Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Physiol Plant ; 161(3): 322-338, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28665551

RESUMO

Salicylic acid (SA) is a plant hormone involved in a number of physiological responses including both local and systemic resistance of plants to pathogens. In Arabidopsis, SA is glucosylated to form either SA 2-O-ß-d-glucose (SAG) or SA glucose ester (SGE). In this study, we show that SAG accumulates in the vacuole of Arabidopsis, while the majority of SGE was located outside the vacuole. The uptake of SAG by vacuolar membrane-enriched vesicles isolated from Arabidopsis was stimulated by the addition of MgATP and was inhibited by both vanadate (ABC transporter inhibitor) and bafilomycin A1 (vacuolar H+ -ATPase inhibitor), suggesting that SAG uptake involves both an ABC transporter and H+ -antiporter. Despite its absence in the vacuole, we observed the MgATP-dependent uptake of SGE by Arabidopsis vacuolar membrane-enriched vesicles. SGE uptake was not inhibited by vanadate but was inhibited by bafilomycin A1 and gramicidin D providing evidence that uptake was dependent on an H+ -antiporter. The uptake of both SAG and SGE was also inhibited by quercetin and verapamil (two known inhibitors of multidrug efflux pumps) and salicin and arbutin. MgATP-dependent SAG and SGE uptake exhibited Michaelis-Menten-type saturation kinetics. The vacuolar enriched-membrane vesicles had a 46-fold greater affinity and a 10-fold greater transport activity with SGE than with SAG. We propose that in Arabidopsis, SAG is transported into the vacuole to serve as a long-term storage form of SA while SGE, although also transported into the vacuole, is easily hydrolyzed to release the active hormone which can then be remobilized to other cellular locations.


Assuntos
Arabidopsis/metabolismo , Glucose/metabolismo , Membranas Intracelulares/metabolismo , Ácido Salicílico/metabolismo , Vesículas Transportadoras/metabolismo , Vacúolos/metabolismo , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Arabidopsis/efeitos dos fármacos , Arbutina/farmacologia , Álcoois Benzílicos/farmacologia , Radioisótopos de Carbono/metabolismo , Cromatografia Líquida de Alta Pressão , Glucosídeos/farmacologia , Gramicidina/farmacologia , Membranas Intracelulares/efeitos dos fármacos , Cinética , Metaboloma , Protoplastos/metabolismo , Quercetina/farmacologia , Ácido Salicílico/química , Fatores de Tempo , Vesículas Transportadoras/efeitos dos fármacos , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , ATPases Vacuolares Próton-Translocadoras/metabolismo , Vacúolos/efeitos dos fármacos , Verapamil/farmacologia
2.
J Mol Cell Cardiol ; 76: 73-83, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25134464

RESUMO

Cardiac fibroblasts (CFs) produce and degrade the myocardial extracellular matrix and are critical in maladaptive ventricular remodeling that can result in heart failure (HF). ß-Arrestins are important signaling molecules involved in ß-adrenergic receptor (ß-AR) desensitization and can also mediate signaling in a G protein-independent fashion. We hypothesize that ß-arrestins play an important role in the regulation of adult human CF biology with regard to myofibroblast transformation, increased collagen synthesis, and myocardial fibrosis which are important in the development of HF. ß-Arrestin1 & 2 expression is significantly upregulated in adult human CF isolated from failing left ventricles and ß-AR signaling is uncoupled with loss of ß-agonist-mediated inhibition of collagen synthesis versus normal control CF. Knockdown of either ß-arrestin1 or 2 restored ß-AR signaling and ß-agonist mediated inhibition of collagen synthesis. Overexpression of ß-arrestins in normal CF led to a failing phenotype with increased baseline collagen synthesis, impaired ß-AR signaling, and loss of ß-agonist-mediated inhibition of collagen synthesis. ß-Arrestin knockdown in failing CF diminished TGF-ß stimulated collagen synthesis and also inhibited ERK phosphorylation. Overexpression of ß-arrestins in normal CF increased basal ERK1/2 and Smad2/3 phosphorylation and enhanced TGF-ß-stimulated collagen synthesis. This was prevented by pre-treatment with a MEK1/2 inhibitor. Enhanced ß-arrestin signaling appears to be deleterious in CF by promoting a pro-fibrotic phenotype via uncoupling of ß-AR signaling as well as potentiating ERK and Smad signaling. Targeted inhibition of ß-arrestins in CF may represent a therapeutic strategy to prevent maladaptive myocardial fibrosis.


Assuntos
Arrestinas/fisiologia , Colágenos Fibrilares/biossíntese , Miocárdio/patologia , Miofibroblastos/fisiologia , Remodelação Ventricular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Insuficiência Cardíaca/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Receptores Adrenérgicos beta/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/fisiologia , beta-Arrestinas
3.
Circulation ; 126(24): 2859-69, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23124027

RESUMO

BACKGROUND: The cause and consequences of impaired adrenergic signaling in right ventricular failure/hypertrophy (RVH) are poorly understood. We hypothesized that G protein-coupled receptor kinase-2 (GRK2)-mediated uncoupling of ß-adrenergic receptor signaling impairs inotropic reserve. The implications of right ventricular (RV) adrenergic remodeling for inotrope selection and the therapeutic benefit of interrupting Gßγ-GRK2 interaction, using gallein, were tested. METHODS AND RESULTS: Chamber-specificity and cellular localization of adrenergic remodeling were compared in rodent RVH associated with pulmonary arterial hypertension (PAH-RVH; SU5416+chronic-hypoxia or Monocrotaline) versus pulmonary artery banding-induced RVH (PAB-RVH). Results were corroborated in RV arrays from 10 PAH patients versus controls. Inotropic reserve was assessed in RV- and left ventricular-Langendorff models and in vivo. Gallein therapy (1.8 mg/kg/day ×2-weeks) was assessed. Despite similar RVH, cardiac output (58.3±4.9 versus 82.9±4.8 mL/min; P<0.001) and treadmill distance (41.5±11.6 versus 244.1±12.4 m; P<0.001) were lower in PAH-RVH versus PAB-RVH. In PAH-RVH versus PAB-RVH there was greater downregulation of ß1-, α1- and dopamine-1 receptors, more left ventricular involvement, and greater impairment of RV contractile reserve. RV GRK2 activity increased in parallel with a reduction in both adrenergic receptor expression and inotrope-stimulated cAMP levels (P<0.01). ß1-receptor downregulation also occurred in human PAH-RVH. Dobutamine was superior to dopamine as an RV inotrope, both ex vivo and in vivo. CONCLUSIONS: GRK2-mediated desensitization-downregulation of adrenergic and dopaminergic receptors impairs inotropic reserve in PAH-RVH. Acute inotropic support in RVH is best accomplished by dobutamine, reflecting its better coupling to adenylyl cyclase and the reliance of dopamine on dopamine-1-receptor signaling, which is impaired in RVH. Inhibiting Gßγ-GRK2 interactions has therapeutic benefit in RVH.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Hipertensão Pulmonar/tratamento farmacológico , Hipertrofia Ventricular Direita/tratamento farmacológico , Receptores Adrenérgicos beta/metabolismo , Receptores de Dopamina D1/metabolismo , Xantenos/farmacologia , Animais , Cardiotônicos/farmacologia , Células Cultivadas , Dobutamina/farmacologia , Dopamina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Feminino , Humanos , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/patologia , Masculino , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta/genética , Receptores de Dopamina D1/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
4.
J Biol Chem ; 286(17): 15507-16, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21357420

RESUMO

Cardiac fibroblasts (CF) make up 60-70% of the total cell number in the heart and play a critical role in regulating normal myocardial function and in adverse remodeling following myocardial infarction and the transition to heart failure. Recent studies have shown that increased intracellular cAMP can inhibit CF transformation and collagen synthesis in adult rat CF; however, mechanisms by which cAMP production is regulated in CF have not been elucidated. We investigated the potential role of G protein-coupled receptor kinase-2 (GRK2) in modulating collagen synthesis by adult human CF isolated from normal and failing left ventricles. Baseline collagen synthesis was elevated in failing CF and was not inhibited by ß-agonist stimulation in contrast to normal controls. ß-adrenergic receptor (ß-AR) signaling was markedly uncoupled in the failing CF, and expression and activity of GRK2 were increased 3-fold. Overexpression of GRK2 in normal CF recapitulated a heart failure phenotype with minimal inhibition of collagen synthesis following ß-agonist stimulation. In contrast, knockdown of GRK2 expression in normal CF enhanced cAMP production and led to greater ß-agonist-mediated inhibition of basal and TGFß-stimulated collagen synthesis versus control. Inhibition of GRK2 activity in failing CF by expression of the GRK2 inhibitor, GRK2ct, or siRNA-mediated knockdown restored ß-agonist-stimulated inhibition of collagen synthesis and decreased collagen synthesis in response to TGFß stimulation. GRK2 appears to play a significant role in regulating collagen synthesis in adult human CF, and increased activity of this kinase may be an important mechanism of maladaptive ventricular remodeling as mediated by cardiac fibroblasts.


Assuntos
Colágeno/biossíntese , Fibroblastos/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Miocárdio/citologia , Adulto , Colágeno/antagonistas & inibidores , AMP Cíclico/metabolismo , Fibroblastos/enzimologia , Insuficiência Cardíaca , Ventrículos do Coração/citologia , Humanos , Remodelação Ventricular
5.
Am J Physiol Heart Circ Physiol ; 300(5): H1733-42, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21335476

RESUMO

Apoptosis plays a significant role in maladaptive remodeling and ventricular dysfunction following ischemia-reperfusion injury. There is a critical need for novel approaches to inhibit apoptotic cell death following reperfusion, as this loss of cardiac myocytes can progressively lead to heart failure. We investigated the ability and signaling mechanisms of a high-molecular-weight polyethylene glycol-based copolymer, PEG 15-20, to protect cardiac myocytes from hypoxia-reoxygenation (H-R)-induced cell death and its efficacy in preserving ventricular function following extended hypothermic ischemia and warm reperfusion as relevant to cardiac transplantation. Pretreatment of neonatal rat ventricular myocytes with a 5% PEG solution led to a threefold decline in apoptosis after H-R relative to untreated controls. There was a similar decline in caspase-3 activity in conjunction with inhibition of cytochrome c release from the inner mitochondrial membrane. Treatment with PEG also reduced reactive oxygen species production after H-R, and sarcolemmal lipid-raft architecture was preserved, consistent with membrane stabilization. Cell survival signaling was upregulated after H-R with PEG, as demonstrated by increased phosphorylation of Akt, GSK-3ß, and ERK1/2. There was also maintenance of cardiac myocyte ß-adrenergic signaling, which is critical for myocardial function. PEG 15-20 was very effective in preserving left ventricular function following prolonged hypothermic ischemia and warm reperfusion. PEG 15-20 has a potent protective antiapoptotic effect in cardiac myocytes exposed to H-R injury and may represent a novel therapeutic strategy to decrease myocardial cell death and ventricular dysfunction at the time of reperfusion during acute coronary syndrome or following prolonged donor heart preservation.


Assuntos
Apoptose/efeitos dos fármacos , Miócitos Cardíacos/patologia , Oxigênio/efeitos adversos , Polietilenoglicóis/farmacologia , Função Ventricular/efeitos dos fármacos , Animais , Apoptose/fisiologia , Caspase 3/metabolismo , Hipóxia Celular/fisiologia , Células Cultivadas , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/fisiologia , Modelos Animais , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Oxigênio/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Função Ventricular/fisiologia
6.
Cell Signal ; 28(3): 190-203, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26631573

RESUMO

Cardiac myocyte oxidative stress and apoptosis are considered important mechanisms for the development of heart failure (HF). Chronic HF is characterized by increased circulating catecholamines to augment cardiac output. Long-term stimulation of myocardial ß-adrenergic receptors (ß-ARs) is deleterious in cardiac myocytes, however, the potential mechanisms underlying increased cell death are unclear. We hypothesize that GRK2, a critical regulator of myocardial ß-AR signaling, plays an important role in mediating cellular oxidative stress and apoptotic cell death in response to ß-agonist stimulation. Stimulation of H9c2 cells with a non-selective ß-agonist, isoproterenol (Iso) caused increased oxidative stress and apoptosis. There was also increased Nox4 expression, but no change in Nox2, the primary NADPH isoforms and major sources of ROS generation in cardiac myocytes. Adenoviral-mediated overexpression of GRK2 led to similar increases in ROS production and apoptosis as seen with Iso stimulation. These increases in oxidative stress were abolished by pre-treatment with the non-specific Nox inhibitor, apocynin, or siRNA knockdown of Nox4. Adenoviral-mediated expression of a GRK2 inhibitor prevented ROS production and apoptosis in response to Iso stimulation. ß-Arrestins are signaling proteins that function downstream of GRK2 in ß-AR uncoupling. Adenoviral-mediated overexpression of ß-arrestins increased ROS production and Nox4 expression. Chronic ß-agonist stimulation in mice increased Nox4 expression and apoptosis compared to PBS or AngII treatment. These data demonstrate that GRK2 may play an important role in regulating oxidative stress and apoptosis in cardiac myocytes and provides an additional novel mechanism for the beneficial effects of cardiac-targeted GRK2 inhibition to prevent the development of HF.


Assuntos
Apoptose , NADPH Oxidases/metabolismo , Estresse Oxidativo , Acetofenonas/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Angiotensina II/farmacologia , Animais , Apoptose/efeitos dos fármacos , Arrestinas , Linhagem Celular , AMP Cíclico/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Isoproterenol/farmacologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microscopia Confocal , Mitocôndrias/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Receptores Adrenérgicos beta/química , Receptores Adrenérgicos beta/metabolismo , Rodopsina/metabolismo , Transdução de Sinais/efeitos dos fármacos , beta-Arrestinas
7.
Dis Model Mech ; 8(12): 1579-89, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26449263

RESUMO

Oxidative stress in cardiac fibroblasts (CFs) promotes transformation to myofibroblasts and collagen synthesis leading to myocardial fibrosis, a precursor to heart failure (HF). NADPH oxidase 4 (Nox4) is a major source of cardiac reactive oxygen species (ROS); however, mechanisms of Nox4 regulation are unclear. ß-arrestins are scaffold proteins that signal in G-protein-dependent and -independent pathways; for example, in ERK activation. We hypothesize that ß-arrestins regulate oxidative stress in a Nox4-dependent manner and increase fibrosis in HF. CFs were isolated from normal and failing adult human left ventricles. Mitochondrial ROS/superoxide production was quantitated using MitoSox. ß-arrestin and Nox4 expressions were manipulated using adenoviral overexpression or short interfering RNA (siRNA)-mediated knockdown. Mitochondrial oxidative stress and Nox4 expression in CFs were significantly increased in HF. Nox4 knockdown resulted in inhibition of mitochondrial superoxide production and decreased basal and TGF-ß-stimulated collagen and α-SMA expression. CF ß-arrestin expression was upregulated fourfold in HF. ß-arrestin knockdown in failing CFs decreased ROS and Nox4 expression by 50%. ß-arrestin overexpression in normal CFs increased mitochondrial superoxide production twofold. These effects were prevented by inhibition of either Nox or ERK. Upregulation of Nox4 seemed to be a primary mechanism for increased ROS production in failing CFs, which stimulates collagen deposition. ß-arrestin expression was upregulated in HF and plays an important and newly identified role in regulating mitochondrial superoxide production via Nox4. The mechanism for this effect seems to be ERK-mediated. Targeted inhibition of ß-arrestins in CFs might decrease oxidative stress as well as pathological cardiac fibrosis.


Assuntos
Arrestinas/metabolismo , Fibroblastos/metabolismo , Mitocôndrias/metabolismo , Miocárdio/patologia , Estresse Oxidativo , Linhagem Celular Transformada , Células Cultivadas , Colágeno/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Insuficiência Cardíaca/patologia , Humanos , Mitocôndrias/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , NADPH Oxidase 4 , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Superóxidos/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Regulação para Cima/efeitos dos fármacos , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA