Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 6: 168, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26321954

RESUMO

Histone deacetylase inhibitors (HDIs) are under investigation for the treatment of a number of human health problems. HDIs have proven therapeutic value in refractory cases of cutaneous T-cell lymphoma. Electrocardiographic ST segment morphological changes associated with HDIs were observed during development. Because ST segment morphology is typically linked to changes in ATP sensitive potassium (KATP) channel activity, we tested the hypothesis that HDIs affect cardiac KATP channel subunit expression. Two different HDIs, romidepsin and trichostatin A, caused ~20-fold increase in SUR2 (Abcc9) subunit mRNA expression in HL-1 cardiomyocytes. The effect was specific for the SUR2 subunit as neither compound causes a marked change in SUR1 (Abcc8) expression. Moreover, the effect was cell specific as neither HDI markedly altered KATP subunit expression in MIN6 pancreatic ß-cells. We observe significant enrichment of the H3K9Ac histone mark specifically at the SUR2 promoter consistent with the conclusion that chromatin remodeling at this locus plays a role in increasing SUR2 gene expression. Unexpectedly, however, we also discovered that HDI-dependent depletion of cellular cholesterol is required for the observed effects on SUR2 expression. Taken together, the data in the present study demonstrate that KATP subunit expression can be epigenetically regulated in cardiomyocytes, defines a role for cholesterol homeostasis in mediating epigenetic regulation and suggests a potential molecular basis for the cardiac effects of the HDIs.

2.
Am J Respir Cell Mol Biol ; 52(4): 492-502, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25188881

RESUMO

Autopsy specimens from human victims or experimental animals that die due to acute chlorine gas exposure present features of cardiovascular pathology. We demonstrate acute chlorine inhalation-induced reduction in heart rate and oxygen saturation in rats. Chlorine inhalation elevated chlorine reactants, such as chlorotyrosine and chloramine, in blood plasma. Using heart tissue and primary cardiomyocytes, we demonstrated that acute high-concentration chlorine exposure in vivo (500 ppm for 30 min) caused decreased total ATP content and loss of sarcoendoplasmic reticulum calcium ATPase (SERCA) activity. Loss of SERCA activity was attributed to chlorination of tyrosine residues and oxidation of an important cysteine residue, cysteine-674, in SERCA, as demonstrated by immunoblots and mass spectrometry. Using cardiomyocytes, we found that chlorine-induced cell death and damage to SERCA could be decreased by thiocyanate, an important biological antioxidant, and by genetic SERCA2 overexpression. We also investigated a U.S. Food and Drug Administration-approved drug, ranolazine, used in treatment of cardiac diseases, and previously shown to stabilize SERCA in animal models of ischemia-reperfusion. Pretreatment with ranolazine or istaroxime, another SERCA activator, prevented chlorine-induced cardiomyocyte death. Further investigation of responsible mechanisms showed that ranolazine- and istaroxime-treated cells preserved mitochondrial membrane potential and ATP after chlorine exposure. Thus, these studies demonstrate a novel critical target for chlorine in the heart and identify potentially useful therapies to mitigate toxicity of acute chlorine exposure.


Assuntos
Cloro/toxicidade , Cardiopatias/enzimologia , Exposição por Inalação , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Antioxidantes/farmacologia , Apoptose , Sinalização do Cálcio , Cardiotônicos/farmacologia , Células Cultivadas , Etiocolanolona/análogos & derivados , Etiocolanolona/farmacologia , Cardiopatias/induzido quimicamente , Masculino , Mitocôndrias Cardíacas , Miocárdio/enzimologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ranolazina/farmacologia , Ratos Sprague-Dawley , Tiocianatos/farmacologia
3.
J Cell Mol Med ; 16(8): 1709-19, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21992629

RESUMO

Recently the protein myozap, a 54-kD polypeptide which is not a member of any of the known cytoskeletal and junctional protein multigene families, has been identified as a constituent of the plaques of the composite junctions in the intercalated disks connecting the cardiomyocytes of mammalian hearts. Using a set of novel, highly sensitive and specific antibodies we now report that myozap is also a major constituent of the cytoplasmic plaques of the adherens junctions (AJs) connecting the endothelial cells of the mammalian blood and lymph vascular systems, including the desmoplakin-containing complexus adhaerentes of the virgultar cells of lymph node sinus. In light and electron microscopic immunolocalization experiments we show that myozap colocalizes with several proteins of desmosomal plaques as well as with AJ-specific transmembrane molecules, including VE-cadherin. In biochemical analyses, rigorous immunoprecipitation experiments have revealed N-cadherin, desmoplakin, desmoglein-2, plakophilin-2, plakoglobin and plectin as very stably bound complex partners. We conclude that myozap is a general component of cell-cell junctions not only in the myocardium but also in diverse endothelia of the blood and lymph vascular systems of adult mammals, suggesting that this protein not only serves a specific role in the heart but also a broader set of functions in the vessel systems. We also propose to use myozap as an endothelial cell type marker in diagnoses.


Assuntos
Junções Aderentes/metabolismo , Endotélio Vascular/metabolismo , Linfa/metabolismo , Vasos Linfáticos/metabolismo , Proteínas de Membrana/sangue , Proteínas de Membrana/metabolismo , Animais , Especificidade de Anticorpos/imunologia , Bovinos , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Células Endoteliais/metabolismo , Endotélio Vascular/ultraestrutura , Imunofluorescência , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/ultraestrutura , Ratos , Sus scrofa
4.
Mol Biol Cell ; 21(22): 3952-62, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20861308

RESUMO

Ablation of nonmuscle myosin (NM) II-A or NM II-B results in mouse embryonic lethality. Here, we report the results of ablating NM II-C as well as NM II-C/II-B together in mice. NM II-C ablated mice survive to adulthood and show no obvious defects compared with wild-type littermates. However, ablation of NM II-C in mice expressing only 12% of wild-type amounts of NM II-B results in a marked increase in cardiac myocyte hypertrophy compared with the NM II-B hypomorphic mice alone. In addition, these hearts develop interstitial fibrosis associated with diffuse N-cadherin and ß-catenin localization at the intercalated discs, where both NM II-B and II-C are normally concentrated. When both NM II-C and II-B are ablated the B-C-/B-C- cardiac myocytes show major defects in karyokinesis. More than 90% of B-C-/B-C- myocytes demonstrate defects in chromatid segregation and mitotic spindle formation accompanied by increased stability of microtubules and abnormal formation of multiple centrosomes. This requirement for NM II in karyokinesis is further demonstrated in the HL-1 cell line derived from mouse atrial myocytes, by using small interfering RNA knockdown of NM II or treatment with the myosin inhibitor blebbistatin. Our study shows that NM II is involved in regulating cardiac myocyte karyokinesis by affecting microtubule dynamics.


Assuntos
Divisão do Núcleo Celular/fisiologia , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo II/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Animais , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Divisão do Núcleo Celular/genética , Chlorocebus aethiops , Feminino , Células HT29 , Células HeLa , Coração/embriologia , Coração/crescimento & desenvolvimento , Humanos , Pulmão/embriologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microtúbulos/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Cadeias Pesadas de Miosina/genética , Miosina Tipo II/genética , Miosina não Muscular Tipo IIB/genética , Interferência de RNA
5.
J Biol Chem ; 285(26): 20369-80, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20424170

RESUMO

The molecular mechanism underlying the post-Golgi transport of G protein-coupled receptors (GPCRs) remains poorly understood. Here we determine the role of Rab8 GTPase, which modulates vesicular protein transport between the trans-Golgi network (TGN) and the plasma membrane, in the cell surface targeting of alpha(2B)- and beta(2)-adrenergic receptors (AR). Transient expression of GDP- and GTP-bound Rab8 mutants and short hairpin RNA-mediated knockdown of Rab8 more potently inhibited the cell surface expression of alpha(2B)-AR than beta(2)-AR. The GDP-bound Rab8(T22N) mutant attenuated ERK1/2 activation by alpha(2B)-AR, but not beta(2)-AR, and arrested alpha(2B)-AR in the TGN compartment. Co-immunoprecipitation revealed that both alpha(2B)-AR and beta(2)-AR physically interacted with Rab8 and glutathione S-transferase fusion protein pulldown assays demonstrated that Rab8 interacted with the C termini of both receptors. Interestingly, mutation of the highly conserved membrane-proximal C terminus dileucine motif selectively blocked beta(2)-AR interaction with Rab8, whereas mutation of residues Val(431)-Phe(432)-Asn(433)-Gln(434), Pro(447)-Trp(448), Gln(450)-Thr(451), and Trp(453) in the C terminus impaired alpha(2B)-AR interaction with Rab8. Furthermore, transport inhibition by Rab8(T22N) of a chimeric beta(2)-AR carrying the alpha(2B)-AR C terminus was similar to alpha(2B)-AR. These data provide strong evidence indicating that Rab8 GTPase interacts with distinct motifs in the C termini of alpha(2B)-AR and beta(2)-AR and differentially modulates their traffic from the TGN to the cell surface.


Assuntos
Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Motivos de Aminoácidos , Animais , Sítios de Ligação , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Immunoblotting , Microscopia de Fluorescência , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mutação , Ligação Proteica , Transporte Proteico , Interferência de RNA , Ratos , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos beta 2/genética , Transfecção , Proteínas rab de Ligação ao GTP/genética , Rede trans-Golgi/metabolismo
6.
Crit Care Med ; 38(3): 903-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20081527

RESUMO

OBJECTIVE: To test whether preconditioning with a toll-like receptor (TLR) 2 agonist protects against myocardial ischemia and reperfusion by interfering with chemokine CXCL1 release from cardiomyocytes. DESIGN: C3H mice were challenged with vehicle or synthetic TLR2 agonist Pam3Cys-Ser-Lys4 (Pam3CSK4; 1 mg/kg) 24 hrs before myocardial ischemia (20 mins) and reperfusion (2 hrs or 24 hrs). Infarct size, troponin T release, and leukocyte recruitment were quantified. In murine cardiomyocytes (HL-1), we studied the expression/activation profile of TLR2 in response to stimulation with Pam3CSK4 (0.01-1 mg/mL). Furthermore, we studied the chemokine ligand 1 (CXCL1) response to Pam3CSK4 and ischemia/reperfusion in vivo and in vitro. SETTING: University hospital research laboratory. SUBJECTS: Anesthetized male mice and murine cardiomyocytes. MEASUREMENTS AND MAIN RESULTS: Preconditioning by Pam3CSK4 reduced infarct size and troponin T release. This was accompanied by a decreased recruitment of leukocytes into the ischemic area and an improved cardiac function. In HL-1 cells, TLR2 activation amplified the expression of the receptor in a time-dependent manner and led to CXCL1 release in a concentration-dependent manner. Preconditioning by Pam3CSK4 impaired CXCL1 release in response to a second inflammatory stimulus in vivo and in vitro. CONCLUSIONS: Preconditioning by TLR2 agonist Pam3CSK4 reduces myocardial infarct size after myocardial ischemia/reperfusion. One of the mechanisms involved is a diminished chemokine release from cardiomyocytes, which subsequently limits leukocyte infiltration.


Assuntos
Quimiocina CXCL1/fisiologia , Precondicionamento Isquêmico Miocárdico , Leucócitos/imunologia , Lipopeptídeos/farmacologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/imunologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/imunologia , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/genética , Animais , Linhagem Celular , Quimiocina CXCL1/antagonistas & inibidores , Quimiocinas/metabolismo , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C3H , Infarto do Miocárdio/imunologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Troponina T/metabolismo
7.
J Mol Endocrinol ; 42(6): 479-89, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19318426

RESUMO

Despite our knowledge on the regulation of urocortin (Ucn) I and its related peptides in the heart, the possible involvement of cardiovascular stress substances, such as cytokines or angiotensin II (Ang II), on this regulation remains to be fully elucidated. We therefore evaluated the potential role of cardiovascular stress substances on the regulation of the Ucn-corticotropin-releasing hormone (CRH) receptor system in HL-1 cardiomyocytes using a Ucn I-specific RIA, conventional reverse transcription-PCR (RT-PCR) and quantitative real-time RT-PCR. Ucn I mRNA levels were shown to be up-regulated by lipopolysaccarides (LPS), tumor necrosis factor-alpha (TNF-alpha), Ang II, H(2)O(2), and pyrrolidinedithiocarbamate (PDTC). The LPS- and Ang II-induced increase in Ucn I mRNA levels was abolished by tempol. In addition, the secretion of Ucn I from HL-1 cardiomyocytes was stimulated by LPS and TNF-alpha. On the contrary, Ucn II mRNA was increased by TNF-alpha alone and Ang II with tempol, and the TNF-alpha-induced increase in Ucn II mRNA was abolished by erythromycin and PDTC. These results suggested that Ucn I mRNA may be up-regulated by oxidative stress, whereas Ucn II mRNA may be up-regulated by the activated nuclear factor-kappaB, i.e. inflammatory stress. CRH-R2 mRNA may be negatively regulated by the increase in expression of Ucn I and/or Ucn II mRNA. In conclusion, the Ucn-CRH receptor system may be regulated by two major forms of cardiac stresses, i.e. oxidative and inflammatory stress, and may play a critical role in cardiac stress adaptation in heart diseases.


Assuntos
Inflamação/fisiopatologia , Miocárdio/metabolismo , Estresse Oxidativo , Urocortinas/fisiologia , Angiotensina II/farmacologia , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , DNA Complementar , Lipopolissacarídeos/farmacologia , Camundongos , Miocárdio/citologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/farmacologia , Urocortinas/genética
8.
Exp Biol Med (Maywood) ; 234(5): 582-94, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19244544

RESUMO

AIMS: Reactive oxygen species (ROS) activate multiple signaling pathways involved in cardiac hypertrophy. Since HO-1 exerts potent antioxidant effects, we hypothesized that this enzyme inhibits ROS-induced cardiomyocyte hypertrophy. METHODS: HL-1 cardiomyocytes were transduced with an adenovirus constitutively expressing HO-1 (AdHO-1) to increase basal HO-1 expression and then exposed to 200 microM hydrogen peroxide (H2O2). Hypertrophy was measured using 3H-leucine incorporation, planar morphometry and cell-size by forward-scatter flow-cytometry. The pro-oxidant effect of H2O2 was assessed by redox sensitive fluorophores. Inducing intracellular redox imbalance resulted in cardiomyocyte hypertrophy through transactivation of nuclear factor kappa B (NF-kappaB). RESULTS: Pre-emptive HO-1 overexpression attenuated the redox imbalance and reduced hypertrophic indices. This is the first time that HO-1 has directly been shown to inhibit oxidant-induced cardiomyocyte hypertrophy by a NF-kappaB-dependent mechanism. CONCLUSION: These results demonstrate that HO-1 inhibits pro-oxidant induced cardiomyocyte hypertrophy and suggest that HO-1 may yield therapeutic potential in treatment of.


Assuntos
Cardiomegalia/enzimologia , Heme Oxigenase (Desciclizante)/metabolismo , Peróxido de Hidrogênio/farmacologia , Miócitos Cardíacos/enzimologia , Oxidantes/farmacologia , Adenoviridae , Animais , Cardiomegalia/genética , Cardiomegalia/terapia , Linhagem Celular , Heme Oxigenase (Desciclizante)/genética , NF-kappa B/metabolismo , Oxirredução , Ratos , Transdução Genética
9.
Circ Res ; 100(6): 874-83, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17322175

RESUMO

Ca(2+) release via type 2 ryanodine receptors (RyR2) regulates cardiac function. Molecular cloning of human RyR2 identified 2 alternatively spliced variants, comprising 30- and 24-bp sequence insertions; yet their role in shaping cardiomyocyte Ca(2+) signaling and cell phenotype is unknown. We profiled the developmental regulation and the tissue and species specificity of these variants and showed that their recombinant expression in HL-1 cardiomyocytes profoundly modulated nuclear and cytoplasmic Ca(2+) release. All splice variants localized to the sarcoplasmic reticulum, perinuclear Golgi apparatus, and to finger-like invaginations of the nuclear envelope (nucleoplasmic reticulum). Strikingly, the 24-bp splice insertion that was present at low levels in embryonic and adult hearts was essential for targeting RyR2 to an intranuclear Golgi apparatus and promoted the intracellular segregation of this variant. The amplitude variability of nuclear and cytoplasmic Ca(2+) fluxes were reduced in nonstimulated cardiomyocytes expressing both 30- and 24-bp splice variants and were associated with lower basal levels of apoptosis. Expression of RyR2 containing the 24-bp insertion also suppressed intracellular Ca(2+) fluxes following prolonged caffeine exposure (1 mmol/L, 16 hours) that protected cells from apoptosis. The antiapoptotic effects of this variant were linked to increased levels of Bcl-2 phosphorylation. In contrast, RyR2 containing the 30-bp insertion, which was abundant in human embryonic heart but was decreased during cardiac development, did not protect cardiomyocytes from caffeine-evoked apoptosis. Thus, we provide the first evidence that RyR2 splice variants exquisitely modulate intracellular Ca(2+) signaling and are key determinants of cardiomyocyte apoptotic susceptibility.


Assuntos
Processamento Alternativo/genética , Apoptose/genética , Sinalização do Cálcio/fisiologia , Miócitos Cardíacos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cafeína/farmacologia , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Transferência de Genes , Humanos , Camundongos , Dados de Sequência Molecular , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/metabolismo , Especificidade da Espécie
10.
Circ Res ; 99(5): 510-9, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16873718

RESUMO

Adenosine plays multiple roles in the efficient functioning of the heart by regulating coronary blood flow, cardiac pacemaking, and contractility. Previous studies have implicated the equilibrative nucleoside transporter family member equilibrative nucleoside transporter-1 (ENT1) in the regulation of cardiac adenosine levels. We report here that a second member of this family, ENT4, is also abundant in the heart, in particular in the plasma membranes of ventricular myocytes and vascular endothelial cells but, unlike ENT1, is virtually absent from the sinoatrial and atrioventricular nodes. Originally described as a monoamine/organic cation transporter, we found that both human and mouse ENT4 exhibited a novel, pH-dependent adenosine transport activity optimal at acidic pH (apparent K(m) values 0.78 and 0.13 mmol/L, respectively, at pH 5.5) and absent at pH 7.4. In contrast, serotonin transport by ENT4 was relatively insensitive to pH. ENT4-mediated nucleoside transport was adenosine selective, sodium independent and only weakly inhibited by the classical inhibitors of equilibrative nucleoside transport, dipyridamole, dilazep, and nitrobenzylthioinosine. We hypothesize that ENT4, in addition to playing roles in cardiac serotonin transport, contributes to the regulation of extracellular adenosine concentrations, in particular under the acidotic conditions associated with ischemia.


Assuntos
Ácidos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Miocárdio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Adenosina/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Proteínas de Transporte de Nucleosídeo Equilibrativas , Glicosilação , Humanos , Concentração de Íons de Hidrogênio , Cinética , Camundongos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Nucleosídeos/metabolismo , Oócitos , Serotonina/metabolismo , Frações Subcelulares/metabolismo , Distribuição Tecidual , Xenopus
11.
J Biol Chem ; 281(16): 11097-103, 2006 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-16484224

RESUMO

We investigate the role of Rab4, a Ras-like small GTPase coordinating protein transport from the endosome to the plasma membrane, on the recycling and activation of endogenous beta-adrenergic receptor (beta-AR) in HL-1 cardiac myocytes in vitro and transgenic mouse hearts in vivo. Beta1-AR, the predominant subtype of beta-AR in HL-1 cardiac myocytes, was internalized after stimulation with isoproterenol (ISO) and fully recycled at 4 h upon ISO removal. Transient expression of Rab4 markedly facilitated recycling of internalized beta-AR to the cell surface and enhanced beta-AR signaling as measured by ISO-stimulated cAMP production. Transgenic overexpression of Rab4 in the mouse myocardium significantly increased the number of beta-AR in the plasma membrane and augmented cAMP production at the basal level and in response to ISO stimulation. Rab4 overexpression induced concentric cardiac hypertrophy with a moderate increase in ventricle/body weight ratio and posterior wall thickness and a selective up-regulation of the beta-myosin heavy chain gene. These data provide the first evidence indicating that Rab4 is a rate-limiting factor for the recycling of endogenous beta-AR and augmentation of Rab4-mediated traffic enhances beta-AR function in cardiac myocytes.


Assuntos
Miócitos Cardíacos/enzimologia , Receptores Adrenérgicos beta/metabolismo , Proteínas rab4 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Ecocardiografia , Eletroforese em Gel de Poliacrilamida , Immunoblotting , Isoproterenol/farmacologia , Ligantes , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fenótipo , Fatores de Tempo , Transfecção , Transgenes , Regulação para Cima , Miosinas Ventriculares/química
12.
J Biol Chem ; 280(6): 4553-67, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15574430

RESUMO

In patients with congestive heart failure, high serum levels of the proinflammatory cytokine interleukin (IL)-18 were reported. A positive correlation was described between serum IL-18 levels and the disease severity. IL-18 has also been shown to induce atrial natriuretic factor (ANF) gene expression in adult cardiomyocytes. Because re-expression of the fetal gene ANF is mostly associated with hypertrophy, a hallmark of heart failure, we hypothesized that IL-18 induces cardiomyocyte hypertrophy. Treatment of the cardiomyocyte cell line HL-1 with IL-18 induced hypertrophy as characterized by increases in protein synthesis, phosphorylated p70 S6 kinase, and ribosomal S6 protein levels as well as cell surface area. Furthermore, IL-18 induced ANF gene transcription in a time-dependent manner as evidenced by increased ANF secretion and ANF promoter-driven reporter gene activity. Investigation into possible signal transduction pathways mediating IL-18 effects revealed that IL-18 activates phosphoinositide 3-kinase (PI3K), an effect that was blocked by wortmannin and LY-294002. IL-18 induced Akt phosphorylation and stimulated its activity, effects that were abolished by Akt inhibitor or knockdown. IL-18 stimulated GATA4 DNA binding activity and increased transcription of a reporter gene driven by multimerized GATA4-binding DNA elements. Pharmacological inhibition or knockdown studies revealed that IL-18 induced cardiomyocyte hypertrophy and ANF gene transcription via PI3K, PDK1, Akt, and GATA4. Most importantly, IL-18 induced ANF gene transcription and hypertrophy of neonatal rat ventricular myocytes via PI3K-, Akt-, and GATA4-dependent signaling. Together these data provide the first evidence that IL-18 induces cardiomyocyte hypertrophy via PI3K-dependent signaling, defines a mechanism of IL-18-mediated ANF gene transcription, and further supports a role for IL-18 in inflammatory heart diseases including heart failure.


Assuntos
Citocinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Interleucina-18/fisiologia , Miócitos Cardíacos/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Androstadienos/farmacologia , Animais , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Northern Blotting , Western Blotting , Linhagem Celular , Núcleo Celular/metabolismo , Células Cultivadas , Cromonas/farmacologia , DNA/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fator de Transcrição GATA4 , Regulação da Expressão Gênica , Genes Reporter , Hipertrofia , Inflamação , Interleucina-18/metabolismo , Subunidade alfa de Receptor de Interleucina-18 , Camundongos , Morfolinas/farmacologia , Miócitos Cardíacos/metabolismo , Fenótipo , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt , Ratos , Receptores de Interleucina/metabolismo , Receptores de Interleucina-18 , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção , Wortmanina
13.
J Biol Chem ; 279(39): 41077-84, 2004 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-15252015

RESUMO

Rab1 GTPase coordinates vesicle-mediated protein transport specifically from the endoplasmic reticulum (ER) to the Golgi apparatus. We recently demonstrated that Rab1 is involved in the export of angiotensin II (Ang II) type 1 receptor (AT1R) to the cell surface in HEK293 cells and that transgenic mice overexpressing Rab1 in the myocardium develop cardiac hypertrophy. To expand these studies, we determined in this report whether the modification of Rab1-mediated ER-to-Golgi transport can alter the cell surface expression and function of endogenous AT1R and AT1R-mediated hypertrophic growth in primary cultures of neonatal rat ventricular myocytes. Adenovirus-mediated gene transfer of wild-type Rab1 (Rab1WT) significantly increased cell surface expression of endogenous AT1R in neonatal cardiomyocytes, whereas the dominant-negative mutant Rab1N124I had the opposite effect. Brefeldin A treatment blocked the Rab1WT-induced increase in AT1R cell surface expression. Fluorescence analysis of the subcellular localization of AT1R revealed that Rab1 regulated AT1R transport specifically from the ER to the Golgi in HL-1 cardiomyocytes. Consistent with their effects on AT1R export, Rab1WT and Rab1N124I differentially modified the AT1R-mediated activation of ERK1/2 and its upstream kinase MEK1. More importantly, adenovirus-mediated expression of Rab1N124I markedly attenuated the Ang II-stimulated hypertrophic growth as measured by protein synthesis, cell size, and sarcomeric organization in neonatal cardiomyocytes. In contrast, Rab1WT expression augmented the Ang II-mediated hypertrophic response. These data strongly indicate that AT1R function in cardiomyocytes can be modulated through manipulating AT1R traffic from the ER to the Golgi and provide the first evidence implicating the ER-to-Golgi transport as a regulatory site for control of cardiomyocyte growth.


Assuntos
Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Miócitos Cardíacos/metabolismo , Receptor Tipo 1 de Angiotensina/química , Proteínas rab1 de Ligação ao GTP/metabolismo , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Transporte Biológico , Western Blotting , Brefeldina A/farmacologia , Divisão Celular , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Citometria de Fluxo , Genes Dominantes , Proteínas de Fluorescência Verde , Humanos , Immunoblotting , Leucina/metabolismo , Proteínas Luminescentes/metabolismo , MAP Quinase Quinase 1 , Microscopia de Fluorescência , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Plasmídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA