Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Cell Cardiol ; 40(5): 619-28, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16600289

RESUMO

We previously found that a canine model of selective surgical ventricular denervation (VD), which does not permit increased sympathetic tone during the pathogenesis of heart failure (HF), tolerated the development of HF better than controls. To investigate the cellular mechanisms, we examined cellular contraction and L-type Ca(2+) channel currents (I(Ca)) and their responses to beta-adrenergic receptor (beta-AR) stimulation in left ventricular myocytes from 1) control, 2) VD, 3) HF induced by rapid pacing, and 4) HF induced in VD (VD-HF) dogs. The magnitude of myocyte contraction and rate of relaxation in VD were similar to control but were depressed in both HF and VD-HF. These changes were associated with reduced protein expression of sarcoplasmic reticulum Ca(2+)-ATPase (SERCA2a) and protein kinase A phosphorylated phospholamban (PLB), which was reduced in HF, but essentially abolished in VD-HF. beta-AR kinase (GRK2) was increased in HF but reduced in VD-HF. Basal I(Ca) density did not differ among control, VD, and HF groups, but VD-HF myocytes showed a markedly reduced I(Ca) density (approximately 40%). Compared to controls, the sensitivity of I(Ca) to isoproterenol (ISO), was significantly higher in VD, but reduced in HF. While I(Ca) responses to ISO in VD-HF were maintained at control levels, the amplitude of the ISO-stimulated I(Ca) was significantly smaller (approximately 50%) compared with HF myocytes. The relative decrease in Ca(2+) influx due to downregulation of I(Ca) density may contribute to the cardioprotective effects in VD-HF hearts by preventing Ca(2+) overload during the development of HF. These findings, in combination with the virtual abolition of phosphorylated PLB in VD-HF and the decrease in GRK2, may explain, in part, why VD dogs tolerate the development of HF better than control dogs.


Assuntos
Proteínas de Ligação ao Cálcio/química , Regulação para Baixo , Insuficiência Cardíaca/patologia , Quinases de Receptores Adrenérgicos beta/biossíntese , Animais , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Cães , Eletrofisiologia , Hemodinâmica , Miócitos Cardíacos/metabolismo , Fosforilação , Quinases de Receptores Adrenérgicos beta/metabolismo
2.
Circ Res ; 89(12): 1130-7, 2001 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-11739277

RESUMO

The Ras-like Rab GTPases regulate vesicle transport in endocytosis and exocytosis. We found that cardiac Rabs1, 4, and 6 are upregulated in a dilated cardiomyopathy model overexpressing beta(2)-adrenergic receptors. To determine if increased Rab GTPase expression can contribute to cardiomyopathy, we transgenically overexpressed in mouse hearts prototypical Rab1a, the small G protein that regulates vesicle transport from endoplasmic reticulum to and through Golgi. In multiple independent mouse lines, Rab1a overexpression caused cardiac hypertrophy that progressed in a time- and transgene dose-dependent manner to heart failure. Isolated cardiac myocytes were hypertrophied and exhibited contractile depression with impaired calcium reuptake. Ultrastructural analysis revealed enlarged Golgi stacks and increased transitional vesicles in ventricular myocytes, with increased secretory atrial natriuretic peptide granules and degenerative myelin figures in atrial myocytes; immunogold studies localized Rab1a to these abnormal vesicular structures. A survey of hypertrophy signaling molecules revealed increased protein kinase C (PKC) alpha and delta, and confocal microscopy showed abnormal subcellular distribution of PKCalpha in Rab1a transgenics. These results indicate that increased expression of Rab1 GTPase in myocardium distorts subcellular localization of proteins and is sufficient to cause cardiac hypertrophy and failure.


Assuntos
Cardiomiopatias/enzimologia , Cardiomiopatias/etiologia , Miocárdio/enzimologia , Proteínas rab de Ligação ao GTP/biossíntese , Animais , Southern Blotting , Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Cardiomiopatias/patologia , Tamanho Celular/genética , Modelos Animais de Doenças , Progressão da Doença , Expressão Gênica , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Humanos , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Miocárdio/patologia , Miocárdio/ultraestrutura , Organelas/ultraestrutura , Técnicas de Patch-Clamp , Proteína Quinase C/metabolismo , Transporte Proteico , RNA Mensageiro/metabolismo , Transdução de Sinais , Especificidade da Espécie , Transgenes , Regulação para Cima/genética , Proteínas rab de Ligação ao GTP/genética , Proteínas rab1 de Ligação ao GTP/biossíntese , Proteínas rab1 de Ligação ao GTP/genética
3.
Circ Res ; 89(9): 831-7, 2001 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-11679414

RESUMO

The mechanism of myocardial stunning has been studied extensively in rodents and is thought to involve a decrease in Ca(2+) responsiveness of the myofilaments, degradation of Troponin I (TnI), and no change in Ca(2+) handling. We studied the mechanism of stunning in isolated myocytes from chronically instrumented pigs. Myocytes were isolated from the ischemic (stunned) and nonischemic (normal) regions after 90-minute coronary stenosis followed by 60-minute reperfusion. Baseline myocyte contraction was reduced, P<0.01, in stunned myocytes (6.3+/-0.4%) compared with normal myocytes (8.8+/-0.4%). The time for 70% relaxation was prolonged, P<0.01, in stunned myocytes (131+/-8 ms) compared with normal myocytes (105+/-5 ms). The impaired contractile function was associated with decreased Ca(2+) transients (stunned, 0.33+/-0.04 versus normal, 0.49+/-0.05, P<0.01). Action potential measurements in stunned myocytes demonstrated a decrease in plateau potential without a change in resting membrane potential. These changes were associated with decreased L-type Ca(2+)-current density (stunned, -4.8+/-0.4 versus normal, -6.6+/-0.4 pA/pF, P<0.01). There were no differences in TnI, sarcoplasmic reticulum Ca(2+) ATPase (SERCA2a), and phospholamban protein quantities. However, the fraction of phosphorylated phospholamban monomer was reduced in stunned myocardium. In rats, stunned myocytes demonstrated reduced systolic contraction but actually accelerated relaxation and no change in Ca(2+) transients. Thus, mechanisms of stunning in the pig are radically different from the widely held concepts derived from studies in rodents and involve impaired Ca(2+) handling and dephosphorylation of phospholamban, but not TnI degradation.


Assuntos
Cálcio/metabolismo , Contração Miocárdica , Miocárdio Atordoado/fisiopatologia , Potenciais de Ação , Animais , Canais de Cálcio Tipo L/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , ATPases Transportadoras de Cálcio/metabolismo , Separação Celular , Estimulação Elétrica , Immunoblotting , Técnicas In Vitro , Isoenzimas/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Ratos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Especificidade da Espécie , Suínos , Troponina I/metabolismo
4.
Circ Res ; 89(2): 160-7, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11463723

RESUMO

Ectopic expression of the sarcoplasmic reticulum (SR) Ca(2+) ATPase (SERCA) 1a pump in the mouse heart results in a 2.5-fold increase in total SERCA pump level. SERCA1a hearts show increased rates of contraction/relaxation and enhanced Ca(2+) transients; however, the cellular mechanisms underlying altered Ca(2+) handling in SERCA1a transgenic (TG) hearts are unknown. In this study, using confocal microscopy, we demonstrate that SERCA1a protein traffics to the cardiac SR and structurally substitutes for the endogenous SERCA2a isoform. SR Ca(2+) load measurements revealed that TG myocytes have significantly enhanced SR Ca(2+) load. Confocal line-scan images of field-stimulated SR Ca(2+) release showed an increased rate of Ca(2+) removal in TG myocytes. On the other hand, ryanodine receptor binding activity was decreased by approximately 30%. However, TG myocytes had a greater rate of spontaneous ryanodine receptor opening as measured by spark frequency. Whole-cell L-type Ca(2+) current density was reduced by approximately 50%, whereas the time course of inactivation was unchanged in TG myocytes. These studies provide important evidence that SERCA1a can substitute both structurally and functionally for SERCA2a in the heart and that SERCA1a overexpression can be used to enhance SR Ca(2+) transport and cardiac contractility.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Cálcio/metabolismo , Miocárdio/metabolismo , Retículo Sarcoplasmático/enzimologia , Animais , Ligação Competitiva , Western Blotting , Cafeína/farmacologia , Canais de Cálcio Tipo L/fisiologia , ATPases Transportadoras de Cálcio/genética , Coração/fisiologia , Proteínas de Homeodomínio/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Contração Miocárdica/fisiologia , Miocárdio/citologia , Miocárdio/enzimologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rianodina/metabolismo , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo
5.
J Biol Chem ; 276(26): 24145-52, 2001 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-11328820

RESUMO

To determine whether selective impairment of cardiac sarcoplasmic reticulum (SR) Ca(2+) transport may drive the progressive functional deterioration leading to heart failure, transgenic mice, overexpressing a phospholamban Val(49) --> Gly mutant (2-fold), which is a superinhibitor of SR Ca(2+)-ATPase affinity for Ca(2+), were generated, and their cardiac phenotype was examined longitudinally. At 3 months of age, the increased EC(50) level of SR Ca(2+) uptake for Ca(2+) (0.67 +/- 0.09 microm) resulted in significantly higher depression of cardiomyocyte rates of shortening (57%), relengthening (31%), and prolongation of the Ca(2+) signal decay time (165%) than overexpression (2-fold) of wild type phospholamban (68%, 64%, and 125%, respectively), compared with controls (100%). Echocardiography also revealed significantly depressed function and impaired beta-adrenergic responses in mutant hearts. The depressed contractile parameters were associated with left ventricular remodeling, recapitulation of fetal gene expression, and hypertrophy, which progressed to dilated cardiomyopathy with interstitial tissue fibrosis and death by 6 months in males. Females also had ventricular hypertrophy at 3 months but exhibited normal systolic function up to 12 months of age. These results suggest a causal relationship between defective SR Ca(2+) cycling and cardiac remodeling leading to heart failure, with a gender-dependent influence on the time course of these alterations.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Insuficiência Cardíaca/etiologia , Contração Miocárdica , Retículo Sarcoplasmático/fisiologia , Envelhecimento , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/fisiologia , Proteínas de Ligação ao Cálcio/genética , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Células Cultivadas , Ecocardiografia , Feminino , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Miocárdio/patologia , Mutação Puntual , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Fatores Sexuais , Taxa de Sobrevida
7.
J Mol Cell Cardiol ; 33(2): 249-59, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11162130

RESUMO

Cardiac-specific expression of an activated calcineurin protein in the hearts of transgenic (CLN) mice produces a profound hypertrophy that rapidly progresses to heart failure. While calcineurin is regulated by Ca2+, the potential effects of calcineurin on cardiac myocyte Ca2+ handling has not been evaluated. To this end, we examined L-type Ca2+ currents (I(Ca)) in left ventricular myocytes. CLN myocytes had larger (approximately 80%) cell capacitance and enhanced I(Ca) density (approximately 20%) compared with non-transgenic (NTG) littermates, but no change in the current-voltage relationship, single-channel conductance or protein levels of alpha 1 or beta 2 subunit of L-type Ca2+ channels. Interestingly, the kinetics of I(Ca) inactivation was faster (approximately two-fold) in CLN myocytes compared with NTG myocytes. Ryanodine application slowed the rate of I(Ca) inactivation in both groups and abolished the kinetic difference, suggesting that Ca2+ dependent inactivation is increased in CLN myocytes due to altered SR Ca2+ release. Treatment of CLN mice with Cyclosporine A (CsA), a calcineurin inhibitor, prevented myocyte hypertrophy and changes in I(Ca) activity and inactivation kinetics. However, there was no direct effect of CsA on I(Ca) in either NTG or CLN myocytes, suggesting that endogenous calcineurin activity does not directly regulate Ca2+ channel activity. This interpretation is consistent with the observation that I(Ca) density, inactivation kinetics and regulation by isoproterenol were normal in cardiac-specific transgenic mice expressing calcineurin inhibitory protein domains from either Cain or AKAP79. Taken together these data suggest that chronic activation of calcineurin is associated with myocyte hypertrophy and a secondary enhancement of intracellular Ca2+ handling that is tied to the hypertrophy response itself.


Assuntos
Calcineurina/metabolismo , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Cardiomegalia/metabolismo , Animais , Western Blotting , Inibidores de Calcineurina , Canais de Cálcio/genética , Ciclosporina/farmacologia , Eletrofisiologia , Isoproterenol/farmacologia , Cinética , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/metabolismo , Estrutura Terciária de Proteína , Rianodina/farmacologia
8.
J Biol Chem ; 276(12): 9392-9, 2001 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-11115498

RESUMO

Cardiac-specific overexpression of murine cardiac calsequestrin results in depressed cardiac contractile parameters, low Ca(2+)-induced Ca(2+) release from sarcoplasmic reticulum (SR) and cardiac hypertrophy in transgenic mice. To test the hypothesis that inhibition of phospholamban activity may rescue some of these phenotypic alterations, the calsequestrin overexpressing mice were cross-bred with phospholamban-knockout mice. Phospholamban ablation in calsequestrin overexpressing mice led to reversal of the depressed cardiac contractile parameters in Langendorff-perfused hearts or in vivo. This was associated with increases of SR Ca(2+) storage, assessed by caffeine-induced Na(+)-Ca(2+) exchanger currents. The inactivation time of the L-type Ca(2+) current (I(Ca)), which has an inverse correlation with Ca(2+)-induced SR Ca(2+) release, and the relation between the peak current density and half-inactivation time were also normalized, indicating a restoration in the ability of I(Ca) to trigger SR Ca(2+) release. The prolonged action potentials in calsequestrin overexpressing cardiomyocytes also reversed to normal upon phospholamban ablation. Furthermore, ablation of phospholamban restored the expression levels of atrial natriuretic factor and alpha-skeletal actin mRNA as well as ventricular myocyte size. These results indicate that attenuation of phospholamban function may prevent or overcome functional and remodeling defects in hypertrophied hearts.


Assuntos
Calsequestrina/metabolismo , Cardiomegalia , Contração Miocárdica , Miocárdio/metabolismo , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Fator Natriurético Atrial/biossíntese , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/fisiologia , Coração/efeitos dos fármacos , Imuno-Histoquímica , Isoproterenol/farmacologia , Camundongos , Camundongos Knockout , Miocárdio/citologia , Técnicas de Patch-Clamp , Retículo Sarcoplasmático/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 279(1): H139-48, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10899051

RESUMO

Transgenic overexpression of G alpha(q) causes cardiac hypertrophy and depressed contractile responses to beta-adrenergic receptor agonists. The electrophysiological basis of the altered myocardial function was examined in left ventricular myocytes isolated from transgenic (G alpha(q)) mice. Action potential duration was significantly prolonged in G alpha(q) compared with nontransgenic (NTG) myocytes. The densities of inward rectifier K(+) currents, transient outward K(+) currents (I(to)), and Na(+)/Ca(2+) exchange currents were reduced in G alpha(q) myocytes. Consistent with functional measurements, Na(+)/Ca(2+) exchanger gene expression was reduced in G alpha(q) hearts. Kinetics or sensitivity of I(to) to 4-aminopyridine was unchanged, but 4-aminopyridine prolonged the action potential more in G alpha(q) myocytes. Isoproterenol increased L-type Ca(2+) currents (I(Ca)) in both groups, with a similar EC(50), but the maximal response in G alpha(q) myocytes was approximately 24% of that in NTG myocytes. In NTG myocytes, the maximal increase of I(Ca) with isoproterenol or forskolin was similar. In G alpha(q) myocytes, forskolin was more effective and enhanced I(Ca) up to approximately 55% of that in NTG myocytes. These results indicate that the changes in ionic currents and multiple defects in the beta-adrenergic receptor/Ca(2+) channel signaling pathway contribute to altered ventricular function in this model of cardiac hypertrophy.


Assuntos
Cardiomegalia/fisiopatologia , Proteínas de Ligação ao GTP/fisiologia , Coração/fisiologia , Receptores Adrenérgicos beta/fisiologia , 4-Aminopiridina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/fisiologia , Células Cultivadas , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica , Coração/efeitos dos fármacos , Coração/fisiopatologia , Ventrículos do Coração , Isoproterenol/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Valores de Referência , Transdução de Sinais , Trocador de Sódio e Cálcio/genética
11.
Circ Res ; 86(11): 1173-9, 2000 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-10850970

RESUMO

Protein kinase C (PKC) is a key mediator of many diverse physiological and pathological responses. Although little is known about the specific in vivo roles of the various cardiac PKC isozymes, activation-induced translocation of PKC is believed to be the primary determinant of isozyme-specific functions. Recently, we have identified a catalytically inactive peptide translocation inhibitor (epsilonV1) and translocation activator (psiepsilonRACK [receptors for activated C kinase]) specifically targeting PKCepsilon. Using cardiomyocyte-specific transgenic expression of these peptides, we combined loss- and gain-of-function approaches to elucidate the in vivo consequences of myocardial PKCepsilon signaling. As expected for a PKCepsilon RACK binding peptide, confocal microscopy showed that epsilonV1 decorated cross-striated elements and intercalated disks of cardiac myocytes. Inhibition of cardiomyocyte PKCepsilon by epsilonV1 at lower expression levels upregulated alpha-skeletal actin gene expression, increased cardiomyocyte cell size, and modestly impaired left ventricular fractional shortening. At high expression levels, epsilonV1 caused a lethal dilated cardiomyopathy. In contrast, enhancement of PKCepsilon translocation with psiepsilonRACK resulted in selectively increased beta myosin heavy chain gene expression and normally functioning concentric ventricular remodeling with decreased cardiomyocyte size. These results identify for the first time a role for PKCepsilon signaling in normal postnatal maturational myocardial development and suggest the potential for PKCepsilon activators to stimulate "physiological" cardiomyocyte growth.


Assuntos
Coração/fisiologia , Isoenzimas/fisiologia , Proteína Quinase C/fisiologia , Actinas/genética , Animais , Transporte Biológico/fisiologia , Cardiomegalia/etiologia , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Cardiomiopatia Dilatada/etiologia , Expressão Gênica/fisiologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos/genética , Contração Miocárdica/fisiologia , Miocárdio/patologia , Cadeias Pesadas de Miosina/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Quinase C-épsilon , Receptores de Quinase C Ativada , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Remodelação Ventricular/fisiologia
12.
Circulation ; 101(14): 1707-14, 2000 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-10758054

RESUMO

BACKGROUND: Transgenic cardiac beta(2)-adrenergic receptor (AR) overexpression has resulted in enhanced signaling and cardiac function in mice, whereas relatively low levels of transgenically expressed G(alphas) or beta(1)AR have resulted in phenotypes of ventricular failure. Potential relationships between the levels of betaAR overexpression and biochemical, molecular, and physiological consequences have not been reported. METHODS AND RESULTS: We generated transgenic mice expressing beta(2)AR at 3690, 7120, 9670, and 23 300 fmol/mg in the heart, representing 60, 100, 150, and 350 times background betaAR expression. All lines showed enhanced basal adenylyl cyclase activation but a decrease in forskolin- and NaF-stimulated adenylyl cyclase activities. Mice of the highest-expressing line developed a rapidly progressive fibrotic dilated cardiomyopathy and died of heart failure at 25+/-1 weeks of age. The 60-fold line exhibited enhanced basal cardiac function without increased mortality when followed for 1 year, whereas 100-fold overexpressors developed a fibrotic cardiomyopathy and heart failure, with death occurring at 41+/-1 weeks of age. Adenylyl cyclase activation did not correlate with early or delayed decompensation. Propranolol administration reduced baseline +dP/dt(max) to nontransgenic levels in all beta(2)AR transgenics except the 350-fold overexpressors, indicating that spontaneous activation of beta(2)AR was present at this level of expression. CONCLUSIONS: These data demonstrate that the heart tolerates enhanced contractile function via 60-fold beta(2)AR overexpression without detriment for a period of >/=1 year and that higher levels of expression result in either aggressive or delayed cardiomyopathy. The consequences for enhanced betaAR function in the heart appear to be highly dependent on which signaling elements are increased and to what extent.


Assuntos
Miocárdio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Animais , Canais de Cálcio/metabolismo , Canais de Cálcio/fisiologia , Baixo Débito Cardíaco/etiologia , Baixo Débito Cardíaco/mortalidade , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Cardiomiopatia Dilatada/diagnóstico por imagem , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Ecocardiografia , Condutividade Elétrica , Fibrose , Hemodinâmica , Humanos , Camundongos , Camundongos Transgênicos/genética , Contração Miocárdica/fisiologia , Miocárdio/patologia , Concentração Osmolar , Estudos Prospectivos , Fatores de Tempo
13.
Mol Pharmacol ; 57(2): 278-87, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10648637

RESUMO

Targeted cardiac overexpression of the alpha-subunit of the heterotrimeric G protein G(q) in transgenic mice evokes hypertrophy and depressed stimulation of cardiac inotropy and chronotropy by beta-adrenergic receptor (betaAR) agonists in vivo, which is a hallmark of many forms of experimental and human heart failure. The molecular basis of this betaAR dysfunction was explored in transgenic mice overexpressing G(alphaq) approximately 5-fold over background. Isoproterenol-stimulated adenylyl cyclase activities in myocardial membranes were significantly depressed in G(alphaq) mice compared with nontransgenic controls (19.7 +/- 2.6 versus 43.7 +/- 5. 6 pmol/min/mg) without a decrease in betaAR expression levels. Functional coupling of both betaAR subtypes was impaired. Similarly, in whole-cell patch-clamp studies, betaAR stimulation of L-type Ca(2+) channel currents was depressed approximately 75% in the G(alphaq) mice. Cardiac betaAR from these mice showed decreased formation of the active high-affinity conformation (R(H) = 29% versus 62% for nontransgenic littermates), confirming a receptor-G(s)-coupling defect. Of the three candidate kinases that might impose this uncoupling by receptor phosphorylation (protein kinase A, betaAR kinase, protein kinase C), only protein kinase C activity was elevated in G(alphaq) mouse hearts. Type V adenylyl cyclase was decreased approximately 45% in these mice, consistent with decreased basal, NaF, and forskolin-stimulated enzyme activities. Although cellular G(s) levels were unaltered, G(i2) and G(i3) were increased in G(alphaq) mice. Pertussis toxin treatment of isolated G(alphaq) myocytes resulted in an improvement in betaAR, but not that of forskolin or NaF, stimulation of adenylyl cyclase. Thus three distinct mechanisms contribute to impaired betaAR function by in vivo G(q) signaling cross-talk in myocytes. Because many elements of hypertrophy and/or failure in cellular and animal models can be initiated by increased G(alphaq) signaling, the current work may be broadly applicable to interfaces whereby modification of heart failure might be considered.


Assuntos
Cardiomegalia/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais/fisiologia , Disfunção Ventricular/metabolismo , Adenilil Ciclases/efeitos dos fármacos , Adenilil Ciclases/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Animais , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Humanos , Radioisótopos do Iodo , Camundongos , Camundongos Transgênicos , Técnicas de Patch-Clamp , Pindolol/análogos & derivados , Pindolol/farmacologia , Receptores Adrenérgicos beta/efeitos dos fármacos
14.
Gen Comp Endocrinol ; 116(3): 422-32, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10603280

RESUMO

Plasma levels of testosterone and corticosterone were measured in free-living male bush warblers captured on their breeding ground at different times of the breeding season. Their territoriality was also estimated from their singing response to song playbacks. The pattern of change detected in the levels of plasma testosterone was different from that of "typical" monogamous species but similar to that of polygynous species. In "typical" monogamous species, plasma testosterone levels elevated during territory settlement and courtship behavior and then declined to low, stable levels during incubation. In bush warblers, plasma levels of testosterone were already high (1-2 ng/ml) upon arrival in late March and peaked (2. 5-4 ng/ml) in early June. They then decreased but relatively high levels were maintained until early August. In late August the testosterone concentration was 0.03 ng/ml or less. Plasma levels of corticosterone also showed a seasonal change, being highest in May to July and declining in late August. Territoriality showed clear seasonality, reflecting the levels of circulating testosterone. Upon arrival, latency periods for responses to song playback were long and singing activity was rather low but this behavior was soon stabilized and a high degree of territoriality was maintained to late August. These results suggest that high levels of circulating testosterone and corticosterone allow males to pursue a polygynous breeding strategy, to hold a territory, and to maintain breeding activity for a prolonged period, characteristics which are likely to be adaptations to dense bushes with high rates of predation and brood parasitism of this species.


Assuntos
Comportamento Animal/fisiologia , Corticosterona/sangue , Comportamento Sexual Animal/fisiologia , Aves Canoras/fisiologia , Testosterona/sangue , Animais , Peso Corporal , Masculino , Reprodução/fisiologia , Estações do Ano , Territorialidade , Vocalização Animal
15.
Eur J Pharmacol ; 384(1): 17-24, 1999 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-10611414

RESUMO

Chronic administration of norepinephrine for 8 weeks has been shown to reduce neuronal norepinephrine uptake activity and increase interstitial norepinephrine concentration in the heart. To determine whether the changes could lead to myocardial beta-adrenoceptor down-regulation or beta-adrenergic subsensitivity, we measured left ventricular contractile responses to dobutamine, myocardial beta-adrenoceptor density, beta subtype distribution, competitive inhibition agonist binding, and adenylyl cyclase activity activation by isoproterenol, 5'-guanylylimidodiphosphate, and forskolin in dogs after a norepinephrine or saline infusion for 8 weeks. We found that norepinephrine infusion reduced myocardial beta-adrenoceptor density, beta(1)-adrenoceptor subtype density, and high-affinity site for isoproterenol. Left ventricular contractile responses to dobutamine were reduced in the norepinephrine-infused animals. In addition, norepinephrine infusion decreased the basal adenylyl cyclase activity and the adenylyl cyclase responses to isoproterenol, 5'-guanylylimidodiphosphate, and forskolin. The findings indicate that a decrease in cardiac norepinephrine uptake predisposes the heart to norepinephrine-induced myocardial beta-adrenoceptor down-regulation, and that norepinephrine, when present in a sufficient amount over a long period as it is in chronic heart failure, can reduce myocardial beta-adrenergic responsiveness by both homologous and heterologous desensitization.


Assuntos
Ventrículos do Coração/efeitos dos fármacos , Norepinefrina/farmacologia , Norepinefrina/farmacocinética , Receptores Adrenérgicos beta/metabolismo , Adenilil Ciclases/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Animais , Ligação Competitiva/efeitos dos fármacos , Colforsina/farmacologia , Dobutamina/farmacologia , Cães , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Guanilil Imidodifosfato/farmacologia , Ventrículos do Coração/metabolismo , Hemodinâmica/efeitos dos fármacos , Imidazóis/farmacologia , Técnicas In Vitro , Radioisótopos do Iodo , Iodocianopindolol/metabolismo , Iodocianopindolol/farmacologia , Isoproterenol/farmacologia , Contração Miocárdica/efeitos dos fármacos , Receptores Adrenérgicos beta/efeitos dos fármacos , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Função Ventricular
16.
Biochemistry ; 38(50): 16706-13, 1999 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-10600134

RESUMO

The limiting element in beta-adrenergic receptor (betaAR)-G(s)-adenylyl cyclase (AC) signal transduction in the cardiomyocyte is not known, but it has been proposed that the level of adenylyl cyclase expression constrains betaAR signaling. To alter the above equilibrium, type V AC was overexpressed in a myocyte-specific manner in the hearts of transgenic mice using the alpha-myosin heavy chain promoter. Expression of type V AC was approximately 75% over endogenous levels as quantitated by [(3)H]forskolin binding. Functional activity of the transgene product was evident in cardiac membrane AC studies, where basal (45 +/- 11 vs 19 +/- 5 pmol min(-)(1) mg(-)(1)) and forskolin+Mn(2+) (695 +/- 104 vs 386 +/- 34 pmol min(-)(1) mg(-)(1)) stimulated activities were increased compared to activities in nontransgenic (NTG) littermates. However, while isoproterenol stimulated activities were higher (74 +/- 12 vs 46 +/- 9.8 pmol min(-)(1) mg(-)(1)), the fold stimulation over basal was not increased in ACV overexpressors compared to NTG (line 14.3 = 2.29 +/- 0.44-fold, line 15.1 = 1.70 +/- 0.1-fold, NTG = 2.62 +/- 0.18-fold). Similarly, in whole cell patch-clamp studies, betaAR-mediated opening of L-type Ca(2+) channels was not found to be enhanced in transgenic ACV myocytes (225 +/- 15 vs 216 +/- 10% of basal currents). Basal and isoproterenol stimulated PKA activities were elevated in the ACV mice compared to NTG, but again the extent of stimulation over basal was not enhanced. Phosphorylated phospholamban was approximately 2-fold greater in myocytes from ACV hearts compared to NTG, indicating that distal elements of the contractile cascade are activated by AC overexpression. ACV mice displayed increased heart rates and fractional shortening as assessed by echocardiography. However, in vivo hemodynamic studies revealed that heart rate and contractility responses to agonist infusion were not enhanced in ACV mice compared to NTG. We conclude that at native stoichiometries, the levels of adenylyl cyclase influence basal activities and cardiac function, but do not constrain betaAR signaling in the cardiomyocyte.


Assuntos
Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Miocárdio/enzimologia , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Adenilil Ciclases/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Western Blotting , Canais de Cálcio Tipo L/metabolismo , Catálise , Eletrocardiografia , Testes de Função Cardíaca , Frequência Cardíaca/genética , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Receptores Adrenérgicos beta/fisiologia , Transdução de Sinais/genética
17.
Am J Physiol ; 277(4): H1609-17, 1999 10.
Artigo em Inglês | MEDLINE | ID: mdl-10516201

RESUMO

Cardiac sympathetic nerve terminal dysfunction plays an important role in the downregulation of myocardial beta-adrenoceptors in heart failure. To determine whether chronic angiotensin-converting enzyme (ACE) inhibition improved cardiac sympathetic nerve terminal function and hence increased myocardial beta-adrenergic responsiveness, we administered ACE inhibitors to dogs with chronic right-sided heart failure (RHF) produced by tricuspid avulsion and pulmonary artery constriction. The RHF animals exhibited fluid retention, elevated right heart filling pressures, blunted inotropic response to isoproterenol, and reduced beta-adrenoceptor density. These changes were accompanied by decreases in right ventricular norepinephrine (NE) uptake and neuronal NE histofluorescence and tyrosine hydroxylase immunoreactive profiles. ACE inhibitors had no effect on the production of heart failure but greatly reduced the attenuation of cardiac NE uptake, neuronal NE histofluorescence, and tyrosine hydroxylase immunoreactive profiles. ACE inhibition also improved the inotropic response to isoproterenol and restored myocardial beta-adrenoceptor density. The changes probably are caused by reduction of cardiac NE release by ACE inhibition and may contribute to the beneficial effects of ACE inhibitor therapy in patients with chronic heart failure.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Insuficiência Cardíaca/fisiopatologia , Miocárdio/metabolismo , Terminações Nervosas/fisiopatologia , Norepinefrina/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Animais , Cães , Enalapril/farmacologia , Fluorescência , Glioxilatos/farmacologia , Insuficiência Cardíaca/metabolismo , Hemodinâmica , Terminações Nervosas/enzimologia , Peptidil Dipeptidase A/metabolismo , Ramipril/farmacologia , Receptores Adrenérgicos beta/metabolismo , Sistema Nervoso Simpático/enzimologia , Tirosina 3-Mono-Oxigenase/metabolismo
18.
J Mol Cell Cardiol ; 31(7): 1327-36, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10403750

RESUMO

Transgenic mice with cardiac-specific overexpression of G alpha q exhibit a biochemical and physiological phenotype of load-independent cardiac hypertrophy with contractile dysfunction. To elucidate the cellular basis for altered contractility, we measured cellular contraction, Ca(2+)transients, and l -type Ca(2+)channel currents (I(Ca)) in left ventricular (LV) myocytes isolated from non transgenic (NT) controls or G alpha q hearts. Although baseline contractile function (% shortening) and the amplitude of Ca(2+)transients in G alpha q myocytes were similar to NT myocytes, the rates of cellular shortening and relengthening and the duration of Ca(2+)transients were prolonged in G alpha q myocytes. Myocytes from G alpha q hearts had larger cell capacitance but no change in I(Ca)density, voltage-dependence of activation and inactivation. The responses of I(Ca)to dihydropyridine drugs and a membrane permeable cAMP analog, 8-(4-chlorophenylthio) cAMP, were not altered; however, the time course of I(Ca)inactivation was significantly slower in G alpha q myocytes compared to NT myocytes. The kinetic difference in inactivation was abolished when Ba(2+)was used as the charge carrier or when the sarcoplasmic reticulum (SR) Ca(2+)was depleted by ryanodine, suggesting that Ca(2+)-dependent inactivation is reduced in G alpha q myocytes due to altered SR Ca(2+)release. Consistent with this hypothesis, the function of SR as assessed by the maximal Ca(2+)uptake rates and the apparent affinity of SR Ca(2+)-ATPase for Ca(2+)was reduced in ventricles of G alpha q heart. These results suggest that the reduced SR function contributes to the depressed contractility associated with this form of cardiac hypertrophy.


Assuntos
Proteínas de Ligação ao GTP/biossíntese , Miocárdio/metabolismo , Animais , Cálcio/metabolismo , Cálcio/fisiologia , Canais de Cálcio/fisiologia , Eletrofisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Proteínas de Ligação ao GTP/genética , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Camundongos , Camundongos Transgênicos , Contração Miocárdica , Retículo Sarcoplasmático/metabolismo , Fatores de Tempo
19.
Cell Signal ; 11(5): 337-42, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10376806

RESUMO

Beta1- and beta2-adrenergic receptors (beta-ARs) co-exist in mammalian heart, and it is generally accepted that both activate adenylyl cyclase (AC), resulting in increased levels of cAMP and subsequent activation of L-type Ca2+ channels (CaCh). To investigate the contribution of each beta-AR subtype in AC and CaCh coupling, we stably expressed cardiac CaCh alpha1 and beta2 subunits along with either beta1-AR or beta2-AR in CHW fibroblasts. Co-expression of either beta-AR with CaCh subunits conferred responsiveness of AC and CaCh to isoproterenol (ISO), which was not observed in non-transfected cells. ISO-promoted cAMP formation occurred at a lower EC50 through the beta2-AR than through the beta1-AR (0.13 +/- 0.01 vs. 0.6 +/- 0.14 nM). In contrast, activation of CaCh was more efficacious via the beta1-AR than the beta2-AR (EC50 for CaCh activation = 238 +/- 33 vs. 1057 +/- 113 nM). Pre-treatment with pertussis toxin (PTX) had no effect upon the responsiveness of either cAMP formation or CaCh activation through either receptor. We conclude (1) that beta1-ARs exhibit preferential coupling to CaCh activation, versus that observed for the beta2-AR; (2) that this preferential coupling cannot be explained solely by cAMP-dependent processes; and (3) that the relative attenuation of beta2-AR-promoted CaCh activation is not due to receptor coupling to PTX-sensitive G proteins. Thus, it is likely that other subtype-specific, cAMP-independent coupling of the beta-AR to CaCh is present.


Assuntos
Canais de Cálcio/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Canais de Cálcio Tipo L , AMP Cíclico/metabolismo , Miocárdio/metabolismo
20.
J Clin Invest ; 103(7): 1089-97, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10194482

RESUMO

We investigated the mechanisms responsible for altered contractile and relaxation function in overexpressed Gsalpha myocytes. Although baseline contractile function (percent contraction) in Gsalpha mice was similar to that of wild-type (WT) mice, left ventricular myocyte contraction, fura-2 Ca2+transients, and Ca2+ channel currents (ICa) were greater in Gsalpha mice in response to 10(-8) M isoproterenol (ISO) compared with WT mice. The late phase of relaxation of the isolated myocytes and fura-2 Ca2+ transients was accelerated at baseline in Gsalpha but did not increase further with ISO. In vivo measurements using echocardiography also demonstrated enhanced relaxation at baseline in Gsalpha mice. Forskolin and CaCl2 increased contraction similarly in WT and Gsalpha mice. Rp-cAMP, an inhibitor of protein kinase, blocked the increases in contractile response and Ca2+ currents to ISO in WT and to forskolin in both WT and Gsalpha. It also blocked the accelerated relaxation in Gsalpha at baseline but not the contractile response to ISO in Gsalpha myocytes. Baseline measurements of cAMP and phospholambation phosphorylation were enhanced in Gsalpha compared with WT. These data indicate that overexpression of Gsalpha accelerates relaxation at end diastolic but does not affect baseline systolic function in isolated myocytes. However, the enhanced responses to sympathetic stimulation partly reflect increased Ca2+ channel activity; i.e the cellular mechanisms mediating these effects appear to involve a cAMP-independent as well as a cAMP-dependent pathway.


Assuntos
Canais de Cálcio/metabolismo , AMP Cíclico/farmacologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Contração Miocárdica/efeitos dos fármacos , Animais , Cálcio/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Colforsina/farmacologia , Ventrículos do Coração/efeitos dos fármacos , Isoproterenol/farmacologia , Cinética , Camundongos , Relaxamento Muscular/efeitos dos fármacos , Técnicas de Patch-Clamp , Fosforilação , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...