Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Basic Res Cardiol ; 111(1): 4, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26611208

RESUMO

Persistent elevation of Ca(2+) influx due to prolongation of the action potential (AP), chronic activation of the ß-adrenergic system and molecular remodeling occurs in stressed and diseased hearts. Increases in Ca(2+) influx are usually linked to prolonged myocyte action potentials and arrhythmias. However, the contribution of chronic enhancement of Cav1.2 activity on cardiac electrical remodeling and arrhythmogenicity has not been completely defined and is the subject of this study. Chronically increased Cav1.2 activity was produced with a cardiac specific, inducible double transgenic (DTG) mouse system overexpressing the ß2a subunit of Cav (Cavß2a). DTG myocytes had increased L-type Ca(2+) current (ICa-L), myocyte shortening, and Ca(2+) transients. DTG mice had enhanced cardiac performance, but died suddenly and prematurely. Telemetric electrocardiograms revealed shortened QT intervals in DTG mice. The action potential duration (APD) was shortened in DTG myocytes due to significant increases of potassium currents and channel abundance. However, shortened AP in DTG myocytes did not fully limit excess Ca(2+) influx and increased the peak and tail ICa-L. Enhanced ICa promoted sarcoplasmic reticulum (SR) Ca(2+) overload, diastolic Ca(2+) sparks and waves, and increased NCX activity, causing increased occurrence of early and delayed afterdepolarizations (EADs and DADs) that may contribute to premature ventricular beats and ventricular tachycardia. AV blocks that could be related to fibrosis of the AV node were also observed. Our study suggests that increasing ICa-L does not necessarily result in AP prolongation but causes SR Ca(2+) overload and fibrosis of AV node and myocardium to induce cellular arrhythmogenicity, arrhythmias, and conduction abnormalities.


Assuntos
Potenciais de Ação/fisiologia , Arritmias Cardíacas/fisiopatologia , Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Arritmias Cardíacas/metabolismo , Western Blotting , Camundongos , Camundongos Transgênicos , Microscopia Confocal
2.
FASEB J ; 28(2): 956-65, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24132623

RESUMO

To clarify the potential utility of targeting GRK2/3-mediated desensitization as a means of manipulating airway smooth muscle (ASM) contractile state, we assessed the specificity of GRK2/3 regulation of procontractile and relaxant G-protein-coupled receptors in ASM. Functional domains of GRK2/3 were stably expressed, or siRNA-mediated GRK2/3 knockdown was performed, in human ASM cultures, and agonist-induced signaling was assessed. Regulation of contraction of murine tracheal rings expressing GRK2 C terminus was also assessed. GRK2/3 knockdown or expression of the GRK2 C terminus caused a significant (∼ 30-90%) increase in maximal ß-agonist and histamine [phosphoinositide (PI) hydrolysis] signaling, without affecting the calculated EC50. GRK2 C-terminal expression did not affect signaling by methacholine, thrombin, or LTD4. Expression of the GRK2 N terminus or kinase-dead holo-GRK2 diminished (∼ 30-70%) both PI hydrolysis and Ca(2+) mobilization by every Gq-coupled receptor examined. Under conditions of GRK2 C-terminal expression, ß-agonist inhibition of methacholine-stimulated PI hydrolysis was greater. Finally, transgenic expression of the GRK2 C terminus in murine ASM enabled ∼ 30-50% greater ß-agonist-mediated relaxation of methacholine-induced contraction. Collectively these data demonstrate the relative selectivity of GRKs for the ß2AR in ASM and the ability to exploit GRK2/3 functional domains to render ASM hyporesponsive to contractile agents while increasing responsiveness to bronchodilating ß-agonist.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Músculo Liso/metabolismo , Cálcio/metabolismo , AMP Cíclico/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/química , Quinase 3 de Receptor Acoplado a Proteína G/química , Humanos , RNA Interferente Pequeno , Receptores Acoplados a Proteínas G/metabolismo , Sistema Respiratório/citologia , Transdução de Sinais/fisiologia
3.
J Inherit Metab Dis ; 38(6): 1029-39, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25875217

RESUMO

Global metabolic profiling currently achievable by untargeted mass spectrometry-based metabolomic platforms has great potential to advance our understanding of human disease states, including potential utility in the detection of novel and known inborn errors of metabolism (IEMs). There are few studies of the technical reproducibility, data analysis methods, and overall diagnostic capabilities when this technology is applied to clinical specimens for the diagnosis of IEMs. We explored the clinical utility of a metabolomic workflow capable of routinely generating semi-quantitative z-score values for ~900 unique compounds, including ~500 named human analytes, in a single analysis of human plasma. We tested the technical reproducibility of this platform and applied it to the retrospective diagnosis of 190 individual plasma samples, 120 of which were collected from patients with a confirmed IEM. Our results demonstrate high intra-assay precision and linear detection for the majority compounds tested. Individual metabolomic profiles provided excellent sensitivity and specificity for the detection of a wide range of metabolic disorders and identified novel biomarkers for some diseases. With this platform, it is possible to use one test to screen for dozens of IEMs that might otherwise require ordering multiple unique biochemical tests. However, this test may yield false negative results for certain disorders that would be detected by a more well-established quantitative test and in its current state should be considered a supplementary test. Our findings describe a novel approach to metabolomic analysis of clinical specimens and demonstrate the clinical utility of this technology for prospective screening of IEMs.


Assuntos
Biomarcadores/análise , Erros Inatos do Metabolismo/diagnóstico , Metabolômica/métodos , Triagem Neonatal/métodos , Humanos , Recém-Nascido , Reprodutibilidade dos Testes , Estudos Retrospectivos , Sensibilidade e Especificidade
4.
Nat Med ; 13(3): 315-23, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17322894

RESUMO

Cardiac overstimulation by the sympathetic nervous system (SNS) is a salient characteristic of heart failure, reflected by elevated circulating levels of catecholamines. The success of beta-adrenergic receptor (betaAR) antagonists in heart failure argues for SNS hyperactivity being pathogenic; however, sympatholytic agents targeting alpha2AR-mediated catecholamine inhibition have been unsuccessful. By investigating adrenal adrenergic receptor signaling in heart failure models, we found molecular mechanisms to explain the failure of sympatholytic agents and discovered a new strategy to lower SNS activity. During heart failure, there is substantial alpha2AR dysregulation in the adrenal gland, triggered by increased expression and activity of G protein-coupled receptor kinase 2 (GRK2). Adrenal gland-specific GRK2 inhibition reversed alpha2AR dysregulation in heart failure, resulting in lowered plasma catecholamine levels, improved cardiac betaAR signaling and function, and increased sympatholytic efficacy of a alpha2AR agonist. This is the first demonstration, to our knowledge, of a molecular mechanism for SNS hyperactivity in heart failure, and our study identifies adrenal GRK2 activity as a new sympatholytic target.


Assuntos
Glândulas Suprarrenais/enzimologia , Hiperfunção Adrenocortical/enzimologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Insuficiência Cardíaca/enzimologia , Regulação para Cima/fisiologia , Quinases de Receptores Adrenérgicos beta/biossíntese , Glândulas Suprarrenais/patologia , Glândulas Suprarrenais/fisiopatologia , Hiperfunção Adrenocortical/fisiopatologia , Animais , Células Cultivadas , Quinase 2 de Receptor Acoplado a Proteína G , Insuficiência Cardíaca/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Ratos , Ratos Sprague-Dawley , Quinases de Receptores Adrenérgicos beta/antagonistas & inibidores , Quinases de Receptores Adrenérgicos beta/fisiologia
5.
Circ Res ; 108(1): 27-39, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21106943

RESUMO

RATIONALE: The G(ßγ)-sequestering peptide ß-adrenergic receptor kinase (ßARK)ct derived from the G-protein-coupled receptor kinase (GRK)2 carboxyl terminus has emerged as a promising target for gene-based heart failure therapy. Enhanced downstream cAMP signaling has been proposed as the underlying mechanism for increased ß-adrenergic receptor (ßAR) responsiveness. However, molecular targets mediating improved cardiac contractile performance by ßARKct and its impact on G(ßγ)-mediated signaling have yet to be fully elucidated. OBJECTIVE: We sought to identify G(ßγ)-regulated targets and signaling mechanisms conveying ßARKct-mediated enhanced ßAR responsiveness in normal (NC) and failing (FC) adult rat ventricular cardiomyocytes. METHODS AND RESULTS: Assessing viral-based ßARKct gene delivery with electrophysiological techniques, analysis of contractile performance, subcellular Ca²(+) handling, and site-specific protein phosphorylation, we demonstrate that ßARKct enhances the cardiac L-type Ca²(+) channel (LCC) current (I(Ca)) both in NCs and FCs on ßAR stimulation. Mechanistically, ßARKct augments I(Ca) by preventing enhanced inhibitory interaction between the α1-LCC subunit (Cav1.2α) and liberated G(ßγ) subunits downstream of activated ßARs. Despite improved ßAR contractile responsiveness, ßARKct neither increased nor restored cAMP-dependent protein kinase (PKA) and calmodulin-dependent kinase II signaling including unchanged protein kinase (PK)Cε, extracellular signal-regulated kinase (ERK)1/2, Akt, ERK5, and p38 activation both in NCs and FCs. Accordingly, although ßARKct significantly increases I(Ca) and Ca²(+) transients, being susceptible to suppression by recombinant G(ßγ) protein and use-dependent LCC blocker, ßARKct-expressing cardiomyocytes exhibit equal basal and ßAR-stimulated sarcoplasmic reticulum Ca²(+) load, spontaneous diastolic Ca²(+) leakage, and survival rates and were less susceptible to field-stimulated Ca²(+) waves compared with controls. CONCLUSION: Our study identifies a G(ßγ)-dependent signaling pathway attenuating cardiomyocyte I(Ca) on ßAR as molecular target for the G(ßγ)-sequestering peptide ßARKct. Targeted interruption of this inhibitory signaling pathway by ßARKct confers improved ßAR contractile responsiveness through increased I(Ca) without enhancing regular or restoring abnormal cAMP-signaling. ßARKct-mediated improvement of I(Ca) rendered cardiomyocytes neither susceptible to ßAR-induced damage nor arrhythmogenic sarcoplasmic reticulum Ca²(+) leakage.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Cardiotônicos/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Terapia Genética/métodos , Insuficiência Cardíaca , Contração Miocárdica/genética , Miócitos Cardíacos/metabolismo , Peptídeos/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Sobrevivência Celular/genética , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/terapia , Ventrículos do Coração/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Peptídeos/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Ratos , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo
6.
Circ Res ; 107(9): 1140-9, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20814022

RESUMO

RATIONALE: Activation of prosurvival kinases and subsequent nitric oxide (NO) production by certain G protein-coupled receptors (GPCRs) protects myocardium in ischemia/reperfusion injury (I/R) models. GPCR signaling pathways are regulated by GPCR kinases (GRKs), and GRK2 has been shown to be a critical molecule in normal and pathological cardiac function. OBJECTIVE: A loss of cardiac GRK2 activity is known to arrest progression of heart failure (HF), at least in part by normalization of cardiac ß-adrenergic receptor (ßAR) signaling. Chronic HF studies have been performed with GRK2 knockout mice, as well as expression of the ßARKct, a peptide inhibitor of GRK2 activity. This study was conducted to examine the role of GRK2 and its activity during acute myocardial ischemic injury using an I/R model. METHODS AND RESULTS: We demonstrate, using cardiac-specific GRK2 and ßARKct-expressing transgenic mice, a deleterious effect of GRK2 on in vivo myocardial I/R injury with ßARKct imparting cardioprotection. Post-I/R infarct size was greater in GRK2-overexpressing mice (45.0±2.8% versus 31.3±2.3% in controls) and significantly smaller in ßARKct mice (16.8±1.3%, P<0.05). Importantly, in vivo apoptosis was found to be consistent with these reciprocal effects on post-I/R myocardial injury when levels of GRK2 activity were altered. Moreover, these results were reflected by higher Akt activation and induction of NO production via ßARKct, and these antiapoptotic/survival effects could be recapitulated in vitro. Interestingly, selective antagonism of ß(2)ARs abolished ßARKct-mediated cardioprotection, suggesting that enhanced GRK2 activity on this GPCR is deleterious to cardiac myocyte survival. CONCLUSION: The novel effect of reducing acute ischemic myocardial injury via increased Akt activity and NO production adds significantly to the therapeutic potential of GRK2 inhibition with the ßARKct not only in chronic HF but also potentially in acute ischemic injury conditions.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Apoptose/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Animais , Células Cultivadas , Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Camundongos , Camundongos Transgênicos , Ratos
7.
J Mol Cell Cardiol ; 50(3): 460-70, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21111744

RESUMO

Pathological cardiac hypertrophy (PCH) is associated with the development of arrhythmia and congestive heart failure. While calcium (Ca(2+)) is implicated in hypertrophic signaling pathways, the specific role of Ca(2+) influx through the L-type Ca(2+) channel (I(Ca-L)) has been controversial and is the topic of this study. To determine if and how sustained increases in I(Ca-L) induce PCH, transgenic mouse models with low (LE) and high (HE) expression levels of the ß2a subunit of Ca(2+) channels (ß2a) and in cultured adult feline (AF) and neonatal rat (NR) ventricular myocytes (VMs) infected with an adenovirus containing a ß2a-GFP were used. In vivo, ß2a LE and HE mice had increased heart weight to body weight ratio, posterior wall and interventricular septal thickness, tissue fibrosis, myocyte volume, and cross-sectional area and the expression of PCH markers in a time- and dose-dependent manner. PCH was associated with a hypercontractile phenotype including enhanced I(Ca-L), fractional shortening, peak Ca(2+) transient, at the myocyte level, greater ejection fraction, and fractional shortening at the organ level. In addition, LE mice had an exaggerated hypertrophic response to transverse aortic constriction. In vitro overexpression of ß2a in cultured AFVMs increased I(Ca-L), cell volume, protein synthesis, NFAT, and HDAC translocations and in NRVMs increased surface area. These effects were abolished by the blockade of I(Ca-L), intracellular Ca(2+), calcineurin, CaMKII, and SERCA. In conclusion, increasing I(Ca-L) is sufficient to induce PCH through the calcineurin/NFAT and CaMKII/HDAC pathways. Both cytosolic and SR/ER-nuclear envelop Ca(2+) pools were shown to be involved.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Animais , Calcineurina/metabolismo , Canais de Cálcio Tipo L/biossíntese , Canais de Cálcio Tipo L/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Gatos , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Histona Desacetilases/metabolismo , Camundongos , Camundongos Transgênicos , Contração Miocárdica , Fatores de Transcrição NFATC/metabolismo , Membrana Nuclear/metabolismo , Fenótipo , Ratos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Transdução de Sinais
8.
Biochim Biophys Acta ; 1802(12): 1268-75, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20060896

RESUMO

Hypertension represents a complex, multifactorial disease and contributes to the major causes of morbidity and mortality in industrialized countries: ischemic and hypertensive heart disease, stroke, peripheral atherosclerosis and renal failure. Current pharmacological therapy of essential hypertension focuses on the regulation of vascular resistance by inhibition of hormones such as catecholamines and angiotensin II, blocking them from receptor activation. Interaction of G-protein coupled receptor kinases (GRKs) and regulator of G-protein signaling (RGS) proteins with activated G-protein coupled receptors (GPCRs) effect the phosphorylation state of the receptor leading to desensitization and can profoundly impair signaling. Defects in GPCR regulation via these modulators have severe consequences affecting GPCR-stimulated biological responses in pathological situations such as hypertension, since they fine-tune and balance the major transmitters of vessel constriction versus dilatation, thus representing valuable new targets for anti-hypertensive therapeutic strategies. Elevated levels of GRKs are associated with human hypertensive disease and are relevant modulators of blood pressure in animal models of hypertension. This implies therapeutic perspective in a disease that has a prevalence of 65million in the United States while being directly correlated with occurrence of major adverse cardiac and vascular events. Therefore, therapeutic approaches using the inhibition of GRKs to regulate GPCRs are intriguing novel targets for treatment of hypertension and heart failure.


Assuntos
Hipertensão/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Angiotensina II/genética , Angiotensina II/metabolismo , Animais , Anti-Hipertensivos/uso terapêutico , Pressão Sanguínea/genética , Catecolaminas/genética , Catecolaminas/metabolismo , Modelos Animais de Doenças , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/epidemiologia , Hipertensão/genética , Prevalência , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Acoplados a Proteínas G/genética , Estados Unidos/epidemiologia , Vasoconstrição/genética , Vasodilatação/genética
10.
Proc Natl Acad Sci U S A ; 105(34): 12457-62, 2008 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-18711143

RESUMO

G protein-coupled receptor (GPCR) kinases (GRKs) are critical regulators of cellular signaling and function. In cardiomyocytes, GRK2 and GRK5 are two GRKs important for myocardial regulation, and both have been shown to be up-regulated in the dysfunctional heart. We report that increased levels and activity of GRK5 in failing myocardium may have unique significance due to its nuclear localization, a property not shared by GRK2. We find that transgenic mice with elevated cardiac GRK5 levels have exaggerated hypertrophy and early heart failure compared with control mice after pressure overload. This pathology is not present in cardiac GRK2-overexpressing mice or in mice with overexpression of a mutant GRK5 that is excluded from the nucleus. Nuclear accumulation of GRK5 is enhanced in myocytes after aortic banding in vivo and in vitro in myocytes after increased G alpha q activity, the trigger for pressure-overload hypertrophy. GRK5 enhances activation of MEF2 in concert with Gq signals, demonstrating that nuclear localized GRK5 regulates gene transcription via a pathway critically linked to myocardial hypertrophy. Mechanistically, we show that this is due to GRK5 acting, in a non-GPCR manner, as a class II histone deacetylase (HDAC) kinase because it can associate with and phosphorylate the myocyte enhancer factor-2 repressor, HDAC5. Moreover, significant HDAC activity can be found with GRK5 in the heart. Our data show that GRK5 is a nuclear HDAC kinase that plays a key role in maladaptive cardiac hypertrophy apparently independent of any action directly on GPCRs.


Assuntos
Núcleo Celular/enzimologia , Quinase 5 de Receptor Acoplado a Proteína G/fisiologia , Miócitos Cardíacos/enzimologia , Animais , Quinase 5 de Receptor Acoplado a Proteína G/análise , Quinase 5 de Receptor Acoplado a Proteína G/genética , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/etiologia , Histona Desacetilases/metabolismo , Hipertrofia/enzimologia , Hipertrofia/etiologia , Fatores de Transcrição MEF2 , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/ultraestrutura , Fatores de Regulação Miogênica/metabolismo , Regulação para Cima
11.
Circ Res ; 103(4): 413-22, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18635825

RESUMO

Myocardial G protein-coupled receptor kinase (GRK)2 is a critical regulator of cardiac beta-adrenergic receptor (betaAR) signaling and cardiac function. Its upregulation in heart failure may further depress cardiac function and contribute to mortality in this syndrome. Preventing GRK2 translocation to activated betaAR with a GRK2-derived peptide that binds G(beta)gamma (betaARKct) has benefited some models of heart failure, but the precise mechanism is uncertain, because GRK2 is still present and betaARKct has other potential effects. We generated mice in which cardiac myocyte GRK2 expression was normal during embryonic development but was ablated after birth (alphaMHC-Cre x GRK2 fl/fl) or only after administration of tamoxifen (alphaMHC-MerCreMer x GRK2 fl/fl) and examined the consequences of GRK2 ablation before and after surgical coronary artery ligation on cardiac adaptation after myocardial infarction. Absence of GRK2 before coronary artery ligation prevented maladaptive postinfarction remodeling and preserved betaAR responsiveness. Strikingly, GRK2 ablation initiated 10 days after infarction increased survival, enhanced cardiac contractile performance, and halted ventricular remodeling. These results demonstrate a specific causal role for GRK2 in postinfarction cardiac remodeling and heart failure and support therapeutic approaches of targeting GRK2 or restoring betaAR signaling by other means to improve outcomes in heart failure.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Insuficiência Cardíaca/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Antagonistas de Estrogênios/farmacologia , Quinase 2 de Receptor Acoplado a Proteína G/genética , Regulação da Expressão Gênica , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/prevenção & controle , Ligadura , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Infarto do Miocárdio/complicações , Infarto do Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Receptores Adrenérgicos beta/metabolismo , Tamoxifeno/farmacologia
12.
Circ Res ; 102(7): 786-94, 2008 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-18292599

RESUMO

S100A1, a Ca(2+)-binding protein of the EF-hand type, is known to modulate sarcoplasmic reticulum Ca(2+) handling in skeletal muscle and cardiomyocytes. Recently, S100A1 has been shown to be expressed in endothelial cells (ECs). Because intracellular Ca(2+) ([Ca(2+)](i)) transients can be involved in important EC functions and endothelial NO synthase activity, we sought to investigate the impact of endothelial S100A1 on the regulation of endothelial and vascular function. Thoracic aortas from S100A1 knockout mice (SKO) showed significantly reduced relaxation in response to acetylcholine compared with wild-type vessels, whereas direct vessel relaxation using sodium nitroprusside was unaltered. Endothelial dysfunction attributable to the lack of S100A1 expression could also be demonstrated in vivo and translated into hypertension of SKO. Mechanistically, both basal and acetylcholine-induced endothelial NO release of SKO aortas was significantly reduced compared with wild type. Impaired endothelial NO production in SKO could be attributed, at least in part, to diminished agonist-induced [Ca(2+)](i) transients in ECs. Consistently, silencing endothelial S100A1 expression in wild type also reduced [Ca(2+)](i) and NO generation. Moreover, S100A1 overexpression in ECs further increased NO generation that was blocked by the inositol-1,4,5-triphosphate receptor blocker 2-aminoethoxydiphenylborate. Finally, cardiac endothelial S100A1 expression was shown to be downregulated in heart failure in vivo. Collectively, endothelial S100A1 critically modulates vascular function because lack of S100A1 expression leads to decreased [Ca(2+)](i) and endothelial NO release, which contributes, at least partially, to impaired endothelium-dependent vascular relaxation and hypertension in SKO mice. Targeting endothelial S100A1 expression may, therefore, be a novel therapeutic means to improve endothelial function in vascular disease or heart failure.


Assuntos
Endotélio Vascular/metabolismo , Óxido Nítrico/metabolismo , Proteínas S100/metabolismo , Vasoconstrição/fisiologia , Vasodilatação/fisiologia , Acetilcolina/farmacologia , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Cálcio/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Feminino , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Hipertensão/metabolismo , Hipertensão/patologia , Hipertensão/fisiopatologia , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitroprussiato/farmacologia , Ratos , Ratos Wistar , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
13.
J Mol Cell Cardiol ; 46(1): 100-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18930063

RESUMO

Chronic ventricular pressure overload states, such as hypertension, and elevated levels of neurohormones (norepinephrine, angiotensin II, endothelin-1) initiate cardiac hypertrophy and dysfunction and share the property of being able to bind to Gq-coupled 7-transmembrane receptors. The goal of the current study was to determine the role of endogenous cardiac myocyte Gq signaling and its role in cardiac hypertrophy and dysfunction during high blood pressure (BP). We induced renal artery stenosis for 8 weeks in control mice and mice expressing a peptide inhibitor of Gq signaling (GqI) using a 2 kidney, 1 clip renal artery stenosis model. 8 weeks following chronic high BP, control mice had cardiac hypertrophy and depressed function. Inhibition of cardiomyocyte Gq signaling did not reverse cardiac hypertrophy but attenuated increases in a profile of cardiac profibrotic genes and genes associated with remodeling. Inhibition of Gq signaling also attenuated the loss of cardiac function. We determined that Gq signaling downstream of angiotensin II receptor stimulation negatively impacted beta-adrenergic receptor (AR) responses and inhibition of Gq signaling was sufficient to restore betaAR-mediated responses. Therefore, in this study we found that Gq signaling negatively impacts cardiac function during high BP. Specifically, we found that inhibition of AT1-Gq signaling augmented betaAR mediated effects in a renal artery stenosis model of hypertension. These observations may underlie additional, beneficial effects of angiotensinogen converting enzyme (ACE) inhibitors and angiotensin receptor antagonists observed during times of hemodynamic stress.


Assuntos
Angiotensina II/antagonistas & inibidores , Angiotensina II/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Obstrução da Artéria Renal/patologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Ecocardiografia/métodos , Feminino , Hemodinâmica , Hipertensão , Masculino , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais
14.
Circulation ; 117(11): 1378-87, 2008 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-18316484

RESUMO

BACKGROUND: A salient characteristic of dysfunctional myocardium progressing to heart failure is an upregulation of the adenylyl cyclase inhibitory guanine nucleotide (G) protein alpha subunit, G alpha(i2). It has not been determined conclusively whether increased Gi activity in the heart is beneficial or deleterious in vivo. Gi signaling has been implicated in the mechanism of cardioprotective agents; however, no in vivo evidence exists that any of the G alpha subunits are cardioprotective. We have created a novel molecular tool to specifically address the role of Gi proteins in normal and dysfunctional myocardium. METHODS AND RESULTS: We have developed a class-specific Gi inhibitor peptide, GiCT, composed of the region of G alpha(i2) that interacts specifically with G protein-coupled receptors. GiCT inhibits Gi signals specifically in vitro and in vivo, whereas Gs and Gq signals are not affected. In vivo expression of GiCT in transgenic mice effectively causes a "functional knockout" of cardiac G alpha(i2) signaling. Inducible, cardiac-specific GiCT transgenic mice display a baseline phenotype consistent with nontransgenic mice. However, when subjected to ischemia/reperfusion injury, GiCT transgenic mice demonstrate a significant increase in infarct size compared with nontransgenic mice (from 36.9+/-2.5% to 50.9+/-4.3%). Mechanistically, this post-ischemia/reperfusion phenotype includes increased myocardial apoptosis and resultant decreased contractile performance. CONCLUSIONS: Overall, our results demonstrate the in vivo utility of GiCT to dissect specific mechanisms attributed to Gi signaling in stressed myocardium. Our results with GiCT indicate that upregulation of G alpha(i2) is an adaptive protective response after ischemia to shield myocytes from apoptosis.


Assuntos
Apoptose/fisiologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Mitocôndrias Cardíacas/fisiologia , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/química , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Humanos , Isoproterenol/farmacologia , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Estresse Oxidativo , Fragmentos de Peptídeos/genética , Ratos , Receptores Acoplados a Proteínas G/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais/fisiologia , Transdução Genética
15.
Zhonghua Bing Li Xue Za Zhi ; 38(1): 50-4, 2009 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-19489226

RESUMO

OBJECTIVE: To study the activation of sterol regulatory element binding protein (SREBP) and its critical role in endothelial cell migration. METHODS: Bovine aortic endothelial cells (ECs) were cultured. The expression of SREBP and Cdc42 were determined by Western blot and quantitative real-time PCR. Moreover, outward growth migration model and transwell chamber assay were used to detect ECs migration. RESULTS: (1) SREBP was activated during ECs migration. Western blot analysis demonstrated increased active form SREBP in migrating as compared to non-migrating ECs population. SREBP activation decreased as ECs migration slowed;(2) Coincidental with SREBP activation, mRNA expression of its target genes such as low density lipoprotein receptor, HMG-CoA reductase, and fatty acid synthase also increased in migrating ECs population as detected by real-time PCR; (3) Migration induced SREBP activation in ECs was inhibited by SREBP-acting protein RNAi and pharmacologically by 25-hydroxycholesterol; (4) Inhibition of SREBP led to decreased ECs migration in various models; (5) Cells genetically deficient in SREBP-acting protein, S1P, or S2P, phenotypically exhibited impaired migration; (6) SREBP inhibition in ECs suppressed the activity of small GTPase Cdc42, a key molecule for ECs motility. CONCLUSIONS: SREBP is activated during and plays a critical role in ECs migration. Targeting SREBP could become a novel approach in fighting diseases involving abnormal ECs migration.


Assuntos
Movimento Celular , Ácido Graxo Sintases/metabolismo , Hidroximetilglutaril-CoA Redutases/metabolismo , Receptores de LDL/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Animais , Aorta/citologia , Células CHO , Bovinos , Células Cultivadas , Cricetinae , Cricetulus , Células Endoteliais , Ácido Graxo Sintases/genética , Hidroxicolesteróis/farmacologia , Hidroximetilglutaril-CoA Redutases/genética , Interferência de RNA , RNA Mensageiro/metabolismo , Receptores de LDL/genética , Proteínas de Ligação a Elemento Regulador de Esterol/fisiologia
16.
Cardiovasc Res ; 78(2): 376-84, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18267956

RESUMO

AIMS: Vascular endothelial growth factor (VEGF)-induced endothelial cell migration and angiogenesis are associated with the vascular complications of diabetes mellitus, and adiponectin is an abundant plasma adipokine that exhibits salutary effects on endothelial function. We investigated whether adiponectin suppresses VEGF-induced migration and related signal transduction responses in human coronary artery endothelial cells (HCAECs). METHODS AND RESULTS: Using a modified Boyden chamber technique and a monolayer 'wound-healing' assay, both the recombinant adiponectin globular domain and full-length adiponectin protein potently suppressed the migration of HCAEC induced by VEGF. Adiponectin did not increase endothelial cell apoptosis, as measured by terminal deoxynucleotidyl transferase biotin-dUTP Nick End Labelling assay. Adiponectin also suppressed VEGF-induced reactive oxygen species generation, activation of Akt, the mitogen-activated protein kinase ERK and the RhoGTPase RhoA, and induction of the formation of actin stress fibres and focal cellular adhesions. VEGF-stimulated cell migration was inhibited by activation of adenylyl cyclase with forskolin, and adiponectin treatment increased cellular cyclic adenosine monophosphate (cAMP) levels and protein kinase A (PKA) enzymatic activity. Pharmacological inhibition of either adenylyl cyclase or PKA significantly abrogated the effect of adiponectin globular domain to suppress VEGF-induced cell migration. CONCLUSION: Adiponectin suppresses VEGF-stimulated HCAEC migration via cAMP/PKA-dependent signalling, an important effect with implications for a regulatory role of adiponectin in vascular processes associated with diabetes and atherosclerosis.


Assuntos
Movimento Celular , Células Endoteliais/metabolismo , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inibidores de Adenilil Ciclases , Adenilil Ciclases/metabolismo , Adiponectina/metabolismo , Apoptose , Movimento Celular/efeitos dos fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Adesões Focais/metabolismo , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/metabolismo , Fibras de Estresse/metabolismo , Cicatrização , Proteína rhoA de Ligação ao GTP/metabolismo
17.
Circulation ; 115(19): 2506-15, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17470693

RESUMO

BACKGROUND: The incidence of heart failure is ever-growing, and it is urgent to develop improved treatments. An attractive approach is gene therapy; however, the clinical barrier has yet to be broken because of several issues, including the lack of an ideal vector supporting safe and long-term myocardial transgene expression. METHODS AND RESULTS: Here, we show that the use of a recombinant adeno-associated viral (rAAV6) vector containing a novel cardiac-selective enhancer/promoter element can direct stable cardiac expression of a therapeutic transgene, the calcium (Ca2+)-sensing S100A1, in a rat model of heart failure. The chronic heart failure-rescuing properties of myocardial S100A1 expression, the result of improved sarcoplasmic reticulum Ca2+ handling, included improved contractile function and left ventricular remodeling. Adding to the clinical relevance, long-term S100A1 therapy had unique and additive beneficial effects over beta-adrenergic receptor blockade, a current pharmacological heart failure treatment. CONCLUSIONS: These findings demonstrate that stable increased expression of S100A1 in the failing heart can be used for long-term reversal of LV dysfunction and remodeling. Thus, long-term, cardiac-targeted rAAV6-S100A1 gene therapy may be of potential clinical utility in human heart failure.


Assuntos
Terapia Genética , Insuficiência Cardíaca/terapia , Proteínas S100/fisiologia , Actinas/genética , Animais , Sítios de Ligação , Sinalização do Cálcio , Cardiomegalia/prevenção & controle , Dependovirus/genética , Elementos Facilitadores Genéticos , Genes Reporter , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Proteínas de Fluorescência Verde/genética , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Testes de Função Cardíaca , Humanos , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica , Infarto do Miocárdio/complicações , Especificidade de Órgãos , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes de Fusão/fisiologia , Proteínas S100/genética
18.
Endocrinology ; 149(7): 3569-75, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18356277

RESUMO

The angiotensin II (AngII) type 1 receptor (AT(1)) plays a critical role in hypertrophy of vascular smooth muscle cells (VSMCs). Although it is well known that G(q) is the major G protein activated by the AT(1) receptor, the requirement of G(q) for AngII-induced VSMC hypertrophy remains unclear. By using cultured VSMCs, this study examined the requirement of G(q) for the epidermal growth factor receptor (EGFR) pathway, the Rho-kinase (ROCK) pathway, and subsequent hypertrophy. AngII-induced intracellular Ca(2+) elevation was completely inhibited by a pharmacological G(q) inhibitor as well as by adenovirus encoding a G(q) inhibitory minigene. AngII (100nm)-induced EGFR transactivation was almost completely inhibited by these inhibitors, whereas these inhibitors only partially inhibited AngII (100nm)-induced phosphorylation of a ROCK substrate, myosin phosphatase target subunit-1. Stimulation of VSMCs with AngII resulted in an increase of cellular protein and cell volume but not in cell number. The G(q) inhibitors completely blocked these hypertrophic responses, whereas a G protein-independent AT(1) agonist did not stimulate these hypertrophic responses. In conclusion, G(q) appears to play a major role in the EGFR pathway, leading to vascular hypertrophy induced by AngII. Vascular G(q) seems to be a critical target of intervention against cardiovascular diseases associated with the enhanced renin-angiotensin system.


Assuntos
Angiotensina II/farmacologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Adenoviridae/genética , Animais , Cálcio/metabolismo , Crescimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/química , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Hipertrofia , Immunoblotting , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 1/metabolismo , Ratos , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 1 de Angiotensina/fisiologia , Quinases Associadas a rho/metabolismo
19.
Clin Sci (Lond) ; 115(3): 79-89, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18593382

RESUMO

Hypertension is a prevalent condition in the developed world and disease severity is directly correlated with additional cardiovascular complications. It is estimated that 30% of the adult population in the United States has hypertension, which is classified as a systolic blood pressure > or =140 mmHg and/or a diastolic blood pressure > or =90 mmHg. A prolonged increase in afterload ultimately leads to congestive heart failure in the majority of cases. Currently, medication designed to treat hypertension is inadequate, thus new therapies need to be explored. Blood pressure is tightly regulated by blood vessel radius, which is established by hormones and/or peptides binding to GPCRs (G-protein-coupled receptors). Catecholamines and peptide hormones, such as AngII (angiotensin II), are elevated in hypertension and, therefore, signalling by these GPCRs is increased. Their signalling is tightly controlled by a class of proteins, the GRKs (GPCR kinases). Elevated levels of either GRK2 or GRK5 in both the lymphocytes and VSM (vascular smooth muscle) are associated with human hypertension and animal models of the disease. The focus of the present review is on the role GRKs, and their regulation of GPCRs, play in high blood pressure.


Assuntos
Quinases de Receptores Acoplados a Proteína G/fisiologia , Hipertensão/fisiopatologia , Receptores Acoplados a Proteínas G/fisiologia , Adenilil Ciclases/fisiologia , Animais , Insuficiência Cardíaca/fisiopatologia , Humanos , Músculo Liso Vascular/fisiopatologia , Proteínas RGS/fisiologia , Transdução de Sinais
20.
Life Sci ; 82(3-4): 174-81, 2008 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-18068195

RESUMO

Postintervention restenosis (PIRS) after balloon angioplasty or stent implantation is a limitation for these interventional procedures even with the advent of new drug-eluting stents. Sterol regulatory element-binding proteins (SREBP) are transcription factors governing cellular lipid biosynthesis and thus critical in the regulation of the lipid-rich cell membranes. PIRS following injury results partially from newly proliferating cells expressing vascular smooth muscle cell (VSMC) markers. Platelet-derived growth factor (PDGF), lysophosphatidic acid (LPA) and alpha(1)-adrenergic receptor stimulation are well recognized diverse mitogens for VSMC activation in PIRS. We examined whether PDGF, LPA and alpha(1)-adrenergic receptor stimulation with phenylephrine (PE) regulate SREBP expression and subsequently, VSMC proliferation. Our results show that PDGF, LPA and PE upregulate SREBP-1 in a time- and dose-dependent manner. PDGF, LPA and PE-mediated proliferation is dependent on SREBP since inhibition of SREBP expression using targeted knockdown of the SREBP precursor SREBP activating protein (SCAP) by siRNA led to an attenuation of SREBP expression and decreased PDGF, LPA and PE induced proliferation. In two different in vivo PIRS models we found that SREBP-1 was enhanced in the injured blood vessel wall, especially within the neointima and co-localized with alpha-smooth muscle actin positive cells. Thus, SREBP is enhanced in the vessel wall following PIRS and is important in the regulation of pro-hyperplasia molecular signaling. SREBP inhibition may be a powerful tool to limit PIRS.


Assuntos
Vasos Sanguíneos/metabolismo , Reestenose Coronária/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Biomarcadores/metabolismo , Vasos Sanguíneos/efeitos dos fármacos , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lisofosfolipídeos/farmacologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fenilefrina/farmacologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1 , Stents
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa