influenced by the circadian rhythm, the rising and the setting of the sun. The ubiquitous effect of exercise is widely believed to maximize health benefits but has not been formally investigated for cardiac responses in the exercise-induced circadian rhythms. We hypothesized that the exercise-related proteome is differentially influenced by circadian rhythm and analyzed the differences between the effects of morning and evening exercise. Twenty-four Sprague-Dawley rats were randomly divided into four groups (n = 6 per group): morning control, morning exercise, evening control, and evening exercise groups. The exercise groups were subjected to 12-week treadmill exercise (5 days/week) performed either during daytime or nighttime. After 12 weeks, the physiological characteristics (e.g., body weight, heart weight, visceral fat, and blood metabolites), cardiovascular capacity (ejection fraction (%) and fractional shortening (%)), circadian gene expression levels (clock, ball1, per1, per2, cry1, and cry2), and the proteomic data were obtained and subjected to univariate and multivariate analysis. The mRNA levels of per1 and cry2 increased in the evening group compared with those in the morning group. We also found that per2 decreased and cry2 increased in the evening exercise groups. The evening exercise groups showed more decreased triacylglycerides and increased blood insulin levels than the morning exercise group. The principal component analysis, partial least squares discriminant analysis, and orthogonal partial least squares discriminant analysis indicated that the circadian rhythm differently influenced the protein networks of the exercise groups. In the morning exercise group, the transcription-translation feedback loop (TTFL) (clock, per1, per2, cry1, and cry2) formed a protein-protein interaction network with Nme2, Hint1, Ddt, Ndufb8, Ldha, and Eef1a2. In contrast, the TTFL group appeared close to Maoa, Hist2h4, and Macrod1 in the evening exercise group. Interestingly, the evening exercise group decreased the mRNA level of per2 but not per1. Per1 and Per2 are known to transport Cry1 and Cry2 into the nucleus. Taken together, we summarized the characteristics of enriched proteins in the aspect of their molecular function, cellular component, and biological process. Our results might provide a better understanding of the circadian effect on exercise-related proteins.


Assuntos
Adaptação Fisiológica , Ritmo Circadiano , Miocárdio/metabolismo , Condicionamento Físico Animal , Proteoma/metabolismo , Animais , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Coração/fisiologia , Masculino , Mapas de Interação de Proteínas , Proteoma/genética , Ratos , Ratos Sprague-Dawley
3.
Adv Physiol Educ ; 44(3): 323-333, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32568002

RESUMO

To understand the excitation-contraction (E-C) coupling of cardiomyocytes, including the electrophysiological mechanism of their characteristically long action potential duration, is one of the major learning goals in medical physiology. However, the integrative interpretation of the responses occurring during the contraction-relaxation cycle is challenging due to the dynamic interaction of underlying factors. Starting in 2017, we adopted the mathematical computer simulation model of human ventricular myocyte (Cardiac E-C_Sim), hypothesizing that this educational technology may facilitate students' learning of cardiac physiology. Here, we describe the overall process for the educational application of Cardiac E-C_Sim in the human physiology practicum of Seoul National University College of Medicine. We also report the results from questionnaires covering detailed assessment of the practicum class. The analysis of results and feedback opinions enabled us to understand how the students had approached the problem-solving process. As a whole, the students could better accomplish the learning goals using Cardiac E-C_Sim, followed by constructive discussions on the complex and dynamic mechanisms of cardiac E-C coupling. We suggest that the combined approach of lecture-based teaching and computer simulations guided by a manual containing clinical context would be broadly applicable in physiology education.


Assuntos
Contração Miocárdica , Miócitos Cardíacos , Potenciais de Ação , Simulação por Computador , Humanos , Aprendizagem , Ensino
4.
Biophys J ; 117(4): 767-779, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31400920

RESUMO

Pacemaker depolarization in interstitial cells of Cajal (ICCs) is believed to be induced by Ca2+ transients and activation of anoctamin-1 (Ano1) channels in the plasma membrane. However, block of store-operated calcium entry (SOCE) or the Na-K-2Cl cotransporter (NKCC1) terminates pacemaker activity in ICC, indicating these transporters are involved in the initiation or maintenance of pacemaker activity. We hypothesized that SOCE contributes to pacemaker depolarization by maintaining [Ca2+] in the endoplasmic reticulum, which is the underlying source of Ca2+ transients for activation of Ano1. NKCC1 maintains the Cl- gradient supporting the driving force for inward current mediated by Ano1. Currently mechanisms sustaining release of Ca2+ and activation of Ano1 channels during the plateau phase of slow waves are unknown, but the reverse mode of the Na+/Ca2+ exchange may contribute. We generated a mathematical model of pacemaker activity based on current empirical observations from ICC of mouse small intestine that incorporates functions of SOCE and NKCC1. This model reproduces experimental findings, suggesting roles for SOCE and Ano1 channels: blocking of either NKCC1 or SOCE in our model terminates pacemaker activity. Direct contribution of NKCC1 to pacemaker activity in a beat-to-beat manner is not predicted by our model. Instead, NKCC1 plays a maintenance role supporting the driving force for Cl- efflux. Incorporation of SOCE allows the model to drive pacemaker activity without a diastolic depolarization, as observed in cardiac pacemaking. Further biological experiments are necessary to validate and further refine the roles of NKCC1, Na+/Ca2+ exchange, and Ano1 in the pacemaker mechanism of ICC.


Assuntos
Relógios Biológicos , Sinalização do Cálcio , Células Intersticiais de Cajal/metabolismo , Modelos Neurológicos , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Potenciais de Ação , Animais , Cálcio/metabolismo , Humanos , Células Intersticiais de Cajal/fisiologia
6.
Int J Mol Sci ; 20(24)2019 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-31842522

RESUMO

Diabetes mellitus is associated with cardiovascular, ophthalmic, and renal comorbidities. Among these, diabetic cardiomyopathy (DCM) causes the most severe symptoms and is considered to be a major health problem worldwide. Exercise is widely known as an effective strategy for the prevention and treatment of many chronic diseases. Importantly, the onset of complications arising due to diabetes can be delayed or even prevented by exercise. Regular exercise is reported to have positive effects on diabetes mellitus and the development of DCM. The protective effects of exercise include prevention of cardiac apoptosis, fibrosis, oxidative stress, and microvascular diseases, as well as improvement in cardiac mitochondrial function and calcium regulation. This review summarizes the recent scientific findings to describe the potential mechanisms by which exercise may prevent DCM and heart failure.


Assuntos
Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/terapia , Terapia por Exercício , Exercício Físico , Animais , Biomarcadores , Estudos Clínicos como Assunto , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/fisiopatologia , Terapia por Exercício/métodos , Humanos , Miocárdio/metabolismo , Estresse Oxidativo
7.
Pflugers Arch ; 470(2): 263-275, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29032504

RESUMO

Metabolic disturbance and mitochondrial dysfunction are a hallmark of diabetic cardiomyopathy (DC). Resistance exercise (RE) not only enhances the condition of healthy individuals but could also improve the status of those with disease. However, the beneficial effects of RE in the prevention of DC and mitochondrial dysfunction are uncertain. Therefore, this study investigated whether RE attenuates DC by improving mitochondrial function using an in vivo rat model of diabetes. Fourteen Otsuka Long-Evans Tokushima Fatty rats were assigned to sedentary control (SC, n = 7) and RE (n = 7) groups at 28 weeks of age. Long-Evans Tokushima Otsuka rats were used as the non-diabetic control. The RE rats were trained by 20 repetitions of climbing a ladder 5 days per week. RE rats exhibited higher glucose uptake and lower lipid profiles, indicating changes in energy metabolism. RE rats significantly increased the ejection fraction and fractional shortening compared with the SC rats. Isolated mitochondria in RE rats showed increase in mitochondrial numbers, which were accompanied by higher expression of mitochondrial biogenesis proteins such as proliferator-activated receptor-γ coactivator-1α and TFAM. Moreover, RE rats reduced proton leakage and reactive oxygen species production, with higher membrane potential. These results were accompanied by higher superoxide dismutase 2 and lower uncoupling protein 2 (UCP2) and UCP3 levels in RE rats. These data suggest that RE is effective at ameliorating DC by improving mitochondrial function, which may contribute to the maintenance of diabetic cardiac contractility.


Assuntos
Cardiomiopatias Diabéticas/prevenção & controle , Metabolismo Energético , Mitocôndrias Musculares/metabolismo , Contração Miocárdica , Condicionamento Físico Animal/métodos , Animais , Cardiomiopatias Diabéticas/fisiopatologia , Metabolismo dos Lipídeos , Masculino , Ratos , Ratos Long-Evans
8.
Pflugers Arch ; 468(4): 609-22, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26687128

RESUMO

Na(+)/Ca(2+) exchanger current (INCX) triggered by spontaneous Ca(2+) release from sarcoplasmic reticulum (SR) has been suggested as one of the cardiac pacemaker mechanisms ("Ca(2+) clock model"). In human embryonic stem cell-derived cardiomyocytes (hESC-CMs) showing spontaneous action potentials (APs), we found that substantial population (35 %) showed regular oscillation of inward currents (SICs) in nystatin-perforated voltage clamp between -40 and 40 mV (-80 ± 10.6 pA, at -20 mV). SICs were similarly observed between nodal, atrial, and ventricular hESC-CMs. Oscillations of [Ca(2+)]i synchronized with SICs were observed under voltage clamp. SICs were eliminated by lowering [Ca(2+)]e, L-type Ca(2+) channel (VOCCL) blocker (nifedipine, 10 µM), ryanodine receptor (RyR) agonist (caffeine, 10 mM), or NCX inhibitor (1 µM SN-6 and 10 µM KB-R7943). Plasma membrane expression of NCX1 was confirmed using immunofluorescence confocal microcopy. Both caffeine and SN-6 slowed the pacemaker potential but did not abolish the AP generation. The inhibitors of funny current (3 µM ivabradine) or voltage-gated K(+) channel currents (1 µM E4031 and 10 µM chromanol-293B) also did not abolish but slowed the pacemaker potential. In a computational model of cardiac pacemaker by Maltsev and Lakatta (2009), after modifying the spatial distribution of RyR, VOCCL, and NCX by using our multiparameter adjust algorithm, we could successfully reproduce spontaneous SR Ca(2+) release and SICs under voltage clamp. It was proposed that, under the membrane depolarization activating VOCCL, oscillatory Ca(2+) releases via RyR induce sharp increases in subsarcolemmal [Ca(2+)]i and inward INCX (SICs). Since the hESC-CMs without SICs still showed spontaneous APs, the putative "Ca(2+) clock" would provide a redundant pacemaker or augmenting mechanism in hESC-CMs.


Assuntos
Potenciais de Ação , Sinalização do Cálcio , Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Diferenciação Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
9.
Pflugers Arch ; 468(11-12): 1995-2006, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27796576

RESUMO

Fatty acid (FA)-dependent oxidation is the predominant process for energy supply in normal heart. Impaired FA metabolism and metabolic insufficiency underlie the failing of the myocardium. So far, FA metabolism in normal cardiac physiology and heart failure remains undetermined. Here, we evaluate the mechanisms of FA and major metabolic substrates (termed NF) on the contraction, relaxation, and Ca2+ handling in rat left ventricular (LV) myocytes. Our results showed that NF significantly increased myocyte contraction and facilitated relaxation. Moreover, NF increased the amplitudes of diastolic and systolic Ca2+ transients ([Ca2+]i), abbreviated time constant of [Ca2+]i decay (tau), and prolonged the peak duration of [Ca2+]i. Whole-cell patch-clamp experiments revealed that NF increased Ca2+ influx via L-type Ca2+ channels (LTCC, ICa-integral) and prolonged the action potential duration (APD). Further analysis revealed that NF shifted the relaxation phase of sarcomere lengthening vs. [Ca2+]i trajectory to the right and increased [Ca2+]i for 50 % of sarcomere relengthening (EC50), suggesting myofilament Ca2+ desensitization. Butanedione monoxime (BDM), a myosin ATPase inhibitor that reduces myofilament Ca2+ sensitivity, abolished the NF-induced enhancement of [Ca2+]i amplitude and the tau of [Ca2+]i decay, indicating the association of myofilament Ca2+ desensitization with the changes in [Ca2+]i profile in NF. NF reduced intracellular pH ([pHi]). Increasing [pH]i buffer capacity with HCO3/CO2 attenuated Δ [pH]i and reversed myofilament Ca2+ desensitization and Ca2+ handling in NF. Collectively, greater Ca2+ influx through LTCCs and myofilament Ca2+ desensitization, via reducing [pH]i, are likely responsible for the positive inotropic and lusitropic effects of NF. Computer simulation recapitulated the effects of NF.


Assuntos
Sinalização do Cálcio , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Animais , Bicarbonatos/metabolismo , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Ácidos Graxos/metabolismo , Ventrículos do Coração/citologia , Masculino , Miócitos Cardíacos/fisiologia , Ratos , Ratos Sprague-Dawley
10.
Proc Natl Acad Sci U S A ; 110(22): E2064-73, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23676270

RESUMO

Ca(2+) signaling regulates cell function. This is subject to modulation by H(+) ions that are universal end-products of metabolism. Due to slow diffusion and common buffers, changes in cytoplasmic [Ca(2+)] ([Ca(2+)]i) or [H(+)] ([H(+)]i) can become compartmentalized, leading potentially to complex spatial Ca(2+)/H(+) coupling. This was studied by fluorescence imaging of cardiac myocytes. An increase in [H(+)]i, produced by superfusion of acetate (salt of membrane-permeant weak acid), evoked a [Ca(2+)]i rise, independent of sarcolemmal Ca(2+) influx or release from mitochondria, sarcoplasmic reticulum, or acidic stores. Photolytic H(+) uncaging from 2-nitrobenzaldehyde also raised [Ca(2+)]i, and the yield was reduced following inhibition of glycolysis or mitochondrial respiration. H(+) uncaging into buffer mixtures in vitro demonstrated that Ca(2+) unloading from proteins, histidyl dipeptides (HDPs; e.g., carnosine), and ATP can underlie the H(+)-evoked [Ca(2+)]i rise. Raising [H(+)]i tonically at one end of a myocyte evoked a local [Ca(2+)]i rise in the acidic microdomain, which did not dissipate. The result is consistent with uphill Ca(2+) transport into the acidic zone via Ca(2+)/H(+) exchange on diffusible HDPs and ATP molecules, energized by the [H(+)]i gradient. Ca(2+) recruitment to a localized acid microdomain was greatly reduced during intracellular Mg(2+) overload or by ATP depletion, maneuvers that reduce the Ca(2+)-carrying capacity of HDPs. Cytoplasmic HDPs and ATP underlie spatial Ca(2+)/H(+) coupling in the cardiac myocyte by providing ion exchange and transport on common buffer sites. Given the abundance of cellular HDPs and ATP, spatial Ca(2+)/H(+) coupling is likely to be of general importance in cell signaling.


Assuntos
Trifosfato de Adenosina/metabolismo , Sinalização do Cálcio/fisiologia , Citoplasma/metabolismo , Dipeptídeos/metabolismo , Histidina/metabolismo , Miócitos Cardíacos/metabolismo , Prótons , Animais , Fluorometria , Microscopia de Fluorescência , Ratos
11.
Pflugers Arch ; 467(8): 1689-97, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25196539

RESUMO

Diabetes mellitus and hypertension are common diseases frequently coexisting. Although augmentation of L-type Ca(2+) channel (ICaL) activity has been reported in vascular smooth muscle cells (VSMCs) of a spontaneously hypertensive rat model, no study on ICaL has been conducted for coexisting hypertension and diabetes. Sprague Dawley rats were assigned to four groups: a sham-operated control group (CG), a unilateral nephrectomy group (UNG), a streptozotocin (STZ)-induced type 1 diabetic group (SDG) and a coexisting hypertension and diabetes group (DHG), which underwent nephrectomy and received STZ injection. Blood pressure (BP) was significantly lower in the CG than in the other three groups. The membrane capacitance of VSMCs was nearly doubled in the SDG and DHG but not in the UNG. The ICaL was increased approximately 2-fold in both the UNG and SDG and approximately 4-fold in the DHG. The current density of ICaL was increased approximately 2-fold in the UNG and DHG, while no significant increase was seen in the SDG. The rate of Ca(2+) removal was inhibited significantly, by ~33 %, in the DHG. In conclusion, the effects of hypertension and diabetes on ICaL were apparently additive, and the vascular consequences of combined diabetes and hypertension may be caused by an elevated ICaL and slowed Ca(2+) removal.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Hipertensão/metabolismo , Mesentério/irrigação sanguínea , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Nefrectomia , Estreptozocina , Animais , Arteríolas/metabolismo , Pressão Sanguínea , Cálcio/metabolismo , Sinalização do Cálcio , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/fisiopatologia , Hipertensão/etiologia , Hipertensão/fisiopatologia , Potenciais da Membrana , Músculo Liso Vascular/fisiopatologia , Ratos Sprague-Dawley , Fatores de Tempo
12.
Pflugers Arch ; 467(10): 2151-63, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25410495

RESUMO

Echinochrome A (Ech A), a marine bio-product isolated from sea urchin eggs, is known to have cardioprotective effects through its strong antioxidant and ATP-sparing capabilities. However, the effects of Ech A on cardiac excitation-contraction (E-C) are not known. In this study, we investigated the effects of Ech A on cardiac contractility and Ca(2+) handling in the rat heart. In ex vivo Langendorff hearts, Ech A (3 µM) decreased left ventricular developing pressure to 77.7 ± 6.5 % of basal level. In isolated ventricular myocytes, Ech A reduced the fractional cell shortening from 3.4 % at baseline to 2.1 %. Ech A increased both diastolic and peak systolic intracellular Ca(2+) ([Ca(2+)]i). However, the ratio of peak [Ca]i to resting [Ca]i was significantly decreased. Ech A did not affect the L-type Ca(2+) current. Inhibiting the Na(+)/Ca(2+) exchanger with either NiCl2 or SEA400 did not affect the Ech A-dependent changes in Ca(2+) handling. Our data demonstrate that treatment with Ech A results in a significant reduction in the phosphorylation of phospholamban at both serine 16 and threonine 17 leading to a significant inhibition of SR Ca(2+)-ATPase 2A (SERCA2A) and subsequent reduced Ca(2+) uptake into the intracellular Ca(2+) store. Taken together, our data show that Ech A negatively regulates cardiac contractility by inhibiting SERCA2A activity, which leads to a reduction in internal Ca(2+) stores.


Assuntos
Sinalização do Cálcio , Proteínas de Ligação ao Cálcio/metabolismo , Cardiotônicos/farmacologia , Miócitos Cardíacos/metabolismo , Naftoquinonas/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Animais , Células Cultivadas , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Masculino , Contração Miocárdica , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Fosforilação , Ratos , Ratos Wistar , Serina/metabolismo , Treonina/metabolismo , Função Ventricular
13.
J Physiol ; 592(5): 991-1007, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24297849

RESUMO

Carbonic anhydrase enzymes (CAs) catalyse the reversible hydration of CO2 to H+ and HCO3- ions. This catalysis is proposed to be harnessed by acid/base transporters, to facilitate their transmembrane flux activity, either through direct protein-protein binding (a 'transport metabolon') or local functional interaction. Flux facilitation has previously been investigated by heterologous co-expression of relevant proteins in host cell lines/oocytes. Here, we examine the influence of intrinsic CA activity on membrane HCO3- or H+ transport via the native acid-extruding proteins, Na+ -HCO3- cotransport (NBC) and Na+ / H+ exchange (NHE), expressed in enzymically isolated mammalian ventricular myocytes. Effects of intracellular and extracellular (exofacial) CA (CAi and CAe) are distinguished using membrane-permeant and -impermeant pharmacological CA inhibitors, while measuring transporter activity in the intact cell using pH and Na+ fluorophores. We find that NBC, but not NHE flux is enhanced by catalytic CA activity, with facilitation being confined to CAi activity alone. Results are quantitatively consistent with a model where CAi catalyses local H+ ion delivery to the NBC protein, assisting the subsequent (uncatalysed) protonation and removal of imported HCO3- ions. In well-superfused myocytes, exofacial CA activity is superfluous, most likely because extracellular CO2/HCO3- buffer is clamped at equilibrium. The CAi insensitivity of NHE flux suggests that, in the native cell, intrinsic mobile buffer-shuttles supply sufficient intracellular H+ ions to this transporter, while intrinsic buffer access to NBC proteins is restricted. Our results demonstrate a selective CA facilitation of acid/base transporters in the ventricular myocyte, implying a specific role for the intracellular enzyme in HCO3- transport, and hence pHi regulation in the heart.


Assuntos
Bicarbonatos/metabolismo , Anidrases Carbônicas/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Sódio/metabolismo , Animais , Bicarbonatos/química , Células Cultivadas , Ativação Enzimática , Ventrículos do Coração/citologia , Concentração de Íons de Hidrogênio , Masculino , Ratos , Ratos Sprague-Dawley
14.
Pflugers Arch ; 466(3): 529-40, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23989646

RESUMO

Atrial fibrillation (AF) is the most common arrhythmia. Gain-of-function mutations in KCNQ1, the pore-forming α-subunit of the slow delayed rectifier K current (IKs) channel, have been associated with AF. The purpose of this study was functional assessment of a mutation in KCNQ1 identified in a family with persistent AF and sinus bradycardia. We investigated whether this KCNQ1 missense mutation could form the genetic basis for AF and bradycardia simultaneously in this family. Sanger sequencing in a family with hereditary persistent AF identified a novel KCNQ1 variant (V241F) in a highly conserved region of S4 domain. The proband and her son developed bradycardia and persistent AF in an age-dependent fashion. The other son was a mutation carrier but he showed sinus bradycardia and not AF. Whole-cell patch clamp electrophysiology showed that V241F mutation in KCNQ1 shifted the activation curve to the left and dramatically slowed deactivation, leading to a constitutively open-like phenotype. Computer modeling showed that V241F would slow pacemaker activity. Also, simulations of atrial excitation predicted that V241F results in extreme shortening of action potential duration, possibly resulting in AF. Our study indicates that V241F might cause sinus bradycardia by increasing IKs. Additionally, V241F likely shortens atrial refractoriness to promote a substrate for reentry. KCNQ1 mutations have previously been described in AF, yet this is the first time a mutation in KCNQ1 is associated with age-dependent bradycardia and persistent AF. This finding further supports the hypothesis that sinus node dysfunction contributes to the development of AF.


Assuntos
Potenciais de Ação , Fibrilação Atrial/fisiopatologia , Bradicardia/fisiopatologia , Canal de Potássio KCNQ1/metabolismo , Mutação de Sentido Incorreto , Adulto , Fatores Etários , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/genética , Bradicardia/diagnóstico , Bradicardia/genética , Feminino , Células HEK293 , Heterozigoto , Humanos , Canal de Potássio KCNQ1/genética , Masculino , Pessoa de Meia-Idade , Modelos Cardiovasculares , Linhagem
15.
J Mol Cell Cardiol ; 61: 51-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23602948

RESUMO

Acid extrusion on Na(+)-coupled pH-regulatory proteins (pH-transporters), Na(+)/H(+) exchange (NHE1) and Na(+)-HCO3(-) co-transport (NBC), drives Na(+) influx into the ventricular myocyte. This H(+)-activated Na(+)-influx is acutely up-regulated at pHi<7.2, greatly exceeding Na(+)-efflux on the Na(+)/K(+) ATPase. It is spatially heterogeneous, due to the co-localisation of NHE1 protein (the dominant pH-transporter) with gap-junctions at intercalated discs. Overall Na(+)-influx via NBC is considerably lower, but much is co-localised with L-type Ca(2+)-channels in transverse-tubules. Through a functional coupling with Na(+)/Ca(2+) exchange (NCX), H(+)-activated Na(+)-influx increases sarcoplasmic-reticular Ca(2+)-loading and release during intracellular acidosis. This raises Ca(2+)-transient amplitude, rescuing it from direct H(+)-inhibition. Functional coupling is biochemically regulated and linked to membrane receptors, through effects on NHE1 and NBC. It requires adequate cytoplasmic Na(+)-mobility, as NHE1 and NCX are spatially separated (up to 60µm). The relevant functional NCX activity must be close to dyads, as it exerts no effect on bulk diastolic Ca(2+). H(+)-activated Na(+)-influx is up-regulated during ischaemia-reperfusion and some forms of maladaptive hypertrophy and heart failure. It is thus an attractive system for therapeutic manipulation. This article is part of a Special Issue entitled "Na(+) Regulation in Cardiac Myocytes".


Assuntos
Desequilíbrio Ácido-Base/metabolismo , Proteínas de Transporte de Cátions/fisiologia , Acoplamento Excitação-Contração , Trocadores de Sódio-Hidrogênio/fisiologia , Animais , Cardiomegalia/metabolismo , Insuficiência Cardíaca/metabolismo , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Humanos , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/fisiologia , Prótons , Trocador 1 de Sódio-Hidrogênio
16.
Pflugers Arch ; 465(8): 1121-34, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23440458

RESUMO

Human ether-á-go-go-related gene (hERG) K(+) channel current (I hERG ) is inhibited by various compounds and genetic mutations, potentially resulting in cardiac arrhythmia. Here, we investigated effects of caffeic acid phenethyl ester (CAPE) and curcumin, two natural anti-inflammatory polyphenols, on I hERG in HEK-293 cells overexpressed with hERG. CAPE dose-dependently decreased repolarization tail current of hERG (I hERG,tail; IC50, 10.6 ± 0.5 µM). CAPE also shifted half-activation voltage (V 1/2) to the left (from -17.5 to -26.5 mV) and accelerated activation and inactivation kinetics. The CAPE inhibition of I hERG,tail was not attenuated in the pore-blocker site mutants of hERG (Y652A and F656A). A point mutation of Cys723 (C723S) mimicked the effects of CAPE and caused a left shift of V 1/2 and acceleration of I hERG,tail deactivation. However, I hERG,tail inhibition by CAPE was still observed in C723S. Taken together, CAPE inhibits hERG channel by class 3 mechanism, i.e., modification of gating, not by blocking the pore. Curcumin induced changes of I hERG similar to those of CAPE, while additional interaction with pore-blocking sites was suggested from attenuated I hERG,tail inhibition in Y652A and F656A. Interestingly, I hERG induced by human action potential voltage clamp was increased by CAPE while decreased by curcumin. Mathematical simulation of action potential derived from the experimental results of CAPE and curcumin supports that CAPE, but not curcumin, would induce shortening of AP duration by facilitation of I hERG . The above results revealed intriguing roles of Cys723 in hERG kinetics and suggested that conventional drug screening by using step pulse protocol for I hERG,tail would overlook the hERG kinetic modulations that could compensate the decrease of I hERG,tail.


Assuntos
Ácidos Cafeicos/farmacologia , Curcumina/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/metabolismo , Álcool Feniletílico/análogos & derivados , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Potenciais de Ação/efeitos dos fármacos , Canal de Potássio ERG1 , Células HEK293 , Humanos , Cinética , Álcool Feniletílico/farmacologia
17.
Korean J Physiol Pharmacol ; 17(6): 537-46, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24381504

RESUMO

Deiters' cells are the supporting cells in organ of Corti and are suggested to play an important role in biochemical and mechanical modulation of outer hair cells. We successfully isolated functionally different K(+) currents from Deiters' cells of guinea pig using whole cell patch clamp technique. With high K(+) pipette solution, depolarizing step pulses activated strongly outward rectifying currents which were dose-dependently blocked by clofilium, a class III anti-arrhythmic K(+) channel blocker. The remaining outward current was transient in time course whereas the clofilium-sensitive outward current showed slow inactivation and delayed rectification. Addition of 5 mM tetraethylammonium (TEA) further blocked the remaining current leaving a very fast inactivating transient outward current. Therefore, at least three different types of K(+) current were identified in Deiters' cells, such as fast activating and fast inactivating current, fast activating slow inactivating current, and very fast inactivating transient outward current. Physiological role of them needs to be established.

18.
J Physiol ; 590(18): 4447-63, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22674726

RESUMO

L-type Ca(2+) channels (ICaLs) are inactivated by an increase in intracellular [Ca(2+)], known as Ca(2+)-dependent inactivation (CDI). CDI is also induced by Ca(2+) released from the sarcoplasmic reticulum (SR), known as release-dependent inhibition (RDI). As both CDI and RDI occur in the junctional subsarcolemmal nanospace (JSS), we investigated which factors are involved within the JSS using isolated cardiac myocytes from the main pulmonary vein of the rabbit. Using the whole-cell patch clamp technique, RDI was readily observed with the application of a pre-pulse followed by a test pulse, during which the ICaLs exhibited a decrease in peak current amplitude and a slower inactivation. A fast acting Ca(2+) chelator, 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA), abolished this effect. As the time interval between the pre-pulse and test pulse increased, the ICaLs exhibited greater recovery and the RDI was relieved. Inhibition of the ryanodine receptor (RyR) or the SR Ca(2+)-ATPase (SERCA) greatly attenuated RDI and facilitated ICaL recovery. Removal of extracellular Na(+),which inhibits the Na(+)-Ca(2+) exchange (Incx), greatly enhanced RDI and slowed ICaL recovery, suggesting that Incx critically controls the [Ca(2+)] in the JSS. We incorporated the Ca(2+)-binding kinetics of the ICaL into a previously published computational model. By assuming two Ca(2+)-binding sites in the ICaL, of which one is of low-affinity with fast kinetics and the other is of high-affinity with slower kinetics, the new model was able to successfully reproduce RDI and its regulation by Incx. The model suggests that Incx accelerates Ca(2+) removal from the JSS to downregulate CDI and attenuates SR Ca(2+) refilling. The model may be useful to elucidate complex mechanisms involved in excitation­contraction coupling in myocytes.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Cálcio/fisiologia , Modelos Cardiovasculares , Miócitos Cardíacos/fisiologia , Veias Pulmonares/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Técnicas In Vitro , Miócitos Cardíacos/efeitos dos fármacos , Coelhos , Retículo Sarcoplasmático/fisiologia , Trocador de Sódio e Cálcio/fisiologia
19.
Pflugers Arch ; 464(6): 549-59, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23001133

RESUMO

Glucocorticoids (GCs) are essential steroid hormones for homeostasis, development, metabolism, and cognition and possess anti-inflammatory and immunosuppressive actions. Since glucocorticoid receptor II (GR) is nearly ubiquitous, chronic activation or depletion of GCs leads to dysfunction of diverse organs, including the heart and blood vessels, resulting predominantly from changes in gene expression. Most studies, therefore, have focused on the genomic effects of GC to understand its related pathophysiological manifestations. The nongenomic effects of GCs clearly differ from well-known genomic effects, with the former responding within several minutes without the need for protein synthesis. There is increasing evidence that the nongenomic actions of GCs influence various physiological functions. To develop a GC-mediated therapeutic target for the treatment of cardiovascular disease, understanding the genomic and nongenomic effects of GC on the cardiovascular system is needed. This article reviews our current understanding of the underlying mechanisms of GCs on cardiovascular diseases and stress, as well as how nongenomic GC signaling contributes to these conditions. We suggest that manipulation of GC action based on both GC and GR metabolism, mitochondrial impact, and the action of serum- and glucocorticoid-dependent kinase 1 may provide new information with which to treat cardiovascular diseases.


Assuntos
Sistema Cardiovascular/metabolismo , Glucocorticoides/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Genômica/métodos , Humanos
20.
Pflugers Arch ; 464(6): 631-43, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23073892

RESUMO

Telmisartan is an angiotensin II receptor blocker and partial peroxisome proliferator-activated receptor gamma agonist that modulates the renin-angiotensin-aldosterone system. It is used primarily to manage hypertension, diabetic nephropathy, and congestive heart failure. Recent studies have reported that myocardial infarction (MI) has occurred in telmisartan-treated patients. The purpose of the study was to investigate the specific conditions and underlying mechanisms that may result in telmisartan-induced MI. We evaluated the effect of telmisartan on whole hearts, cardiomyocytes, and cardiac sarcolemmal ion channels. Hearts of 8-week-old male Sprague-Dawley rats were perfused with 3, 10, 30, or 100 µM telmisartan or losartan or with normal Tyrode's solution (control) for 3 h. We found that telmisartan induced myocardial infarction, with an infarct size of 21 % of the total at 30 µM (P < 0.0001) and 63 % of the total area at 100 µM (P < 0.001). Telmisartan also induced cardiac dysfunction (e.g., decreased heart rate, diminished coronary flow, hypercontracture, and arrhythmia). Confocal microscopy demonstrated that 30 µM telmisartan significantly elevated the intracellular Ca(2+) level, leading to hypercontracture and cell death. Patch clamp analysis of isolated cardiomyocytes revealed that telmisartan induced Na(+) overload by slowing the inactivation of voltage-gated Na(+) current (I (Na)), activating the reverse mode of Na(+)-Ca(2+) exchanger activity, and causing Ca(2+) overload. Telmisartan significantly delayed the inactivation of the voltage-gated Na(+) channel, causing cytosolic Na(+) overload, prolonged action potential duration, and subsequent Ca(2+) overload. Above 30 µM, telmisartan may potentially cause cardiac cell death and MI.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/toxicidade , Benzimidazóis/toxicidade , Benzoatos/toxicidade , Coração/efeitos dos fármacos , Infarto do Miocárdio/induzido quimicamente , Miócitos Cardíacos/efeitos dos fármacos , PPAR gama/agonistas , Canais de Sódio Disparados por Voltagem/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Coração/fisiopatologia , Losartan/farmacologia , Masculino , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , PPAR gama/metabolismo , Ratos , Ratos Sprague-Dawley , Sarcolema/efeitos dos fármacos , Sarcolema/metabolismo , Sarcolema/patologia , Sódio/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Telmisartan
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA