Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Physiol ; 593(3): 521-39, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25433071

RESUMO

KEY POINTS: The renin-angiotensin system plays a key role in cardiovascular physiology and its overactivation has been implicated in the pathogenesis of several major cardiovascular diseases. There is growing evidence that angiotensin II (Ang-II) may function as an intracellular peptide to activate intracellular/nuclear receptors and their downstream signalling effectors independently of cell surface receptors. Current methods used to study intracrine Ang-II signalling are limited to indirect approaches because of a lack of selective intracellularly-acting probes. Here, we present novel photoreleasable Ang-II analogues used to probe intracellular actions with spatial and temporal precision. The photorelease of intracellular Ang-II causes nuclear and cytosolic calcium mobilization and initiates the de novo synthesis of RNA in cardiac cells, demonstrating the application of the method. ABSTRACT: Several lines of evidence suggest that intracellular angiotensin II (Ang-II) contributes to the regulation of cardiac contractility, renal salt reabsorption, vascular tone and metabolism; however, work on intracrine Ang-II signalling has been limited to indirect approaches because of a lack of selective intracellularly-acting probes. Here, we aimed to synthesize and characterize cell-permeant Ang-II analogues that are inactive without uncaging, but release active Ang-II upon exposure to a flash of UV-light, and act as novel tools for use in the study of intracrine Ang-II physiology. We prepared three novel caged Ang-II analogues, [Tyr(DMNB)(4)]Ang-II, Ang-II-ODMNB and [Tyr(DMNB)(4)]Ang-II-ODMNB, based upon the incorporation of the photolabile moiety 4,5-dimethoxy-2-nitrobenzyl (DMNB). Compared to Ang-II, the caged Ang-II analogues showed 2-3 orders of magnitude reduced affinity toward both angiotensin type-1 (AT1R) and type-2 (AT2R) receptors in competition binding assays, and greatly-reduced potency in contraction assays of rat thoracic aorta. After receiving UV-irradiation, all three caged Ang-II analogues released Ang-II and potently induced the contraction of rat thoracic aorta. [Tyr(DMNB)(4)]Ang-II showed the most rapid photolysis upon UV-irradiation and was the focus of subsequent characterization. Whereas Ang-II and photolysed [Tyr(DMNB)(4)]Ang-II increased ERK1/2 phosphorylation (via AT1R) and cGMP production (AT2R), caged [Tyr(DMNB)(4)]Ang-II did not. Cellular uptake of [Tyr(DMNB)(4)]Ang-II was 4-fold greater than that of Ang-II and significantly greater than uptake driven by the positive-control HIV TAT(48-60) peptide. Intracellular photolysis of [Tyr(DMNB)(4)]Ang-II induced an increase in nucleoplasmic Ca(2+) ([Ca(2+)]n), and initiated 18S rRNA and nuclear factor kappa B mRNA synthesis in adult cardiac cells. We conclude that caged Ang-II analogues represent powerful new tools for use in the selective study of intracrine signalling via Ang-II.


Assuntos
Angiotensina II/análogos & derivados , Sinalização do Cálcio , Receptores de Angiotensina/metabolismo , Raios Ultravioleta , Antagonistas de Receptores de Angiotensina/farmacologia , Animais , Fluoresceínas/efeitos da radiação , Corantes Fluorescentes/efeitos da radiação , Células HEK293 , Humanos , Masculino , Microscopia de Fluorescência/métodos , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/agonistas
2.
Am J Physiol Heart Circ Physiol ; 298(6): H2062-70, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20382853

RESUMO

Various antioxidants, including polyphenols, prevent the development of atherosclerosis in animal models, contrasting with the failure of antioxidants to provide benefits in patients with established atherosclerosis. We therefore tested in a mouse model the hypothesis that although catechin is atheroprotective in prevention, catechin brings no global vascular protection when initiated after established atherosclerosis, because aging associated with dyslipidemia has induced irreversible dysfunctions. To this end, LDLr(-/-); hApoB(+/+) atherosclerotic (ATX, 9 mo old) and pre-ATX (3 mo old) male mice were treated with catechin (30 mg x kg(-1) x day(-1)) up to 12 mo of age. Vascular function and endothelium/leukocyte interactions were studied at 12 mo old. The renal artery endothelium-dependent dilations were impaired with age whereas adhesion of leukocytes onto the native aortic endothelium was increased (P < 0.05). Aortic oxidative stress [reactive oxygen species (ROS)] increased (P < 0.05) at 3 mo in ATX and at 12 mo in wild-type mice. Aorta mRNA expression of NADPH oxidase increased, whereas that of manganese superoxide dismutase decreased in 12-mo-old ATX mice only. In mice with established ATX, catechin (from 9 to 12 mo) reduced (P < 0.05) by approximately 60% ROS without affecting plaque burden. Notably, catechin worsened endothelial dysfunction and further increased leukocyte adhesion (P < 0.05) in ATX mice. In contrast, the same catechin treatment reversed all age-related dysfunctions in wild-type mice. On the other hand, in pre-ATX mice treated for 9 mo with catechin, plaque burden was reduced by 64% (P < 0.05) and all vascular markers were normalized to the 3-mo-old values. These results demonstrate that an antioxidant treatment is deleterious in mice with established atherosclerosis.


Assuntos
Envelhecimento/fisiologia , Antioxidantes/farmacologia , Aterosclerose/fisiopatologia , Catequina/farmacologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Animais , Apolipoproteínas B/genética , Apolipoproteínas B/fisiologia , Aterosclerose/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de LDL/genética , Receptores de LDL/fisiologia , Superóxidos/metabolismo
3.
J Mol Neurosci ; 38(1): 67-79, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19052921

RESUMO

In the current model of gamma-aminobutyric acid (GABA) B receptor function, there is a requirement for GABA-B(1/2) heterodimerisation for targetting to the cell surface. However, different lines of evidence suggest that the GABA-B(1) subunit can form a functional receptor in the absence of GABA-B(2). We observed coupling of endogenous GABA-B(1) receptors in the DI-TNC1 glial cell line to the ERK pathway in response to baclofen even though these cells do not express GABA-B(2). GABA-B(1A) receptors were also able to mediate a rapid, transient, and dose-dependent activation of the ERK1/2 MAP kinase pathway when transfected alone into HEK 293 cells. The response was abolished by G(i/o) and MEK inhibition, potentiated by inhibitors of phospholipase C and protein kinase C and did not involve PI-3-kinase activity. Finally, using bioluminescence resonance energy transfer and co-immunoprecipitation, we show the existence of homodimeric GABA-B(1A) receptors in transfected HEK293 cells. Altogether, our observations show that GABA-B(1A) receptors are able to activate the ERK1/2 pathway despite the absence of surface targetting partner GABA-B(2) in both HEK 293 cells and the DI-TNC1 cell line.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Receptores de GABA-B/metabolismo , Animais , Baclofeno/farmacologia , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/química , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , GABAérgicos/farmacologia , Agonistas GABAérgicos/farmacologia , Agonistas dos Receptores de GABA-B , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Toxina Pertussis/farmacologia , Fosforilação , Multimerização Proteica , Ratos , Ácido gama-Aminobutírico/farmacologia
4.
Mech Ageing Dev ; 129(5): 261-70, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18302967

RESUMO

Endothelial senescence may contribute to the pathogenesis of age-related vascular disorders. Furthermore, chronic exposure to risk factors for cardiovascular disease (CVD) accelerates the effects of chronological aging by generating stress-dependent damages, including oxidative stress, therefore promoting stress-induced premature senescence. Our objective was to determine whether a chronic treatment with an antioxidant (N-acetyl-cystein, NAC) could delay senescence of endothelial cells (EC) isolated and cultured from arterial segments of patients with severe coronary artery disease. If EC were considered as one population (n=26), chronic NAC treatment slightly shortened telomere attrition rate associated with senescence but did not significantly delay the onset of endothelial senescence. However, in a subgroup of NAC-treated EC (n=15) cellular senescence was significantly delayed, NAC decreased lipid peroxidation (HNE), activated the catalytic subunit of telomerase (hTERT) and inhibited telomere attrition. In contrast, in another subgroup of EC (n=11) characterized by initial short telomeres, no effect of NAC on HNE and high levels of DNA damages, the antioxidant was not beneficial on senescence, suggesting an irreversible stress-dependent damage. In conclusion, chronic exposure to NAC can delay senescence of diseased EC via hTERT activation and transient telomere stabilization, unless oxidative stress-associated cell damage has become irreversible.


Assuntos
Acetilcisteína/farmacologia , Antioxidantes/farmacologia , Aterosclerose/patologia , Senescência Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Acetilcisteína/administração & dosagem , Idoso , Antioxidantes/administração & dosagem , Aterosclerose/metabolismo , Células Cultivadas , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Feminino , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Artéria Torácica Interna/cirurgia , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , Estresse Oxidativo/efeitos dos fármacos , Telomerase/metabolismo
5.
Mech Ageing Dev ; 128(11-12): 662-71, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18022214

RESUMO

Risk factors for cardiovascular diseases (CVD) increase oxidative stress, and they are proposed to hasten endothelial cell (EC) damage and dysfunction. Our objective was to elucidate the impact of chronic exposure to risk factors for CVD on senescence of EC isolated and cultured from internal mammary arterial segments of patients with severe coronary artery disease. Senescence induced by serial passages resulted in progressive telomere shortening, and short initial telomeres predicted early appearance of senescence in culture. Neither time course of senescence nor telomere length was age-dependent, suggesting that biological age, rather than chronological age, determined the dynamics. Senescence appeared earlier in patients with longer history of risk factor for CVD, and multivariate analysis suggested that hypertension hastened the onset of senescence. Risk factors for CVD override the effects of chronological aging likely by generating stress-dependent damage: senescent EC exhibited oxidative stress (increase in lipid peroxydation and caveolin-1 gene expression) and cell damage markers (loss of eNOS expression and increase in Cox2 mRNA, lower TRF1 protein level). Thus, cell senescence was triggered both by telomere-dependent and -independent pathways. In conclusion, chronic exposure to risk factors for CVD accelerated the development of endothelial senescence that could contribute to the pathogenesis of CVD.


Assuntos
Doenças Cardiovasculares/etiologia , Senescência Celular , Doença da Artéria Coronariana/patologia , Células Endoteliais/patologia , Artéria Torácica Interna/patologia , Adulto , Fatores Etários , Idoso , Índice de Massa Corporal , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Caveolina 1/genética , Caveolina 1/metabolismo , Proliferação de Células , Células Cultivadas , Doença da Artéria Coronariana/complicações , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/metabolismo , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Feminino , Humanos , Hipertensão/complicações , Hipertensão/patologia , Peroxidação de Lipídeos , Masculino , Artéria Torácica Interna/enzimologia , Artéria Torácica Interna/metabolismo , Pessoa de Meia-Idade , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , RNA Mensageiro/metabolismo , Fatores de Risco , Índice de Gravidade de Doença , Fumar/efeitos adversos , Telômero/metabolismo , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Fatores de Tempo
6.
Cell Signal ; 18(12): 2172-81, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16809021

RESUMO

gamma-aminobutyric acid (GABA) is the principal inhibitory neurotransmitter in the mammalian brain. It acts via both ionotropic GABA-A and metabotropic GABA-B receptors. We evaluated the interaction of receptors with members of the inwardly rectifying potassium (Kir 3) channel family, which also play an important role in neuronal transmission and membrane excitability. These channels are functionally regulated by GABA-B receptors. Possible physical interactions between GABA-B receptor and Kir 3 channels expressed in HEK cells were evaluated using Bioluminescence Resonance Energy Transfer (BRET) experiments, co-immunoprecipitation and confocal microscopy. Our data indicate that Kir 3 channels and Gbetagamma subunits can interact with the GABA-B(1) subunits independently of the GABA-B(2) subunit or Kir 3.4 which are ultimately responsible for their targetting to the cell surface. Thus signalling complexes containing GABA-B receptors, G proteins and Kir channels are formed shortly after biosynthesis most likely in the endoplasmic reticulum.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Receptores de GABA-B/metabolismo , Western Blotting , Linhagem Celular , Transferência Ressonante de Energia de Fluorescência , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Líquido Intracelular/metabolismo , Luciferases/genética , Luciferases/metabolismo , Microscopia Confocal , Plasmídeos/genética , Ligação Proteica , Transporte Proteico/fisiologia , Receptores de GABA-B/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Transfecção
7.
Cell Signal ; 17(1): 49-58, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15451024

RESUMO

The function of beta-adrenergic receptor (betaAR) is modulated by the activity status of alpha1-adrenergic receptors (alpha1ARs) via molecular crosstalk, and this becomes evident when measuring cardiac contractile responses to adrenergic stimulation. The molecular mechanism underlying this crosstalk is unknown. We have previously demonstrated that overexpression of alpha1B-adrenergic receptor (alpha1BAR) in transgenic mice leads to a marked desensitization of betaAR-mediated adenylyl cyclase stimulation which is correlated with increased levels of activated protein kinase C (PKC) beta, delta and [J. Mol. Cell. Cardiol. 30 (1998) 1827]. Therefore, we wished to determine which PKC isoforms play a role in heterologous betaAR desensitization and also which isoforms of the betaAR were the molecular target(s) for PKC. In experiments using constitutively activated PKC expression constructs transfected into HEK 293 cells also expressing the beta2AR, constitutively active (CA)-PKC overexpression was first confirmed by immunoblots using specific anti-PKC antibodies. We then demonstrated that the different PKC subtypes lead to a decreased maximal cAMP accumulation following isoproterenol stimulation with a rank order of PKCalpha > or = PKCzeta>PKC>PKCbetaII. However, a much more dramatic desensitization of adenylyl cyclase stimulation was observed in cells co-transfected with different PKC isoforms and beta1AR. Further, the modulation of beta1AR by PKC isoforms had a different rank order than for the beta2AR: PKCbetaII>PKCalpha>PKC>PKCzeta. PKC-mediated desensitization was reduced by mutating consensus cAMP-dependent protein kinase (PKA)/PKC sites in the third intracellular loop and/or the carboxy-terminal tail of either receptor. Our results demonstrate therefore that the beta1AR is the most likely molecular target for PKC-mediated heterologous desensitization in the mammalian heart and that modulation of adrenergic receptor activity in any given cell type will depend on the complement of PKC isoforms present.


Assuntos
Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Receptores Adrenérgicos beta 1/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Adenilil Ciclases/metabolismo , Animais , Bovinos , Isoproterenol/farmacologia , Cinética , Camundongos , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Mapeamento por Restrição , Especificidade por Substrato , Transfecção
8.
Naunyn Schmiedebergs Arch Pharmacol ; 388(9): 973-81, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25790957

RESUMO

We have previously reported a physiologically relevant interaction between KCNQ1 (Q1) and KCNH2 (H2). While the H2 C-terminus has been suggested to play a role, so far, no more detailed information regarding the interaction site is available. The methods used in the study are cell culture, PCR for mutagenesis, patch clamp for ion current recordings, co-immunoprecipitation for determination of protein interaction. Co-expression of Q1 and H2 resulted in an increase of I H2 (tails after +50 mV; Q1 + H2, 36 ± 6 pA/pF; H2, 14 ± 2 pA/pF; n = 10; 12; P < 0.05). Upon expressing a non-conductive (dominant-negative) Q1-pore mutation (dnQ1), there was still an increase in I H2 (tails after +50 mV; H2 + dnQ1, 24 ± 4 pA/pF; n = 10; P < 0.05) making the pore region unlikely as an interaction site. Experiments using the KCNH2-pore blocking agent quinidine supported these findings. If Q1 and H2 formed hetero-tetramers, steric changes within the pore should change the quinidine half-inhibitory concentrations (IC50). However, I H2 sensitivity did not significantly change in the presence or absence of Q1 (IC50 341 ± 63 vs. 611 ± 293 nmol/L, respectively, P = n.s.), providing further evidence that the pore is not a likely H2-Q1 interaction site. To obtain further insights into the role of intra-cytoplasmic structures, we used both C- and N-terminally truncated mutant H2 proteins. Both H2 mutants co-immunoprecipitated with Q1, suggesting no specific role of C- or N-termini. Accordingly, rather than these, the transmembrane domains of the α-subunits appear relevant for the interaction. Our results largely exclude the formation of hetero-tetramers between H2 and Q1 comprising the pore region or H2 C- or N-termini.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Canal de Potássio KCNQ1/metabolismo , Quinidina/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/química , Células HEK293 , Humanos , Imunoprecipitação , Concentração Inibidora 50 , Canal de Potássio KCNQ1/química , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Quinidina/administração & dosagem
9.
Springerplus ; 3: 337, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25077060

RESUMO

Angiopoietin-like protein 2 (Angptl2) is a secreted glycoprotein that has been implicated in angiogenesis, inflammation and atherosclerosis as well as enhancing the survival of human hematopoietic stem cells. Glycosylation of Angptl2 is required for biological activity and studies of angiopoietin-like protein 2 have been hindered by the lack of a source for the mature form of this protein. We describe a system that permits purification of the glycosylated form of human Angptl2 from conditioned media of stably transfected HEK 293 cells. To facilitate purification while retaining the integrity of Angptl2's endogenous N-terminal secretion signal peptide, GST was fused downstream of the Angptl2 coding sequence. Secreted Angptl2-GST was purified using a one-step glutathione-affinity purification scheme. The purity and identity of the resulting protein were confirmed by SDS-PAGE, immunoblotting, and mass spectrometry. N-Glycosidase treatment reduced the apparent molecular mass of Angptl2-GST on SDS-PAGE, confirming its glycosylation state. Purified human Angptl2-GST stimulated both HUVEC migration and microtubule formation in vitro. The yield of Angptl2-GST obtained was in quantities suitable for multiple applications including functional in vitro and in vivo assays.

10.
Dose Response ; 10(1): 108-19, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22423234

RESUMO

A controlled redox environment is essential for vascular cell maturation and function. During aging, an imbalance occurs, leading to endothelial dysfunction. We hypothesized that, according to the concept of hormesis, exposure to physiologic oxidative stress during the maturation phase of the endothelium will activate protective pathways involved in stress resistance. C57Bl/6 mice were treated with the polyphenol catechin for the last 3 (post-maturation) or 9 months prior study at 12 months of age. Endothelial dysfunction, assessed by acetylcholine-induced dilations of isolated renal arteries, was present at 12 months (P<0.05). Only the 3-month treatment with catechin fully prevented the decline in efficacy and sensitivity to acetylcholine (P<0.05). Splenocytes adhesion to the native endothelium, expression of CD18 and shedding of CD62L and PSGL-1 augmented in 12 months old mice (P<0.05): only 3-month catechin fully normalized adhesion and prevented the expression of adhesion molecules on splenocytes (P<0.05). Aging was associated with vascular gene alterations, which were prevented by 3-month catechin treatment (P<0.05). In contrast, 9-month catechin further increased COX-2, p22(phox) and reduced MnSOD (P<0.05). In conclusion, we demonstrate a pivotal role of cellular redox equilibrium: exposure to physiologic oxidative stress during the maturation phase of the endothelium is essential for its function.

11.
Obesity (Silver Spring) ; 19(4): 722-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20814418

RESUMO

Adiponectin is an adipokine whose plasma levels are inversely correlated to metabolic syndrome components. Adiponectin protects against atherosclerosis and decreases risks in myocardial infarction. Endothelial progenitor cells (EPCs) are a heterogeneous population of circulating cells involved in vascular repair and neovascularization. EPCs number is reduced in patients with cardiovascular disease. We hypothesize that the positive effects of adiponectin against atherosclerosis are explained in part by its interactions with EPCs. Cells were obtained from healthy volunteers' blood by mononuclear cell isolation and plating on collagen-coated dishes. Three sub-populations of EPCs were identified and characterized using flow cytometry. EPCs' expression of adiponectin receptors, AdipoR1, and AdipoR2 was evaluated by quantitative PCR. The effects of recombinant adiponectin on EPCs' susceptibility to apoptosis were assessed. Finally, expression of neutrophil elastase by EPCs and activity of this enzyme on adiponectin processing were assessed. Quantitative PCR analysis of EPCs mRNAs showed that AdipoR1 mRNA is expressed at higher levels than AdipoR2. Expression of AdipoR1 protein was confirmed by western blot. Adiponectin significantly increased survival of two sub-populations of EPCs in conditions of serum deprivation. Such effect could not be demonstrated in the third EPCs sub-population. We also demonstrated that EPCs, particularly one sub-population, express neutrophil elastase. Neutrophil elastase activity was confirmed in EPCs' conditioned media. Adiponectin protects some EPCs sub-populations against apoptosis and therefore could modulate EPCs ability to induce repair of vascular damage. Neutrophil elastase activity of EPCs could locally modulate adiponectin activity by its involvement in the generation of the globular form of adiponectin.


Assuntos
Adiponectina/sangue , Adiponectina/farmacologia , Células Endoteliais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Análise de Variância , Apoptose/efeitos dos fármacos , Análise Química do Sangue/métodos , Western Blotting , Separação Celular , Células Cultivadas , Meios de Cultivo Condicionados , Células Endoteliais/metabolismo , Feminino , Citometria de Fluxo , Humanos , Elastase de Leucócito/metabolismo , Masculino , Neovascularização Patológica/metabolismo , RNA Mensageiro/análise , Receptores de Adiponectina/metabolismo , Células-Tronco/metabolismo
12.
Cell Signal ; 22(10): 1502-12, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20570725

RESUMO

p38 MAP kinase (MAPK) isoforms alpha, beta, and gamma, are expressed in the heart. p38alpha appears pro-apoptotic whereas p38beta is pro-hypertrophic. The mechanisms mediating these divergent effects are unknown; hence elucidating the downstream signaling of p38 should further our understanding. Downstream effectors include MAPK-activated protein kinase (MK)-3, which is expressed in many tissues including skeletal muscles and heart. We cloned full-length MK3 (MK3.1, 384 aa) and a novel splice variant (MK3.2, 266 aa) from murine heart. For MK3.2, skipping of exons 8 and 9 resulted in a frame-shift in translation of the first 85 base pairs of exon 10 followed by an in-frame stop codon. Of 3 putative phosphorylation sites for p38 MAPK, only Thr-203 remained functional in MK3.2. In addition, MK3.2 lacked nuclear localization and export signals. Quantitative real-time PCR confirmed the presence of these mRNA species in heart and skeletal muscle; however, the relative abundance of MK3.2 differed. Furthermore, whereas total MK3 mRNA was increased, the relative abundance of MK3.2 mRNA decreased in MK2(-/-) mice. Immunoblotting revealed 2 bands of MK3 immunoreactivity in ventricular lysates. Ectopically expressed MK3.1 localized to the nucleus whereas MK3.2 was distributed throughout the cell; however, whereas MK3.1 translocated to the cytoplasm in response to osmotic stress, MK3.2 was degraded. The p38alpha/beta inhibitor SB203580 prevented the degradation of MK3.2. Furthermore, replacing Thr-203 with alanine prevented the loss of MK3.2 following osmotic stress, as did pretreatment with the proteosome inhibitor MG132. In vitro, GST-MK3.1 was strongly phosphorylated by p38alpha and p38beta, but a poor substrate for p38delta and p38gamma. GST-MK3.2 was poorly phosphorylated by p38alpha and p38beta and not phosphorylated by p38delta and p38gamma. Hence, differential regulation of MKs may, in part, explain diverse downstream effects mediated by p38 signaling.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Miocárdio/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Processamento Alternativo , Sequência de Aminoácidos , Animais , Clonagem Molecular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Ventrículos do Coração/citologia , Peptídeos e Proteínas de Sinalização Intracelular/análise , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Dados de Sequência Molecular , Proteínas Serina-Treonina Quinases/análise , Proteínas Serina-Treonina Quinases/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Cell Signal ; 22(11): 1634-44, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20600854

RESUMO

p38 mitogen-activated protein kinases (MAPKs) are serine/threonine specific protein kinases that respond to cellular stress and regulate a broad range of cellular activities. There are four major isoforms of p38 MAPK: alpha, beta, gamma, and delta. To date, the prominent isoform in heart has been thought to be p38alpha. We examined the expression of each p38 isoform at both the mRNA and protein level in murine heart. mRNA for all four p38 isoforms was detected. p38gamma and p38delta were expressed at protein levels comparable to p38alpha and 38beta, respectively. In the early phase of pressure-overload hypertrophy (1-7 days after constriction of the transverse aorta), the abundance of p38beta, p38gamma and p38delta mRNA increased; however, no corresponding changes were detected at the protein level. Confocal immunofluorescence studies revealed p38alpha and p38gamma in both the cytoplasm and nucleus. In the established phase of hypertrophy induced by chronic pressure overload (7-28 days after constriction of the transverse aorta), p38gamma immunoreactivity accumulated in the nucleus whereas the distribution of p38alpha remained unaffected. Hence, both p38alpha and p38gamma are prominent p38 isoforms in heart and p38gamma may play a role in mediating the changes in gene expression associated with cardiac remodeling during pressure-overload hypertrophy.


Assuntos
Cardiomegalia/enzimologia , Miocárdio/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Humanos , Isoenzimas/análise , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Pressão , RNA Mensageiro/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/análise , Proteínas Quinases p38 Ativadas por Mitógeno/genética
14.
Mech Ageing Dev ; 131(5): 354-63, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20399802

RESUMO

INTRODUCTION: With aging, oxidative stress accelerates vascular endothelial cell (EC) telomere shortening-induced senescence, and may promote atherosclerosis in humans. Our aim was to investigate whether an antioxidant treatment combined with telomerase (hTERT) over-expression would prevent senescence of EC isolated from patients with severe atherosclerosis. METHODS: Cells were isolated from internal mammary arteries (n=11 donors), cultured until senescence with or without N-acetylcystein (NAC) and infected, or not, with a lentivirus over-expressing hTERT. RESULTS: Compared to control EC, hTERT-NAC cells had increased telomerase activity, longer telomeres and underwent more cell divisions. According to the donor, hTERT-NAC either delayed (n=5) or prevented (n=4) EC senescence, the latter leading to cell immortalization. Lack of cell immortalization by hTERT-NAC was accompanied by an absence of beneficial effect of NAC alone in paired EC. Accordingly, lack of EC immortalization by hTERT-NAC was associated with high endogenous susceptibility to oxidation. In EC where hTERT-NAC did not immortalize EC, p53, p21 and p16 expression increased with senescence, while oxidative-dependent DNA damage associated with senescence was not prevented. CONCLUSION: Our data suggest that irreversible oxidative stress-dependent damages associated with cardiovascular risk factors are responsible for senescence of EC from atherosclerotic patients.


Assuntos
Aterosclerose/fisiopatologia , Senescência Celular , Células Endoteliais/enzimologia , Estresse Oxidativo , Telomerase/metabolismo , Acetilcisteína/farmacologia , Idoso , Aterosclerose/enzimologia , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Células Endoteliais/efeitos dos fármacos , Feminino , Sequestradores de Radicais Livres/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Risco , Proteína Supressora de Tumor p53/metabolismo
15.
Cell Signal ; 22(7): 1063-75, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20214976

RESUMO

MK5, a member of the MAPK-activated protein kinase family, is highly expressed in the heart. Whereas MK2 and MK3 are activated by p38 MAPK, MK5 has also been shown to be activated by ERK3 and ERK4. We studied the regulation of MK5 in mouse heart. mRNA for 5 splice variants (MK5.1-5.5), including the original form (MK5.1), was detected. MK5 comprises 14 exons: exon 12 splicing was modified in MK5.2, MK5.3, and MK5.5. MK5.2 and MK5.5 lacked 6 bases at the 3'-end of exon 12, whereas MK5.3 lacked exon 12, resulting in a frame shift and premature termination of translation at codon 3 of exon 13. MK5.4 and MK5.5 lacked exons 2-6, encoding kinase subdomains I-VI, and were kinase-dead. All 5 MK5 variants were detected at the mRNA level in all mouse tissues examined; however, their relative abundance was tissue-specific. Furthermore, the relative abundance of variant mRNA was altered both during hypertrophy and postnatal cardiac development, suggesting that the generation or the stability of MK5 variant mRNAs is subject to regulation. When expressed in HEK293 cells, MK5.1, MK5.2 and MK5.3 were nuclear whereas MK5.4 and MK5.5 were cytoplasmic. A p38 MAPK activator, anisomycin, induced the redistribution of each variant. In contrast, MK5 co-immunoprecipitated ERK3, but not ERK4 or p38 alpha, in control and hypertrophying hearts. GST pull-down assays revealed unbound ERK4 and p38 alpha but no free MK5 or ERK3 in heart lysates. Hence, 1) in heart MK5 complexes with ERK3 and 2) MK5 splice variants may mediate distinct effects thus increasing the functional diversity of ERK3-MK5 signaling.


Assuntos
Miocárdio/enzimologia , Proteínas Serina-Treonina Quinases/biossíntese , Processamento Alternativo , Sequência de Aminoácidos , Animais , Cardiomegalia/enzimologia , Cardiomegalia/genética , Linhagem Celular , Clonagem Molecular , Coração/crescimento & desenvolvimento , Ventrículos do Coração/enzimologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/metabolismo
16.
J Biol Chem ; 282(18): 13703-15, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17363375

RESUMO

Heterotrimeric G proteins play a central role in intracellular communication mediated by extracellular signals, and both Galpha and Gbetagamma subunits regulate effectors downstream of activated receptors. The particular constituents of the G protein heterotrimer affect both specificity and efficiency of signal transduction. However, little is known about mechanistic aspects of G protein assembly in the cell that would certainly contribute to formation of heterotrimers of specific composition. It was recently shown that phosducin-like protein (PhLP) modulated both Gbetagamma expression and subsequent signaling by chaperoning nascent Gbeta and facilitating heterodimer formation with Ggamma subunits (Lukov, G. L., Hu, T., McLaughlin, J. N., Hamm, H. E., and Willardson, B. M. (2005) EMBO J. 24, 1965-1975; Humrich, J., Bermel, C., Bunemann, M., Harmark, L., Frost, R., Quitterer, U., and Lohse, M. J. (2005) J. Biol. Chem. 280, 20042-20050). Here we demonstrate using a variety of techniques that DRiP78, an endoplasmic reticulum resident protein known to regulate the trafficking of several seven transmembrane receptors, interacts specifically with the Ggamma subunit but not Gbeta or Galpha subunits. Furthermore, we demonstrate that DRiP78 and the Gbeta subunit can compete for the Ggamma subunit. DRiP78 also protects Ggamma from degradation until a stable partner such as Gbeta is provided. Furthermore, DRiP78 interaction may represent a mechanism for assembly of specific Gbetagamma heterodimers, as selectivity was observed among Ggamma isoforms for interaction with DRiP78 depending on the presence of particular Gbeta subunits. Interestingly, we could detect an interaction between DRiP78 and PhLP, suggesting a role of DRiP78 in the assembly of Gbetagamma by linking Ggamma to PhLP.Gbeta complexes. Our results, therefore, suggest a role of DRiP78 as a chaperone in the assembly of Gbetagamma subunits of the G protein.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/biossíntese , Subunidades gama da Proteína de Ligação ao GTP/biossíntese , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Complexos Multiproteicos/biossíntese , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Proteínas Fetais , Subunidades alfa de Proteínas de Ligação ao GTP/biossíntese , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Proteínas de Membrana/genética , Chaperonas Moleculares/genética , Complexos Multiproteicos/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica/fisiologia , Transporte Proteico/fisiologia
17.
J Biol Chem ; 281(45): 34561-73, 2006 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-16959776

RESUMO

Much is known about beta2-adrenergic receptor trafficking and internalization following prolonged agonist stimulation. However, less is known about outward trafficking of the beta2-adrenergic receptor to the plasma membrane or the role that trafficking might play in the assembly of receptor signaling complexes, important for targeting, specificity, and rapidity of subsequent signaling events. Here, by using a combination of bioluminescence resonance energy transfer, bimolecular fluorescence complementation, and confocal microscopy, we evaluated the steps in the formation of the core receptor-G protein heterotrimer complex. By using dominant negative Rab and Sar GTPase constructs, we demonstrate that receptor dimers and receptor-G betagamma complexes initially associate in the endoplasmic reticulum, whereas G alpha subunits are added to the complex during endoplasmic reticulum-Golgi transit. We also observed that G protein heterotrimers adopt different trafficking itineraries when expressed alone or with stoichiometric co-expression with receptor. Furthermore, deliberate mistargeting of specific components of these complexes leads to diversion of other members from their normal subcellular localization, confirming the role of these early interactions in targeting and formation of specific signaling complexes.


Assuntos
Membrana Celular/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Transporte Proteico , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Dimerização , Retículo Endoplasmático/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Genes Dominantes , Complexo de Golgi/metabolismo , Humanos , Imunoprecipitação , Membranas Intracelulares/metabolismo , Rim/citologia , Rim/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Ligação Proteica , Ensaio Radioligante , Ratos , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA