Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Circ Res ; 112(4): 601-5, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23307820

RESUMO

RATIONALE: During the transition from compensated hypertrophy to heart failure, the signaling between L-type Ca(2+) channels in the cell membrane/T-tubules and ryanodine receptors in the sarcoplasmic reticulum becomes defective, partially because of the decreased expression of a T-tubule-sarcoplasmic reticulum anchoring protein, junctophilin-2. MicroRNA (miR)-24, a junctophilin-2 suppressing miR, is upregulated in hypertrophied and failing cardiomyocytes. OBJECTIVE: To test whether miR-24 suppression can protect the structural and functional integrity of L-type Ca(2+) channel-ryanodine receptor signaling in hypertrophied cardiomyocytes. METHODS AND RESULTS: In vivo silencing of miR-24 by a specific antagomir in an aorta-constricted mouse model effectively prevented the degradation of heart contraction, but not ventricular hypertrophy. Electrophysiology and confocal imaging studies showed that antagomir treatment prevented the decreases in L-type Ca(2+) channel-ryanodine receptor signaling fidelity/efficiency and whole-cell Ca(2+) transients. Further studies showed that antagomir treatment stabilized junctophilin-2 expression and protected the ultrastructure of T-tubule-sarcoplasmic reticulum junctions from disruption. CONCLUSIONS: MiR-24 suppression prevented the transition from compensated hypertrophy to decompensated hypertrophy, providing a potential strategy for early treatment against heart failure.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Acoplamento Excitação-Contração/efeitos dos fármacos , Insuficiência Cardíaca/prevenção & controle , Hipertrofia Ventricular Esquerda/tratamento farmacológico , MicroRNAs/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Oligonucleotídeos Antissenso/uso terapêutico , Animais , Estenose Aórtica Subvalvar/complicações , Canais de Cálcio Tipo L/fisiologia , Sinalização do Cálcio/fisiologia , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Hipertrofia Ventricular Esquerda/complicações , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/fisiologia , Modelos Cardiovasculares , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Oligonucleotídeos Antissenso/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/fisiologia , Retículo Sarcoplasmático/ultraestrutura
2.
Circ Res ; 111(7): 837-41, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22891046

RESUMO

RATIONALE: Failing cardiomyocytes exhibit decreased efficiency of excitation-contraction (E-C) coupling. The downregulation of junctophilin-2 (JP2), a protein anchoring the sarcoplasmic reticulum to T-tubules, has been identified as a major mechanism underlying the defective E-C coupling. However, the regulatory mechanism of JP2 remains unknown. OBJECTIVE: To determine whether microRNAs regulate JP2 expression. METHODS AND RESULTS: Bioinformatic analysis predicted 2 potential binding sites of miR-24 in the 3'-untranslated regions of JP2 mRNA. Luciferase assays confirmed that miR-24 suppressed JP2 expression by binding to either of these sites. In the aortic stenosis model, miR-24 was upregulated in failing cardiomyocytes. Adenovirus-directed overexpression of miR-24 in cardiomyocytes decreased JP2 expression and reduced Ca(2+) transient amplitude and E-C coupling gain. CONCLUSIONS: MiR-24-mediated suppression of JP2 expression provides a novel molecular mechanism for E-C coupling regulation in heart cells and suggests a new target against heart failure.


Assuntos
Estenose da Valva Aórtica/metabolismo , Insuficiência Cardíaca/metabolismo , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Regulação para Cima , Animais , Estenose da Valva Aórtica/patologia , Cálcio/metabolismo , Células Cultivadas , Biologia Computacional , Acoplamento Excitação-Contração/fisiologia , Insuficiência Cardíaca/patologia , Proteínas de Membrana/genética , MicroRNAs/genética , Modelos Animais , Miócitos Cardíacos/patologia , RNA Mensageiro/metabolismo , Ratos , Retículo Sarcoplasmático/fisiologia
3.
Acta Pharmacol Sin ; 32(9): 1116-27, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21841810

RESUMO

AIM: To study and establish a proteome reference map and regulation network of neonatal rat cardiomyocyte. METHODS: Cultured cardiomyocytes of neonatal rats were used. All proteins expressed in the cardiomyocytes were separated and identified by two-dimensional polyacrylamide gel electrophoresis (2-DE) and matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF MS). Biological networks and pathways of the neonatal rat cardiomyocytes were analyzed using the Ingenuity Pathway Analysis (IPA) program (www.ingenuity.com). A 2-DE database was made accessible on-line by Make2ddb package on a web server. RESULTS: More than 1000 proteins were separated on 2D gels, and 148 proteins were identified. The identified proteins were used for the construction of an extensible markup language-based database. Biological networks and pathways were constructed to analyze the functions associate with cardiomyocyte proteins in the database. The 2-DE database of rat cardiomyocyte proteins can be accessed at http://2d.bjmu.edu.cn. CONCLUSION: A proteome reference map and regulation network of the neonatal rat cardiomyocytes have been established, which may serve as an international platform for storage, analysis and visualization of cardiomyocyte proteomic data.


Assuntos
Eletroforese em Gel Bidimensional/métodos , Miócitos Cardíacos/química , Proteoma/análise , Proteômica/métodos , Animais , Células Cultivadas , Masculino , Redes e Vias Metabólicas , Proteoma/isolamento & purificação , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos
4.
PLoS Biol ; 5(2): e21, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17214508

RESUMO

Pressure overload-induced hypertrophy is a key step leading to heart failure. The Ca(2+)-induced Ca(2+) release (CICR) process that governs cardiac contractility is defective in hypertrophy/heart failure, but the molecular mechanisms remain elusive. To examine the intermolecular aspects of CICR during hypertrophy, we utilized loose-patch confocal imaging to visualize the signaling between a single L-type Ca(2+) channel (LCC) and ryanodine receptors (RyRs) in aortic stenosis rat models of compensated (CHT) and decompensated (DHT) hypertrophy. We found that the LCC-RyR intermolecular coupling showed a 49% prolongation in coupling latency, a 47% decrease in chance of hit, and a 72% increase in chance of miss in DHT, demonstrating a state of "intermolecular failure." Unexpectedly, these modifications also occurred robustly in CHT due at least partially to decreased expression of junctophilin, indicating that intermolecular failure occurs prior to cellular manifestations. As a result, cell-wide Ca(2+) release, visualized as "Ca(2+) spikes," became desynchronized, which contrasted sharply with unaltered spike integrals and whole-cell Ca(2+) transients in CHT. These data suggested that, within a certain limit, termed the "stability margin," mild intermolecular failure does not damage the cellular integrity of excitation-contraction coupling. Only when the modification steps beyond the stability margin does global failure occur. The discovery of "hidden" intermolecular failure in CHT has important clinical implications.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/fisiologia , Insuficiência Cardíaca/metabolismo , Hipertrofia Ventricular Esquerda/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Aorta/cirurgia , Estenose da Valva Aórtica/metabolismo , Estenose da Valva Aórtica/patologia , Modelos Animais de Doenças , Insuficiência Cardíaca/patologia , Hipertrofia Ventricular Esquerda/patologia , Microscopia Confocal , Contração Miocárdica/fisiologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
5.
Cell Calcium ; 43(2): 165-74, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17583790

RESUMO

Dynamic nuclear Ca(2+) signals play pivotal roles in diverse cellular functions including gene transcription, cell growth, differentiation, and apoptosis. Here we report a novel nuclear Ca(2+) regulatory mechanism mediated by inositol 1,4,5-trisphosphate receptors (IP(3)Rs) around the nucleus in developing cardiac myocytes. Activation of IP(3)Rs by alpha(1)-adrenergic receptor (alpha(1)AR) stimulation or by IP(3) application (in saponin-permeabilized cells) increases Ca(2+) spark frequency preferentially in the region around the nucleus in neonatal rat ventricular myocytes. A nuclear enrichment of IP(3)R distribution supports the higher responsiveness of Ca(2+) release in this particular region. Strikingly, we observed "nuclear Ca(2+)waves" that engulf the entire nucleus without spreading into the bulk cytosol. alpha(1)AR stimulation enhances the occurrence of nuclear Ca(2+) waves and confers them the ability to trigger cytosolic Ca(2+) waves via IP(3)R-dependent pathways. This finding accounts, at least partly, for a profound frequency-dependent modulation of global Ca(2+) oscillations during alpha(1)AR stimulation. Thus, IP(3)R-mediated Ca(2+) waves traveling in the nuclear region provide active, autonomous regulation of nuclear Ca(2+) signaling, which provides for not only the local signal transduction, but also a pacemaker to drive global Ca(2+) transient in the context of alpha(1)AR stimulation in developing cardiac myocytes.


Assuntos
Sinalização do Cálcio/fisiologia , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Miócitos Cardíacos/fisiologia , Membrana Nuclear/fisiologia , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Animais Recém-Nascidos , Permeabilidade da Membrana Celular , Receptores de Inositol 1,4,5-Trifosfato/efeitos dos fármacos , Microscopia Confocal , Fenilefrina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1/efeitos dos fármacos , Receptores Adrenérgicos alfa 1/fisiologia
6.
FEBS J ; 275(14): 3590-7, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18537819

RESUMO

beta-Adrenergic receptor (beta-AR)-induced cardiac remodeling is closely linked with the re-expression of the atrial natriuretic factor (ANF) gene. However, the exact molecular mechanism of this response remains elusive. Here, we demonstrate that the beta-AR agonist isoproterenol potently evokes the tyrosine phosphorylation of STAT3 and increases its transcriptional activity in an extracellularly regulated kinase 1/2 and glycoprotein (gp)130 signaling-dependent manner in rat cardiomyocytes. Interestingly, both specific silencing of signal transducers and activators of transcription 3 (STAT3) expression by lentivirus-mediated RNA interference and antagonism of gp130 signaling lead to significant inhibition of isoproterenol-stimulated ANF expression. Together, these results indicate that gp130/STAT3 signaling has an essential role in ANF expression by beta-AR stimulation.


Assuntos
Fator Natriurético Atrial/biossíntese , Receptor gp130 de Citocina/metabolismo , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Fator de Transcrição STAT3/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Fator Natriurético Atrial/genética , Células Cultivadas , Citocinas/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Expressão Gênica , Isoproterenol/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação , Ratos , Fator de Transcrição STAT3/química , Transdução de Sinais , Transcrição Gênica , Tirosina/metabolismo
7.
Trends Pharmacol Sci ; 27(6): 330-7, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16697055

RESUMO

Recent studies of adrenoceptors have revealed subtype-specific signaling, promiscuous G-protein coupling, time-dependent switching of intracellular signaling pathways, intermolecular interactions within or between adrenoceptor subfamilies, and G-protein-independent signaling pathways. These findings have extended the classical linear paradigm of G-protein-coupled receptor signaling to a complex "signalome" in which an individual adrenoceptor initiates multiple signaling pathways in a temporally and spatially regulated manner. In particular, persistent stimulation of beta-adrenoceptor subtypes causes a time-dependent switch of signaling pathways and elicits different, even opposing, functional roles of these receptors in regulating cardiac structure and function. Recent progress in the understanding of subtype-specific functions and signaling mechanisms of cardiac adrenoceptor subtypes, particularly beta(1)-adrenoceptors, beta(2)-adrenoceptors, alpha(1A)-adrenoceptors and alpha(1B)-adrenoceptors, might have important pathogenic and therapeutic implications for heart disease.


Assuntos
Miocárdio/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Animais , Humanos , Miocárdio/enzimologia , Miócitos Cardíacos/metabolismo , Receptor Cross-Talk
8.
Biophys Chem ; 127(3): 149-54, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17306438

RESUMO

The heterogeneous motion of alpha(1B)-adrenoceptor (alpha(1B)-AR) was visualized in living cells with BODIPY-labeled antagonist of AR by single molecule fluorescence microscopy at high spatial resolution. The moving trajectory was reconstructed by precise localization (better than 20 nm) with a least-square fit of a two-dimensional Gaussian point spread function to each single spot. Trajectory analysis revealed two apparent groups of movements: directed motion and hindered motion. The directed motion had speeds higher than 0.1 mum/s. The histogram of diffusion coefficients of the hindered motion showed distinction between the cell membrane and the cytoplasm: the diffusion coefficient was lower near the cell membrane than in the internal cytoplasm, suggesting that alpha(1B)-AR was located or trapped in different networks, which was consistent with the natural distribution of cytoskeleton in living cells. These results suggested that the heterogeneity in the motion of alpha(1B)-AR in living cell might be associated with different localizations of cell skeleton proteins in the cell, which could provide molecular insight of AR regulation in living cells.


Assuntos
Membrana Celular/metabolismo , Citoplasma/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Sítios de Ligação , Compostos de Boro , Células Cultivadas , Difusão , Humanos , Rim/citologia , Modelos Biológicos , Prazosina , Receptores Adrenérgicos , Distribuição Tecidual
9.
Sheng Li Xue Bao ; 59(2): 175-82, 2007 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-17437040

RESUMO

To test the hypothesis that AMP-activated protein kinase (AMPK) is possibly the downstream signaling molecule of certain subtypes of adrenergic receptor (AR) in the heart, we evaluated AMPK activation mediated by ARs in H9C2 cells, a rat cardiac source cell line, and rat hearts. The AMPK-alpha subunit and the phosphorylation level of Thr(172)-AMPK-alpha subunit were subjected to Western blot analysis. Osmotic minipumps filled with norepinephrine (NE), phenylephrine (PE) or vehicle [0.01% (W/V) vitamin C solution] were implanted into male Sprague-Dawley rats subcutaneously. The pumps delivered NE or PE continuously at the rate of 0.2 mg/kg per hour. After 7-day infusion, the activity of AMPK was examined following immunoprecipitation with anti-AMPK-alpha antibody. At the cellular level, we found that NE elevated AMPK phosphorylation level in a dose- and time-dependent manner, with the maximal effect at 10 micromol/L NE after 10-minute treatment. This effect was insensitive to propranolol, a specific beta-AR antagonist, but abolished by prazosin, an alpha(1)-AR antagonist, suggesting that alpha(1)-AR but not beta-AR mediated the phosphorylation of AMPK. Moreover, the results from rat models of 7-day-infusion of AR agonists demonstrated that the activity of AMPK was significantly higher in NE (7.4-fold) and PE (6.0-fold) infusion groups than that in the vehicle group (P<0.05, n=6). On the other hand, no obvious cardiac hypertrophy and tissue fibrosis changes were observed in PE-infused rats. Taken together, our results demonstrate that alpha(1)-AR stimulation enhances the activity of AMPK, indicating an important role of alpha(1)-AR stimulation in the regulation of AMPK in the heart. Understanding the activation of AMPK mediated by alpha(1)-AR might have clinical implications in the therapy of heart failure.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Miocárdio/metabolismo , Receptores Adrenérgicos alfa/fisiologia , Animais , Linhagem Celular , Ventrículos do Coração , Masculino , Miocárdio/citologia , Norepinefrina/farmacologia , Fenilefrina/farmacologia , Fosforilação , Ratos , Ratos Sprague-Dawley
11.
Pharmacol Ther ; 108(3): 257-68, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15979723

RESUMO

The stimulation of beta-adrenergic receptor (betaAR) plays a pivotal role in regulating myocardial function and morphology in the normal and failing heart. Three genetically and pharmacologically distinct betaAR subtypes, beta1AR, beta2AR, and beta3AR, are identified in various types of cells. While both beta1AR and beta2AR, the predominant betaAR subtypes expressed in the heart of many mammalian species including human, are coupled to the Gs-adenylyl cyclase-cAMP-PKA pathway, beta2AR dually activates pertussis toxin-sensitive Gi proteins. During acute stimulation, beta2AR-Gi coupling partially inhibits the Gs-mediated positive contractile and relaxant effects via a Gi-Gbetagamma-phosphoinositide 3-kinase (PI3K)-dependent mechanism in adult rodent cardiomyocytes. More importantly, persistent beta1AR stimulation evokes a multitude of cardiac toxic effects, including myocyte apoptosis and hypertrophy, via a calmodulin-dependent protein kinase II (CaMKII)-, rather than cAMP-PKA-, dependent mechanism in rodent heart in vivo and cultured cardiomyocytes. In contrast, persistent beta2AR activation protects myocardium by a cell survival pathway involving Gi, PI3K, and Akt. In this review, we attempt to highlight the distinct functionalities and signaling mechanisms of these betaAR subtypes and discuss how these subtype-specific properties of betaARs might affect the pathogenesis of congestive heart failure (CHF) and the therapeutic effectiveness of certain beta-blockers in the treatment of congestive heart failure.


Assuntos
Insuficiência Cardíaca/etiologia , Receptores Adrenérgicos beta/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Coração/fisiologia , Insuficiência Cardíaca/metabolismo , Humanos , Miocárdio/metabolismo , Transdução de Sinais
12.
FASEB J ; 19(1): 109-11, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15489248

RESUMO

Activation of p38 mitogen-activated protein kinase (MAPK) plays a central role in cellular responses to a multitude of stress signals. In the heart, enhanced p38 MAPK signaling has been implicated in cardiac hypoxic and ischemic injury. However, the mechanism underlying hypoxia-induced p38 MAPK activation remains elusive. We investigated p38 MAPK activation during hypoxia in adult rat cardiomyocytes. Here, we reported that hypoxia leads to concurrent intracellular acidosis and activation of p38 MAPK and that the hypoxia-induced p38 MAPK signaling can be fully abolished by neutralizing intracellular pH, whereas intracellular acidosis (intracellular pH<7.0) per se overtly augments activation of p38 MAPK but not ERK1/2 and JNK. Furthermore, inhibition of p38 MAPK protects myocytes against hypoxic cell death, suggesting that acidosis-evoked p38 MAPK signaling plays an important role in hypoxic cell injury and cell death. These results demonstrate, for the first time, that intracellular acidosis constitutes a necessary and sufficient link responsible for hypoxia-activated p38 MAPK signaling and the subsequent hypoxic cardiomyocyte injury and death.


Assuntos
Acidose/enzimologia , Hipóxia/etnologia , Miócitos Cardíacos/química , Miócitos Cardíacos/enzimologia , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Acidose/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Ativação Enzimática/fisiologia , Ventrículos do Coração/citologia , Concentração de Íons de Hidrogênio , MAP Quinase Quinase 3/metabolismo , MAP Quinase Quinase 6/metabolismo , Miócitos Cardíacos/patologia , Fosforilação , Ratos
13.
Med Rev (Berl) ; 1(1): 1, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37724077
14.
Eur J Pharmacol ; 509(2-3): 109-15, 2005 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-15733545

RESUMO

The potential role of endogenous ryanodine receptor (RyR) in modulating Ca2+ handling in HEK293 cells is controversial. Using Fura2/AM, here we provide evidence that caffeine can induce Ca2+ release from inositol 1,4,5-trisphosphate receptor-sensitive stores and Ca2+ entry in early passage numbers of HEK293 cells, but not in late passage ones. Ryanodine blocks caffeine-mediated effect, whereas 4-chloro-m-cresol can mimic these effects. In contrast, an increase in cyclic AMP or activation of voltage-dependent Ca2+ channels does not induce detectable alteration in intracellular Ca2+. Importantly, immunoblotting and staining have revealed that endogenous RyR expression is more abundant in the early than in the late passage cells. Additionally, similar to carbachol, Ca2+ entry in response to caffeine is blocked by capacitative Ca2+ entry inhibitors. These results indicate that the endogenous RyR in HEK293 cells can function as Ca2+ release channels and mediate capacitative Ca2+ entry, but they may be reduced due to cell passage.


Assuntos
Cafeína/farmacologia , Cálcio/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Compostos de Boro/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Carbacol/farmacologia , Linhagem Celular , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Gadolínio/farmacologia , Inositol 1,4,5-Trifosfato/metabolismo , Rim/citologia , Rim/embriologia , Rim/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
15.
Prostaglandins Other Lipid Mediat ; 75(1-4): 141-51, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15789621

RESUMO

Arachidonic acid (AA) modulates intracellular Ca2+ signaling via Ca2+ release or/and Ca2+ entry. However, the mechanism underlies either process is unknown; nor is it clear as to whether the two processes are mechanistically linked. By using Fura2/AM, we found that AA induced mobilization of internal Ca2+ store and an increment in Ca2+, Mn2+ and Ba2+ influx in HEK293 cells. The AA-mediated Ca2+ signaling was not due to AA metabolites, and insensitive to capacitative Ca2+ entry inhibitors. Interestingly, isotetrandrine and Gd3+ inhibited both AA-induced Ca2+ release and Ca2+ entry in a concentration-dependent manner without affecting Ca2+ discharge caused by carbachol, caffeine, or thapsigargin. Additionally, similar pattern of inhibition was observed with tetracaine treatment. More importantly, the three compounds exhibited almost equal potent inhibition of AA-initiated Ca2+ release as well as Ca2+ influx. Therefore, this study, for the first time, provides evidence for a direct coupling between AA-mediated Ca2+ release and Ca2+ entry.


Assuntos
Ácido Araquidônico/farmacologia , Sinalização do Cálcio/fisiologia , Transporte Biológico/efeitos dos fármacos , Cafeína/farmacologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Carbacol/farmacologia , Cátions Bivalentes/metabolismo , Linhagem Celular , Gadolínio/farmacologia , Humanos , Rim , Cinética , Tapsigargina/farmacologia
16.
Sheng Li Xue Bao ; 57(4): 480-5, 2005 Aug 25.
Artigo em Chinês | MEDLINE | ID: mdl-16094496

RESUMO

To investigate the subcellular distribution of three alpha(1)-adrenergic receptor subtypes and their internalization and trafficking upon agonist stimulation in human embryonic kidney (HEK) 293A cell line, saturation radioligand binding assay, laser confocal imaging, and Western blot were applied to examine the distribution and changes in localization of three alpha(1)-AR subtypes in transfected HEK 293A cells prior to and after treatment with phenylephrine. The results are as follows: (1) The transfection efficiency was over 90%and was equal among three alpha(1)-AR subtypes. alpha(1B ) -AR expression in cell membrane was the highest, and alpha(1D ) -AR was the lowest, as determined by (125)I-BE2254 binding assay, however, K(d)s were not significantly different among the three receptor subtypes. (2) Without agonist stimulation, alpha(1A ) -AR was detected not only on the cell surface but also in the cytosol, alpha(1B ) -AR was predominantly located on the cell surface, whereas alpha(1D ) -AR was mostly detected in the cytosol. (3) After 1 h of stimulation with phenylephrine, as observed using confocal microscope, less alpha(1A)- and alpha(1B ) -AR were detected on the cell surface but more in the cytosol. The change was more remarkable in alpha(1B)-AR than that in alpha(1A)-AR, whereas no change of distribution was detected in alpha(1D)-AR in response to phenylephrine. However, when examined by Western blot, no change in distribution was detected in alpha(1A)- and alpha(1D)-AR, only alpha(1B)-AR showed the same change as that shown in confocal imaging. It is suggested that the characteristics of localization and changes of distribution are different among three alpha(1)-AR subtypes in HEK293A cells upon phenylephrine stimulation.

18.
Sci China C Life Sci ; 47(4): 376-81, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15493479

RESUMO

To improve selectivity and specificity of cell membrane chromatography (CMC), the chromatography affinities of nine ligands of alpha1-adrenergic receptor(AR)to alpha1D-AR subtype were investigated. The human embryonic kidney (HEK) 293 cells expressed by cDNA of alpha1D-AR subtypes were cultured and cell membrane stationary phase (CMSP) was prepared. Then the interactions between ligands and alpha1D-AR in CMSP were investigated using CMC. The affinity rank order to alpha1D-AR subtype obtained from CMC for the nine alpha1-adrenoceceptor ligands is: prazosin, BMY7378, phentolamine, oxymetazoline, 5-methylurapidil, norepinephrine, phenylephrine, methoxamine, RS-17053. The affinity rank order is similar and correlates well with that obtained from others' radioligand binding assays (RBA). CMSP prepared by transfected HEK293 cells with alpha1-D-adrenoceptor cDNA and CMC method could be used to evaluate affinities of -drug-receptor and drug-receptor subtypes and to screen drugs selective to alpha1D-AR.


Assuntos
Membrana Celular/metabolismo , Receptores Adrenérgicos alfa 1/classificação , Receptores Adrenérgicos alfa 1/metabolismo , Linhagem Celular , Cromatografia de Afinidade , Avaliação Pré-Clínica de Medicamentos , Expressão Gênica , Humanos , Ligantes , Receptores Adrenérgicos alfa 1/genética , Transfecção
19.
Sheng Li Xue Bao ; 56(1): 1-15, 2004 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-14985822

RESUMO

In the heart, stimulation of beta-adrenergic receptors (betaAR) serves as the most powerful means to increase cardiac contractility and relaxation in response to stress or a "fight-or-flight" situation. However, sustained beta-adrenergic stimulation promotes pathological cardiac remodeling such as myocyte hypertrophy, apoptosis and necrosis, thus contributing to the pathogenesis of chronic heart failure. Over the past decade, compelling evidence has demonstrated that coexisting cardiac betaAR subtypes, mainly beta(1)AR and beta (2)AR, activate markedly different signaling cascades. As a result, acute beta(1)AR stimulation activates the G(s) -adenylyl cyclase-cAMP-PKA signaling that can broadcast throughout the cell, whereas beta(2)AR-evoked cAMP signaling is spatially and functionally compartmentalized, due to concurrent G(i) activation. Chronic stimulation of beta(1)AR and beta(2)AR elicits opposing effects on the fate of cardiomyocytes: beta(1)AR induces hypertrophy and apoptosis; but beta(2)AR promotes cell survival. The cardiac protective effect of beta(2)AR is mediated by a signaling pathway sequentially involving G(i), G(betagamma), PI3K and Akt. Unexpectedly, beta(1)AR-induced myocyte hypertrophy and apoptosis are independent of the classic cAMP/PKA pathway, but require activation of Ca(2+)/calmodulin-dependent kinase II (CaMK II). The outcomes of cardiac-specific transgenic overexpression of either beta AR subtype in mice have reinforced the fundamentally different functional roles of these betaAR subtypes in governing cardiac remodeling and performance. These new insights regarding betaAR subtype stimulation not only provide clues as to cellular and molecular mechanisms underlying the beneficial effects of beta AR blockers in patients with chronic heart failure, but also delineate rationale for combining selective beta(1)AR blockade with moderate beta(2)AR activation as a potential novel therapy for the treatment of chronic heart failure.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Miocárdio/metabolismo , Receptores Adrenérgicos beta/fisiologia , Adenilil Ciclases/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Coração/fisiologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Receptores Adrenérgicos beta/classificação , Receptores Adrenérgicos beta 1/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Transdução de Sinais
20.
Sheng Li Xue Bao ; 55(3): 251-4, 2003 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-12817289

RESUMO

The expression of beta-adrenergic receptor subtypes and its effect on neonatal rat cardiac fibroblast proliferation were investigated by radioligand binding assay and [(3)H]-thymidine incorporation analysis, respectively. The results indicated that there was no significant difference in the beta-adrenergic receptor density (B(max)) and affinity (K(D)) between cardiomyocytes and cardiac fibroblasts. The [(125)I]-pindolol competitive inhibition curves (ICI 118551 and CGP 20712A) were significantly better fit in a one-site model in membrane preparation of cardiac fibroblasts. In cultured cardiac fibroblasts, 0.1 micromol/L isoproterenol-induced [(3)H]-thymidine incorporation was completely inhibited by a selective beta (2)-AR antagonist ICI 118551, or a non-selective beta-AR antagonist propranolol, but not by CGP 20712A, a selective beta(1)-AR antagonist. These results suggest that isoproterenol-induced cardiac fibroblast proliferation is mediated by beta(2)-AR, the preponderant beta-AR subtype in cardiac fibroblasts.


Assuntos
Fibroblastos/citologia , Miócitos Cardíacos/citologia , Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 2/fisiologia , Animais , Animais Recém-Nascidos , Proliferação de Células , Células Cultivadas , Fibroblastos/metabolismo , Miócitos Cardíacos/metabolismo , Ratos , Ratos Wistar , Receptores Adrenérgicos beta 2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA