Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
1.
Cancer Lett ; 523: 135-147, 2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34634383

RESUMO

Epstein-Barr virus (EBV) is closely related to the development of several malignancies, such as B-cell lymphoma (B-CL), by the mechanism through which these malignancies develop remains largely unknown. We previously observed downregulation of the long noncoding RNA (lncRNA) IGFBP7-AS1 in response to EBV infection. However, the role of IGFBP7-AS1 in EBV-associated cancers has not been clarified. Here, we found that expression of IGFBP7-AS1, as well as its sense gene IGFBP7, is decreased in EBV-positive B-CL cells and clinical tissues. IGFBP7-AS1 stabilizes IGFBP7 mRNA by forming a duplex based on their overlapping regions. The tumour suppressor p53 transcriptionally activates IGFBP7-AS1 expression by binding to the promoter region of the lncRNA gene. The IGFBP7-AS1 expression is able to be rescued in EBV-positive cells in wild-type (wt) p53-dependent manner. IGFBP7-AS1 inhibits the proliferation and promotes the apoptosis of B-CL cells. Moreover, tumorigenic properties due to the depletion of IGFBP7-AS1 were restored by exogenous expression of IGFBP7 or wt-p53. Furthermore, the functional p53/IGFBP7-AS1/IGFBP7 axis facilitates apoptosis by suppressing the production and secretion of the NPPB signal peptide and further regulating the cGMP-PKG signalling pathway. This study demonstrates that EBV promotes tumorigenesis, particularly in B-CL progression, by downregulating the novel p53-responsive lncRNA IGFBP7-AS1.


Assuntos
Infecções por Vírus Epstein-Barr/complicações , Herpesvirus Humano 4/patogenicidade , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Linfoma de Células B/etiologia , RNA Longo não Codificante/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose , Carcinogênese , Linhagem Celular Tumoral , GMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Regulação para Baixo , Feminino , Humanos , Linfoma de Células B/patologia , Linfoma de Células B/virologia , Camundongos Endogâmicos BALB C
2.
JCI Insight ; 5(9)2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32315291

RESUMO

Bone fractures are a major cause of morbidity and mortality, particularly in patients with diabetes, who have a high incidence of fractures and exhibit poor fracture healing. Coordinated expression of osteoblast-derived vascular endothelial growth factor (VEGF) and bone morphogenic proteins (BMPs) is essential for fracture repair. The NO/cGMP/protein kinase G (PKG) signaling pathway mediates osteoblast responses to estrogens and mechanical stimulation, but the pathway's role in bone regeneration is unknown. Here, we used a mouse cortical-defect model to simulate bone fractures and studied osteoblast-specific PKG1-knockout and diabetic mice. The knockout mice had normal bone microarchitecture but after injury exhibited poor bone regeneration, with decreased osteoblasts, collagen deposition, and microvessels in the bone defect area. Primary osteoblasts and tibiae from the knockout mice expressed low amounts of Vegfa and Bmp2/4 mRNAs, and PKG1 was required for cGMP-stimulated expression of these genes. Diabetic mice also demonstrated low Vegfa and Bmp2/4 expression in bone and impaired bone regeneration after injury; notably, the cGMP-elevating agent cinaciguat restored Vegfa and BMP2/4 expression and full bone healing. We conclude that PKG1 is a key orchestrator of VEGF and BMP signaling during bone regeneration and propose pharmacological PKG activation as a novel therapeutic approach to enhance fracture healing.


Assuntos
Regeneração Óssea , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Diabetes Mellitus Experimental , Consolidação da Fratura , Osteoblastos , Animais , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Fraturas Ósseas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/metabolismo , Osteoblastos/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Biol Pharm Bull ; 43(4): 707-715, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32238713

RESUMO

Chaihu-Shugan-San (CSS) has been widely used as an alternative treatment for gastrointestinal (GI) diseases in East Asia. Interstitial cells of Cajal (ICCs) are pacemakers in the GI tract. In the present study, we examined the action of CSS on pacemaker potentials in cultured ICCs from the mouse small intestine in vitro and on GI motility in vivo. We used the electrophysiological methods to measure the pacemaker potentials in ICCs. GI motility was investigated by measuring intestinal transit rates (ITR). CSS inhibited the pacemaker potentials in a dose-dependent manner. The capsazepine did not block the effect of CSS. However, the effects of CSS were blocked by glibenclamide. In addition, NG-nitro-L-arginine methyl ester (L-NAME) also blocked the CSS-induced effects. Pretreatment with SQ-22536 or with KT-5720 did not suppress the effects of CSS; however, pretreatment with ODQ or KT-5823 did. Furthermore, CSS significantly suppressed murine ITR enhancement by neostigmine in vivo. These results suggest that CSS exerts inhibitory effects on the pacemaker potentials of ICCs via nitric oxide (NO)/cGMP and ATP-sensitive K+ channel dependent and transient receptor potential vanilloid 1 (TRPV1) channel independent pathways. Accordingly, CSS could provide the basis for the development of new treatments for GI motility dysfunction.


Assuntos
Células Intersticiais de Cajal/efeitos dos fármacos , Intestino Delgado/citologia , Extratos Vegetais/farmacologia , Animais , Células Cultivadas , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Guanilato Ciclase/fisiologia , Células Intersticiais de Cajal/fisiologia , Intestino Delgado/fisiologia , Canais KATP/fisiologia , Masculino , Camundongos Endogâmicos ICR , Óxido Nítrico/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Canais de Cátion TRPV/fisiologia
4.
J Chin Med Assoc ; 83(4): 357-366, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32101891

RESUMO

BACKGROUND: Nitric oxide (NO), which possesses both protective and toxic properties, has been observed to have a complicated biphasic character within various types of tissues, including neuronal cells. NO was also found to cause the increase of another important signaling molecular Zn (termed as NZR). The molecular mechanism of NZR has been extensively investigated, but the source of Zn is present of a major candidate that is yet to be answered. The NO-protein kinase G (PKG) pathway, mitochondria, and metallothioneins (MTs), are all proposed to be the individual source of NZR. However, this hypothesis remains inconclusive. In this study, we examined the function of PKG signaling cascades, the mitochondria storage, and MT-1 during NZR of living PC12 cells. METHODS: We applied live-cell imaging in combination with pharmacological inhibitors and activators as well as in vitro Zn assay to dissect the functions of the above candidates in NZR. RESULTS: Two mechanisms, namely, mitochondria as the only Zn source and the opening of NO-PKG-dependent mitochondrial ATP-sensitive potassium channels (mKATP) as the key to releasing NO-induced increase in mitochondrial Zn, were proven to be the two critical paths of NZR in neuronal-related cells. CONCLUSION: This new finding provides a reasonable explanation to previously existing and contradictory conclusions regarding the function of mitochondria/mKATP and PKG signaling on the molecular mechanism of NZR.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , GMP Cíclico/fisiologia , Mitocôndrias/fisiologia , Neurônios/metabolismo , Óxido Nítrico/fisiologia , Zinco/metabolismo , Animais , Canais KATP/fisiologia , Células PC12 , Ratos
5.
Inflammopharmacology ; 27(6): 1285-1296, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30945072

RESUMO

Rutin is a glycone form of the flavonol quercetin and it reduces inflammatory pain in animal models. Therapy with granulocyte colony-stimulating factor (G-CSF) is known by the pain caused as its main side effect. The effect of rutin and its mechanisms of action were evaluated in a model of hyperalgesia induced by G-CSF in mice. The mechanical hyperalgesia induced by G-CSF was reduced by treatment with rutin in a dose-dependent manner. Treatment with both rutin + morphine or rutin + indomethacin, at doses that are ineffectual per se, significantly reduced the pain caused by G-CSF. The nitric oxide (NO)-cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG)-ATP-sensitive potassium channel (KATP) signaling pathway activation is one of the analgesic mechanisms of rutin. Rutin also reduced the pro-hyperalgesic and increased anti-hyperalgesic cytokine production induced by G-CSF. Furthermore, rutin inhibited the activation of the nuclear factor kappa-light-chain enhancer of activated B cells (NFκB), which might explain the inhibition of the cytokine production. Treatment with rutin upregulated the decreased mRNA expression of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) combined with enhancement of the mRNA expression of the Nrf2 downstream target heme oxygenase (HO-1). Intraperitoneal (i.p.) treatment with rutin did not alter the mobilization of neutrophils induced by G-CSF. The analgesia by rutin can be explained by: NO-cGMP-PKG-KATP channel signaling activation, inhibition of NFκB and triggering the Nrf2/HO-1 pathway. The present study demonstrates rutin as a promising pharmacological approach to treat the pain induced by G-CSF without impairing its primary therapeutic benefit of mobilizing hematopoietic progenitor cells into the blood.


Assuntos
Analgésicos/farmacologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Dor/tratamento farmacológico , Rutina/farmacologia , Animais , GMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Citocinas/biossíntese , Heme Oxigenase-1/fisiologia , Hiperalgesia/tratamento farmacológico , Canais KATP/fisiologia , Masculino , Camundongos , Fator 2 Relacionado a NF-E2/fisiologia , NF-kappa B/antagonistas & inibidores , Neutrófilos/efeitos dos fármacos , Óxido Nítrico/fisiologia , Dor/induzido quimicamente , Transdução de Sinais/efeitos dos fármacos
6.
Adv Exp Med Biol ; 1074: 511-517, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721983

RESUMO

In humans cone photoreceptors are responsible for high-resolution colour vision. A variety of retinal diseases can compromise cone viability, and, at present, no satisfactory treatment options are available. Here, we present data towards establishing a reliable, high-throughput assay system that will facilitate the search for cone neuroprotective compounds using the murine-photoreceptor cell line 661 W. To further characterize 661 W cells, a retinal marker study was performed, followed by the induction of cell death using paradigms over-activating cGMP-dependent protein kinase G (PKG). We found that 661 W cells may be used to mimic specific aspects of cone degeneration and may thus be valuable for future compound screening studies.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Avaliação Pré-Clínica de Medicamentos/métodos , Proteínas do Olho/fisiologia , Ensaios de Triagem em Larga Escala , Fármacos Neuroprotetores/isolamento & purificação , Células Fotorreceptoras Retinianas Cones/enzimologia , Animais , Biomarcadores , Linhagem Celular Tumoral , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/efeitos dos fármacos , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/deficiência , Ativação Enzimática/efeitos dos fármacos , Proteínas do Olho/análise , Camundongos , Camundongos Knockout , Fármacos Neuroprotetores/farmacologia , Especificidade de Órgãos , Inibidores de Fosfodiesterase/farmacologia , Purinonas/farmacologia , Células Fotorreceptoras Retinianas Cones/citologia
7.
Cancer Lett ; 370(2): 279-85, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26551887

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is an aggressive disease with high mortality. Treatments, which can result in significant morbidity, have not substantially changed in three decades. The second messenger cyclic GMP (cGMP), which targets protein kinase G (PKG), is generated by guanylate cyclases (GCs), and is rapidly hydrolyzed by phosphodiesterases (PDEs). Activation of the cGMP/PKG pathway is antineoplastic in several cancer types, but its impact on HNSCC has not been fully exploited. We found differential expression of critical components of this pathway in four HNSCC cell lines. Several activators of soluble GC (sGC), as well as inhibitors of PDE5, increased intracellular cGMP, reduced cell viability, and induced apoptosis in HNSCC cells. The apoptotic effects of the sGC activator BAY 41-2272 and the PDE5 inhibitor Tadalafil (Cialis) were mediated by PKG. Furthermore, Tadalafil substantially reduced the growth of CAL27-derived tumors in athymic mice. Several drugs which either activate sGC or inhibit PDE5 are approved for treatment of nonmalignant conditions. These drugs could be repurposed as novel and effective therapeutics in patients with head and neck cancer.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , GMP Cíclico/fisiologia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Feminino , Guanilato Ciclase/fisiologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Inibidores da Fosfodiesterase 5/uso terapêutico , Pirazóis/uso terapêutico , Piridinas/uso terapêutico , Carcinoma de Células Escamosas de Cabeça e Pescoço , Tadalafila/uso terapêutico
8.
Basic Res Cardiol ; 110(4): 42, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26036467

RESUMO

Cyclic GMP-dependent protein kinase (PKG) is a serine-threonine kinase that mediates the cardioprotective effect of ischemic and pharmacologic preconditioning. Since hydrogen sulfide (H2S) has been implicated in mediating the cardioprotective effects of the cGMP modulators tadalafil and cinaciguat, we tested the hypothesis that myocardial gene therapy with PKG exerts cardioprotection against ischemia/reperfusion (I/R) injury through a mechanism involving H2S. Adult rat cardiomyocytes were infected with adenoviral vector encoding PKGIα or inactive mutant PKGIαK390A (K390A) for 24 h. Necrosis and apoptosis (n = 6/group) were determined after 90 min of simulated ischemia and 1 or 18 h of reoxygenation, respectively. To study the effect of PKGIα in vivo, mice received intramyocardial injections of adenoviral PKGIα or K390A. Four days later, the hearts were subjected to 30 min of ischemia followed by reperfusion for 24 h. The inhibitor of H2S-producing enzyme, cystathionine-γ-lyase (CSE), dl-propargylglycine (PAG, 50 mg/kg, ip) was given 30 min before ischemia. PKGIα overexpression induced CSE expression, whereas cystathionine-ß-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase expression was not changed. PKGIα overexpression increased H2S in the heart and cardiomyocytes in relation to control and PKGIαK390A. Moreover, PAG abolished protection with PKGIα in vitro by increasing necrosis (35.2 ± 1.7%, P < 0.05) and apoptosis (23.5 ± 1.8 %, P < 0.05) as compared to PKGIα-overexpressing cells (necrosis: 17.2 ± 0.9% and apoptosis: 13.2 ± 0.8%). In vivo, PKGIα overexpression reduced infarct size and preserved left ventricular fractional shortening as compared with K390A (P < 0.05) and PAG abolished the cardioprotective effect of PKGIα. The protective effect of myocardial gene therapy with PKGIα against I/R injury is mediated through a mechanism involving H2S signaling.


Assuntos
Proteína Quinase Dependente de GMP Cíclico Tipo I/genética , Terapia Genética , Sulfeto de Hidrogênio/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Células Cultivadas , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Masculino , Camundongos , Ratos , Ratos Wistar
9.
Hypertension ; 66(1): 190-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25916722

RESUMO

Elevated B-type natriuretic peptide (BNP) regulates cGMP-phosphodiesterase activity. Its elevation is regarded as an early compensatory response to cardiac failure where it can facilitate sympathovagal balance and cardiorenal homeostasis. However, recent reports suggest a paradoxical proadrenergic action of BNP. Because phosphodiesterase activity is altered in cardiovascular disease, we tested the hypothesis that BNP might lose its efficacy by minimizing the action of cGMP on downstream pathways coupled to neurotransmission. BNP decreased norepinephrine release from atrial preparations in response to field stimulation and also significantly reduced the heart rate responses to sympathetic nerve stimulation in vitro. Using electrophysiological recording and fluorescence imaging, BNP also reduced the depolarization evoked calcium current and intracellular calcium transient in isolated cardiac sympathetic neurons. Pharmacological manipulations suggested that the reduction in the calcium transient was regulated by a cGMP/protein kinase G pathway. Fluorescence resonance energy transfer measurements for cAMP, and an immunoassay for cGMP, showed that BNP increased cGMP, but not cAMP. In addition, overexpression of phosphodiesterase 2A after adenoviral gene transfer markedly decreased BNP stimulation of cGMP and abrogated the BNP responses to the calcium current, intracellular calcium transient, and neurotransmitter release. These effects were reversed on inhibition of phosphodiesterase 2A. Moreover, phosphodiesterase 2A activity was significantly elevated in stellate neurons from the prohypertensive rat compared with the normotensive control. Our data suggest that abnormally high levels of phosphodiesterase 2A may provide a brake against the inhibitory action of BNP on sympathetic transmission.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/fisiologia , Sistema de Condução Cardíaco/enzimologia , Hipertensão/enzimologia , Peptídeo Natriurético Encefálico/farmacologia , Sistema Nervoso Simpático/efeitos dos fármacos , Animais , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , GMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiologia , Frequência Cardíaca , Hipertensão/genética , Hipertensão/fisiopatologia , Isatina/farmacologia , Masculino , Peptídeo Natriurético Encefálico/fisiologia , Neurônios/enzimologia , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores do Fator Natriurético Atrial/efeitos dos fármacos , Receptores do Fator Natriurético Atrial/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Gânglio Estrelado/citologia , Gânglio Estrelado/efeitos dos fármacos , Gânglio Estrelado/fisiologia , Sistema Nervoso Simpático/fisiologia , Transmissão Sináptica/fisiologia
10.
Int J Urol ; 21(1): 87-92, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23692571

RESUMO

OBJECTIVES: To investigate whether 7-[2-[4-(2-chlorophenyl) piperazinyl] ethyl]-1,3-di-methylxanthine (KMUP-1) inhibits the effects of testosterone on the development of benign prostatic hyperplasia and sensitizes prostate contraction. METHODS: A benign prostatic hyperplasia animal model was established by subcutaneous injections of testosterone (3 mg/kg/day, s.c.) for 4 weeks in adult male Sprague-Dawley rats. Animals were divided into six groups: control, testosterone, testosterone with KMUP-1 (2.5, 5 mg/kg/day), sildenafil (5 mg/kg/day) or doxazosin (5 mg/kg/day). After 4 weeks, the animals were killed, and prostate tissues were prepared for isometric tension measurement and western blotting analysis. KMUP-1, Y27632, zaprinast, doxazosin or tamsulosin were used at various concentrations to determine the contractility sensitized by phenylephrine (10 µmol/L). RESULTS: KMUP-1 inhibited testosterone-induced phosphorylation of extracellular signal-regulated phosphorylated protein kinase and mitogen-activated protein kinase kinase and Rho kinase-II activation. Sildenafil and doxazosin significantly decreased benign prostatic hyperplasia-induced mitogen-activated protein kinase kinase and Rho kinase-II activation. The decreased expressions of soluble guanylate cyclase α1 was reversed by KMUP-1, doxazosin and sildenafil. Soluble guanylate cyclase ß1 and protein kinase G were increased by KMUP-1, doxazosin, and sildenafil in the testosterone-treated benign prostatic hyperplasia group. Phosphodiesterase-5A was increased by testosterone and inhibited by KMUP-1 (5 mg/kg/day) or sildenafil (5 mg/kg/day). KMUP-1 inhibited phenylephrine-sensitized prostate contraction of rats treated with testosterone. CONCLUSIONS: Mitogen-activated protein kinase 1/extracellular regulated protein kinases kinase, soluble guanylate cyclase/cyclic guanosine monophosphate, protein kinase/protein kinase G and Rho kinase-II are related to prostate smooth muscle tone and proliferation induced by testosterone. KMUP-1 inhibits testosterone-induced prostate hyper-contractility and mitogen-activated protein kinase 1/extracellular regulated protein kinases kinase-phosphorylation, and it inactivates Rho kinase-II by cyclic guanosine monophosphate, protein kinase and α1A-adenergic blockade. Thus, KMUP-1 might be a beneficial pharmacotherapy for benign prostatic hyperplasia.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , GMP Cíclico/fisiologia , Guanilato Ciclase/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Piperidinas/farmacologia , Hiperplasia Prostática/prevenção & controle , Receptores Citoplasmáticos e Nucleares/fisiologia , Xantinas/farmacologia , Quinases Associadas a rho/fisiologia , Animais , Proteínas Quinases Dependentes de GMP Cíclico/efeitos dos fármacos , Modelos Animais de Doenças , Guanilato Ciclase/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Guanilil Ciclase Solúvel , Quinases Associadas a rho/efeitos dos fármacos
11.
Circulation ; 129(4): 451-62, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24190960

RESUMO

BACKGROUND: ß1-2-adrenergic receptors (AR) are key regulators of cardiac contractility and remodeling in response to catecholamines. ß3-AR expression is enhanced in diseased human myocardium, but its impact on remodeling is unknown. METHODS AND RESULTS: Mice with cardiac myocyte-specific expression of human ß3-AR (ß3-TG) and wild-type (WT) littermates were used to compare myocardial remodeling in response to isoproterenol (Iso) or Angiotensin II (Ang II). ß3-TG and WT had similar morphometric and hemodynamic parameters at baseline. ß3-AR colocalized with caveolin-3, endothelial nitric oxide synthase (NOS) and neuronal NOS in adult transgenic myocytes, which constitutively produced more cyclic GMP, detected with a new transgenic FRET sensor. Iso and Ang II produced hypertrophy and fibrosis in WT mice, but not in ß3-TG mice, which also had less re-expression of fetal genes and transforming growth factor ß1. Protection from Iso-induced hypertrophy was reversed by nonspecific NOS inhibition at low dose Iso, and by preferential neuronal NOS inhibition at high-dose Iso. Adenoviral overexpression of ß3-AR in isolated cardiac myocytes also increased NO production and attenuated hypertrophy to Iso and phenylephrine. Hypertrophy was restored on NOS or protein kinase G inhibition. Mechanistically, ß3-AR overexpression inhibited phenylephrine-induced nuclear factor of activated T-cell activation. CONCLUSIONS: Cardiac-specific overexpression of ß3-AR does not affect cardiac morphology at baseline but inhibits the hypertrophic response to neurohormonal stimulation in vivo and in vitro, through a NOS-mediated mechanism. Activation of the cardiac ß3-AR pathway may provide future therapeutic avenues for the modulation of hypertrophic remodeling.


Assuntos
Ventrículos do Coração/patologia , Miócitos Cardíacos/metabolismo , Neurotransmissores/farmacologia , Óxido Nítrico Sintase/fisiologia , Receptores Adrenérgicos beta 3/metabolismo , Remodelação Ventricular/efeitos dos fármacos , Angiotensina II/efeitos adversos , Angiotensina II/farmacologia , Animais , Células Cultivadas , GMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Modelos Animais de Doenças , Ventrículos do Coração/fisiopatologia , Humanos , Hipertrofia/induzido quimicamente , Hipertrofia/patologia , Hipertrofia/fisiopatologia , Técnicas In Vitro , Isoproterenol/efeitos adversos , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Neurotransmissores/efeitos adversos , Receptores Adrenérgicos beta 3/genética , Transdução de Sinais/fisiologia , Remodelação Ventricular/fisiologia
13.
Acta cir. bras ; 28(7): 537-542, July 2013. ilus, graf
Artigo em Inglês | LILACS | ID: lil-679087

RESUMO

PURPOSE: To investigate the role of protein kinase G (PKG) in blocking post-shock mesenteric lymph (PSML) return ameliorating the calcium sensitivity in hemorrhagic shock rats. METHODS: Male Wistar rats were randomly divided into sham, shock, shock+ligation (shock plus mesenteric lymph duct ligation (MLDL)), shock+drainage (shock plus PSML drainage) groups. After shock (hypotension 40mmHg) for three hours or corresponding times, the superior mesenteric artery (SMA) was taken out for detecting the PKG and phospho PKG (p-PKG) contents, and the vascular rings of SMA were prepared for assaying the calcium sensitivity using an isolated organ perfusion system. RESULTS: The PKG and p-PKG contents of SMA in shock group were significantly increased than that of sham group, and MLDL or PSML drainage reducing the levels of PKG and p-PKG. Meanwhile, the vascular calcium sensitivity in shock group was significantly lower than that of sham group, MLDL or PSML drainage enhanced the calcium sensitivity. After incubating with PKG regulators in shock+ligation and shock+drainage groups, the PKG agonist 8Br-cGMP reduced the contractility of vascular rings to gradient calcium ions and Emax and the PKG inhibitor agonist KT5823 elevated the calcium sensitivity significantly. CONCLUSION: Protein kinase G plays an important role in post-shock mesenteric lymph blockage improving vascular calcium sensitivity.


Assuntos
Animais , Masculino , Ratos , Cálcio/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Artéria Mesentérica Superior/metabolismo , Choque Hemorrágico/metabolismo , Western Blotting , Cálcio/análise , Proteínas Quinases Dependentes de GMP Cíclico/análise , Ensaio de Imunoadsorção Enzimática , Contração Muscular , Artéria Mesentérica Superior/fisiopatologia , Distribuição Aleatória , Ratos Wistar , Choque Hemorrágico/fisiopatologia
14.
Cardiovasc Res ; 99(4): 648-56, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23764881

RESUMO

AIMS: Titin-based myofilament stiffness is defined by the expression levels of the cardiac titin-isoforms, N2B and N2BA, and by phosphorylation of the elastic titin domains N2-B unique sequence (N2-Bus) and PEVK. Phosphorylation of the N2-Bus by cGMP-dependent protein kinase (PKG) or cAMP-dependent protein kinase (PKA) decreases titin stiffness, whereas phosphorylation of the PEVK-domain by PKC increases it. We aimed to identify specific sites within the N2-Bus phosphorylated by PKA and PKG and to determine whether differential changes in titin domain phosphorylation could affect passive stiffness in human failing hearts. METHODS AND RESULTS: Using mass spectrometry, we identified seven partly conserved PKA/PKG-targeted phosphorylation motifs in human and rat N2-Bus. Polyclonal antibodies to pSer4185, pSer4010, and pSer4099 in the N2-Bus, and to pSer11878 in the PEVK-region were used to quantify titin-domain phosphorylation by western blot analyses of a set of human donor and failing hearts with similar titin-isoform composition. Passive tension determined in skinned human myocardial fibre preparations was significantly increased in failing compared with donor hearts, notably at shorter sarcomere lengths where titin contributes most to total passive tension. Phosphorylation of Ser4185, Ser4010, and Ser4099 in the N2-Bus was significantly reduced in failing hearts, whereas phosphorylation of Ser11878 in the PEVK-region was increased compared with donor hearts. CONCLUSION: We conclude that hypo-phosphorylation of the N2-Bus and hyper-phosphorylation of the PEVK domain can act complementary to elevate passive tension in failing human hearts. Differential changes in titin-domain phosphorylation may be important to fine-tune passive myocardial stiffness and diastolic function of the heart.


Assuntos
Conectina/metabolismo , Insuficiência Cardíaca/metabolismo , Miócitos Cardíacos/metabolismo , Miofibrilas/fisiologia , Sequência de Aminoácidos , Animais , Conectina/química , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Humanos , Dados de Sequência Molecular , Fosforilação , Estrutura Terciária de Proteína , Ratos
15.
Am J Physiol Renal Physiol ; 304(9): F1210-6, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23445619

RESUMO

The rate of urine secretion by insect Malpighian tubules (MTs) is regulated by multiple diuretic and antidiuretic hormones, often working either synergistically or antagonistically. In the Drosophila melanogaster MT, only diuretic factors have been reported. Two such agents are the biogenic amine tyramine (TA) and the peptide drosokinin (DK), both of which act on the stellate cells of the tubule to increase transepithelial chloride conductance. In the current study, TA and DK signaling was quantified by microelectrode recording of the transepithelial potential in isolated Drosophila MTs. Treatment of tubules with cGMP caused a significant reduction in the depolarizing responses to both TA and DK, while cAMP had no effect on these responses. To determine whether a specific cGMP-dependent protein kinase (PKG) was mediating this inhibition, PKG expression was knocked down by RNAi in either the principal cells or the stellate cells. Knockdown of Pkg21D in the stellate cells eliminated the modulation of TA and DK signaling. Knockdown of Pkg21D with a second RNAi construct also reduced the modulation of TA signaling. In contrast, knockdown of the expression of foraging or CG4839, which encodes a known and a putative PKG, respectively, had no effect. These data indicate that cGMP, acting through the Pkg21D gene product in the stellate cells, can inhibit signaling by the diuretic agents TA and DK. This represents a novel function for cGMP and PKG in the Drosophila MT and suggests the existence of an antidiuretic hormone in Drosophila.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Diuréticos/farmacologia , Drosophila melanogaster/fisiologia , Animais , AMP Cíclico/farmacologia , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/genética , Proteínas de Drosophila/farmacologia , Epitélio/efeitos dos fármacos , Epitélio/fisiologia , Feminino , Túbulos de Malpighi/efeitos dos fármacos , Túbulos de Malpighi/fisiologia , Modelos Animais , Neuropeptídeos/farmacologia , Interferência de RNA , Tiramina/farmacologia
16.
Invest Ophthalmol Vis Sci ; 54(3): 1636-49, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23385799

RESUMO

PURPOSE: Multidrug, resistance-associated protein-4 (MRP4) is a membrane transporter that regulates the cellular efflux of cyclic nucleotides (cAMP and cGMP) involved in various physiologic responses. This study examined the expression and distribution of MRP4 in the trabecular meshwork (TM) cells and its role in homeostasis of IOP. METHODS: Expression and distribution of MRP4 in human TM (HTM) cells and aqueous humor (AH) outflow pathway was determined by RT-PCR, immunoblotting, and immunofluorescence. Effects of inhibiting MRP4 activity and suppression of MRP4 expression on cAMP and cGMP levels, myosin light chain (MLC) phosphorylation, actin filament organization and activity of protein kinase G (PKG), protein kinase A (PKA), Rho guanosine triphosphatase (GTPase), and MLC phosphatase was monitored in HTM cells using ELISA, siRNA, biochemical, and immunofluorescence analyses. Topical application of the MRP4 inhibitor MK571 was tested to assess changes in IOP in rabbits. RESULTS: RT-PCR, immunoblot, and immunofluorescence analyses confirmed the expression of MRP4 in HTM cells and distribution in human AH outflow pathway. Inhibition of MRP4 in HTM cells by MK571 or probenecid resulted in cell shape changes and decreases in actin stress fibers and MLC phosphorylation. Levels of intracellular cAMP and cGMP in HTM cells were increased significantly under these conditions. MK571-induced HTM cell relaxation appeared to be mediated predominantly via activation of the cGMP-dependent PKG signaling pathway. Topical application of MK571 significantly decreased IOP in Dutch-Belted rabbits. CONCLUSIONS: These observations reveal that cyclic nucleotide efflux controlling transporter-MRP4 plays a significant role in IOP homeostasis potentially by regulating the relaxation characteristics of AH outflow pathway cells.


Assuntos
AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Homeostase/fisiologia , Pressão Intraocular/fisiologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Malha Trabecular/citologia , Actinas/fisiologia , Actinas/ultraestrutura , Adulto , Idoso , Animais , Humor Aquoso/citologia , Humor Aquoso/fisiologia , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Humanos , Antagonistas de Leucotrienos/farmacologia , Pessoa de Meia-Idade , Proteínas Associadas à Resistência a Múltiplos Medicamentos/antagonistas & inibidores , Cadeias Leves de Miosina/fisiologia , Fosforilação/fisiologia , Probenecid/farmacologia , Propionatos/farmacologia , Quinolinas/farmacologia , RNA Interferente Pequeno/fisiologia , Coelhos , Transdução de Sinais/fisiologia , Malha Trabecular/metabolismo
17.
Antioxid Redox Signal ; 18(9): 1042-52, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22867279

RESUMO

SIGNIFICANCE: Oxidants were once principally considered perpetrators of injury and disease. However, this has become an antiquated view, with cumulative evidence showing that the oxidant hydrogen peroxide serves as a signaling molecule. Hydrogen peroxide carries vital information about the redox state of the cell and is crucial for homeostatic regulation during health and adaptation to stress. RECENT ADVANCES: In this review, we examine the contemporary concepts for how hydrogen peroxide is sensed and transduced into a biological response by introducing post-translational oxidative modifications on select proteins. Oxidant sensing and signaling by kinases are of particular importance as they integrate oxidant signals into phospho-regulated pathways. We focus on CAMKII, PKA, and PKG, kinases whose redox regulation has notable impact on cardiovascular function. CRITICAL ISSUES: In addition, we examine the mechanism for regulating intracellular hydrogen peroxide, considering the net concentrations that may accumulate. The effects of endogenously generated oxidants are often modeled by applying exogenous hydrogen peroxide to cells or tissues. Here we consider whether model systems exposed to exogenous hydrogen peroxide have relevance to systems where the oxidant is generated endogenously, and if so, what concentration can be justified in terms of relevance to health and disease. FUTURE DIRECTIONS: Improving our understanding of hydrogen peroxide signaling and the sensor proteins that it can modify will help us develop new strategies to regulate intracellular signaling to prevent disease.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Sistema Cardiovascular/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Peróxido de Hidrogênio/metabolismo , Transdução de Sinais/fisiologia , Animais , Cisteína/metabolismo , Homeostase , Humanos , Peróxido de Hidrogênio/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Metionina/metabolismo , Modelos Cardiovasculares , Oxirredução , Oxirredutases/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Sistemas do Segundo Mensageiro/fisiologia , Compostos de Sulfidrila/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Superóxidos/metabolismo
18.
Hepatology ; 57(4): 1384-93, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23174781

RESUMO

UNLABELLED: Obesity is associated with many severe chronic diseases and deciphering its development and molecular mechanisms is necessary for promoting treatment. Previous studies have revealed that mitochondrial content is down-regulated in obesity, diabetes, and nonalcoholic fatty liver disease (NAFLD) and proposed that NAFLD and diabetes are mitochondrial diseases. However, the exact mechanisms underlying these processes remain unclear. In this study, we discovered that resistin down-regulated the content and activities of mitochondria, enhanced hepatic steatosis, and induced insulin resistance (IR) in mice. The time course indicated that the change in mitochondrial content was before the change in fat accumulation and development of insulin resistance. When the mitochondrial content was maintained, resistin did not stimulate hepatic fat accumulation. The present mutation study found that the residue Thr464 of the p65 subunit of nuclear factor kappa B was essential for regulating mitochondria. A proximity ligation assay revealed that resistin inactivated peroxisome proliferator activated receptor gamma coactivator 1 alpha (PGC-1α) and diminished the mitochondrial content by promoting the interaction of p65 and PGC-1α. Signaling-transduction analysis demonstrated that resistin down-regulated mitochondria by a novel protein kinase C/protein kinase G/p65/PGC-1α-signaling pathway. CONCLUSION: Resistin induces hepatic steatosis through diminishing mitochondrial content. This reveals a novel pathway for mitochondrial regulation, and suggests that the maintenance of normal mitochondrial content could be a new strategy for treatment of obesity-associated diseases.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Fígado Gorduroso/induzido quimicamente , Mitocôndrias Hepáticas/efeitos dos fármacos , Proteína Quinase C/fisiologia , Resistina/efeitos adversos , Resistina/farmacologia , Transativadores/fisiologia , eIF-2 Quinase/fisiologia , Animais , Modelos Animais de Doenças , Regulação para Baixo , Fígado Gorduroso/fisiopatologia , Resistência à Insulina/fisiologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Hepáticas/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Transdução de Sinais/fisiologia , Fatores de Transcrição
19.
J Physiol ; 590(20): 5053-73, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22826131

RESUMO

Mouse chromaffin cells (MCCs) express high densities of L-type Ca2+ channels (LTCCs), which control pacemaking activity and catecholamine secretion proportionally to their density of expression. In vivo phosphorylation of LTCCs by cAMP-PKA and cGMP­PKG, regulate LTCC gating in two opposing ways: the cAMP-PKA pathway potentiates while the cGMP­PKG cascade inhibits LTCCs. Despite this, no attempts have been made to answer three key questions related to the two Cav1 isoforms expressed in MCCs (Cav1.2 and Cav1.3): (i) how much are the two Cav1 channels basally modulated by PKA and PKG?, (ii) to what extent can Cav1.2 and Cav1.3 be further regulated by PKA or PKG activation?, and (iii) are the effects of both kinases cumulative when simultaneously active? Here, by comparing the size of L-type currents of wild-type (WT; Cav1.2+Cav1.3) and Cav1.3−/− KO (Cav1.2) MCCs, we provide new evidence that both PKA and PKG pathways affect Cav1.2 and Cav1.3 to the same extent either under basal conditions or induced stimulation. Inhibition of PKA by H89 (5 µM) reduced the L-type current in WT and KO MCCs by∼60%,while inhibition of PKG by KT 5823 (1 µM) increased by∼40% the same current in both cell types. Given that Cav1.2 and Cav1.3 carry the same quantity of Ca2+ currents, this suggests equal sensitivity of Cav1.2 and Cav1.3 to the two basal modulatory pathways. Maximal stimulation of cAMP­PKA by forskolin (100 µM) and activation of cGMP­PKG by pCPT-cGMP (1mM) uncovered a∼25% increase of L-type currents in the first case and∼65% inhibition in the second case in both WT and KO MCCs, suggesting equal sensitivity of Cav1.2 and Cav1.3 during maximal PKA or PKG stimulation. The effects of PKA and PKG were cumulative and most evident when one pathway was activated and the other was inhibited. The two extreme combinations(PKA activation­PKG inhibition vs. PKG activation-PKA inhibition) varied the size of L-type currents by one order of magnitude (from 180% to 18% of control size). Taken together our data suggest that: (i) Cav1.2 and Cav1.3 are equally sensitive to PKA and PKG action under both basal conditions and maximal stimulation, and (ii) PKA and PKG act independently on both Cav1.2 and Cav1.3, producing cumulative effects when opposingly activated. These extreme Cav1 channel modulations may occur either during high-frequency sympathetic stimulation to sustain prolonged catecholamine release (maximal L-type current) or following activation of the NO­cGMP­PKG signalling pathway (minimal L-type current) to limit the steady release of catecholamines.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Animais , Células Cultivadas , Células Cromafins/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
20.
Biochim Biophys Acta ; 1820(11): 1724-33, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22766195

RESUMO

BACKGROUND: Phytoestrogens are plant-derived polyphenolic compounds that exert beneficial effects on human health, mostly related to their estrogen mimetic activity. In particular a strong correlation between phytoestrogens intake and a lower risk of cardiovascular diseases has been reported. The flavanone 8-prenylnaringenin, extracted from hop flowers, has been identified as a novel phytoestrogen, unique with respect to estrogen receptors specificity and potency. However, to date no investigations on the 8-prenylnaringenin role in modulating platelet function have been undertaken. METHODS: We evaluated the effect of 8-prenylnaringenin on platelet aggregation, intracellular calcium mobilization and protein phosphorylation triggered by thrombin and collagen, and platelet adhesion and dense granule secretion triggered by collagen. RESULTS: 8-Prenylnaringenin inhibited platelet aggregation induced by different agonists and platelet adhesion to collagen matrix. 8-Prenylnaringenin directly increased intracellular cAMP and cGMP levels and thus promoted VASP phosphorylation. However, these molecular events were not responsible for the inhibitory action of 8-prenylnaringenin on platelets. Moreover, 8-prenylnaringenin inhibited the phosphorylation of Pyk2, Akt, and ERK1/2. Finally, 8-prenylnaringenin suppressed the mobilization of calcium and the secretion of dense granules. All these effects were independent of estrogen receptors recruitment. CONCLUSIONS: 8-Prenylnaringenin exerted anti-aggregatory and anti-adhesive effects on human platelets, independently of estrogen receptors, acting as an inhibitor of multiple proteins essential for the morphological and biochemical transformations that occur during platelet activation and aggregation. GENERAL SIGNIFICANCE: 8-Prenylnaringenin may represent a useful tool in the therapy and prevention of vascular diseases associated with platelet aggregation, such as atherosclerosis, myocardial infarction, coronary artery disease, and thrombosis.


Assuntos
Flavanonas/farmacologia , Fitoestrógenos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Cálcio/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Grânulos Citoplasmáticos/efeitos dos fármacos , Grânulos Citoplasmáticos/metabolismo , Humanos , Fosforilação , Agregação Plaquetária/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA