Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
São Paulo; s.n; s.n; 2019. 79 p. tab, graf, ilus.
Thesis in Portuguese | LILACS | ID: biblio-1049916

ABSTRACT

A doença de Chagas é uma doença negligenciada causada pelo protozoário Trypanosoma cruzi constituindo-se em um problema de saúde pública em vários países da América Latina. No seu complexo ciclo de vida, o protozoário passa por quatro estágios diferentes: tripomastigota metacíclica, amastigota, tripomastigota sanguíneo e epimastigota, que permitem sua sobrevivência nos diferentes ambientes com os quais o parasita entra em contato. A diferenciação dos tripomastigotas de T. cruzi em amastigotas (amastigogênese) ocorre com grandes mudanças morfológicas, estruturais e metabólicas no parasita e pode ser reproduzido in vitro por exemplo, pela acidificação do meio extracelular. Apesar dos vários trabalhos descritos na literatura, o processo ainda não é totalmente compreendido. A participação de NO na transdução de sinal durante a amastigogênese, sugerida por dados não publicados de nosso grupo, assim como a via de sinalização dependente de AMPc, foram o foco do presente estudo. A indução da amastigogênese foi obtida por incubação de tripomastigotas em meio de cultura acidificado (pH 6,0) e os parâmetros estudados comparados com parasitas controle (meio de cultura, pH 7,4). Estudamos a variação no perfil de nucleotídios cíclicos (AMPc, GMPc), de quinases (PKA, MAPK- ERK1/2), de uma fosfatase (PP2A), assim como o perfil de proteínas fosforiladas, S-nitrosiladas e nitradas até 6 h do início da amastigogênese. O processo foi dividido nas etapas: inicial (até 60 minutos) e tardio (em torno de 3-4 h), caracterizados por um aumento de formas amastigotas na etapa tardia. Houve um aumento de aproximadamente 17 vezes no nível de AMPc nos primeiros 15 minutos da amastigogênese (meio pH 6,0), seguido por aumento discreto no nível de PKA fosforilada, utilizado como indicador de atividade enzimática, este mais evidente na etapa tardia (360 minutos). Quanto à subunidade catalítica fosforilada da MAPK (ativa), há uma aparente diminuição no nível de fosforilação na fase inicial (30 minutos) e aumento na etapa tardia (120 minutos) do processo de amastigogênese. Quanto ao perfil geral de fosforilação de proteínas, há uma diminuição de fosforilação em torno de 30 minutos, seguida de aumento de fosforilação em proteínas de aproximadamente 5 e 100 kDa, mas de maneira geral, não se observaram grandes mudanças nesse perfil com a metodologia utilizada. Quanto às modificações por NO e seus derivados, foram observadas modificações por S-nitrosilação e nitração das proteínas, além do aumento de GMPc em torno de 60 minutos. Embora essas modificações modulem a atividade biológica de uma grande diversidade de proteínas, seu papel biológico não foi explorado.8 Em resumo, nossos resultados apontam para uma variação no perfil de fosforilação, S-nitrosilação e nitração de proteínas, além do aumento de AMPc e GMPc ao longo do processo de amastigogênese in vitro, com a via de sinalização dependente de quinases/ fosfatases e de óxido nítrico ocorrendo ao longo do processo de amastigogênese


Chagas disease is a neglected disease caused by the parasite Trypanosoma cruzi and is a public health problem in several Latin American countries. In its complex life cycle, the protozoan goes through four different stages: metacyclic trypomastigote, amastigote, blood trypomastigote and epimastigote, which allow its survival in the different environments which the parasite comes into contact. The differentiation of T. cruzi trypomastigotes into amastigotes (amastigogenesis) occurs with large morphological, structural and metabolic changes in the parasite and can be reproduced in vitro by, for example, acidification of the extracellular medium. Despite the many data described in the literature, the process is not yet fully understood. The participation of NO in signal transduction during amastigogenesis, suggested by unpublished data from our group, as well as the cAMP-dependent signaling pathway, were the focus of the present study. The induction of amastigogenesis was obtained by incubating trypomastigotes in acidified culture medium (pH 6.0) and the studied parameters compared with control parasites (culture medium, pH 7.4). We studied the variation in the profile of cyclic nucleotides (cAMP, cGMP), kinases (PKA, MAPK-ERK1 / 2), phosphatase (PP2A), as well as the profile of phosphorylated, S-nitrosylated and nitrated proteins up to 6 h. onset of amastigogenesis. The process was divided into early (up to 60 minutes) and late (around 3-4 hours), characterized by an increase in amastigote forms in the late stage. There was an approximately 17-fold increase in cAMP level in the first 15 minutes of amastigogenesis (pH 6.0 medium), followed by a slight increase in phosphorylated PKA level, most evident in the late stage (360 minutes). As for the phosphorylated catalytic subunit of MAPK (active), there is an apparent decrease in the phosphorylation level in the early phase (30 minutes) and increase in the late stage (120 minutes) of the amastigogenesis process. As for the general protein phosphorylation profile, there is a decrease in phosphorylation around 30 minutes, followed by an increase in phosphorylation of proteins (approximately 5 and 100 kDa), but overall, no major changes were observed in this profile with the methodology used. As for modifications by NO and its derivatives, modifications were observed by S-nitrosylation and protein nitration, besides the increase of cGMP around 60 minutes. Although these modifications modulate the biological activity of a wide range of proteins, their biological role has not been explored. In summary, our results point to a variation in phosphorylation, S-nitrosylation and nitration profile of proteins, as well as an increase in cAMP and cGMP along the amastigogenesis process, implicating kinases / phosphatases and nitric oxide dependent signaling pathways in this differentiation


Subject(s)
Phosphorylation , Trypanosoma cruzi/metabolism , Nitric Oxide Synthase/chemistry , Receptors, Cyclic AMP/analysis , Cyclic GMP-Dependent Protein Kinases/analysis , MAP Kinase Kinase Kinases/analysis , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/analysis
2.
The Korean Journal of Physiology and Pharmacology ; : 315-324, 2016.
Article in English | WPRIM | ID: wpr-728441

ABSTRACT

Human cardiac fibroblasts (HCFs) have various voltage-dependent K+ channels (VDKCs) that can induce apoptosis. Hydrogen peroxide (H2O2) modulates VDKCs and induces oxidative stress, which is the main contributor to cardiac injury and cardiac remodeling. We investigated whether H2O2 could modulate VDKCs in HCFs and induce cell injury through this process. In whole-cell mode patch-clamp recordings, application of H2O2 stimulated Ca2+-activated K+ (K(Ca)) currents but not delayed rectifier K+ or transient outward K+ currents, all of which are VDKCs. H2O2-stimulated K(Ca) currents were blocked by iberiotoxin (IbTX, a large conductance K(Ca) blocker). The H2O2-stimulating effect on large-conductance K(Ca) (BK(Ca)) currents was also blocked by KT5823 (a protein kinase G inhibitor) and 1 H-[1, 2, 4] oxadiazolo-[4, 3-a] quinoxalin-1-one (ODQ, a soluble guanylate cyclase inhibitor). In addition, 8-bromo-cyclic guanosine 3', 5'-monophosphate (8-Br-cGMP) stimulated BK(Ca) currents. In contrast, KT5720 and H-89 (protein kinase A inhibitors) did not block the H2O2-stimulating effect on BK(Ca) currents. Using RT-PCR and western blot analysis, three subtypes of K(Ca) channels were detected in HCFs: BK(Ca) channels, small-conductance K(Ca) (SK(Ca)) channels, and intermediate-conductance K(Ca) (IK(Ca)) channels. In the annexin V/propidium iodide assay, apoptotic changes in HCFs increased in response to H2O2, but IbTX decreased H2O2-induced apoptosis. These data suggest that among the VDKCs of HCFs, H2O2 only enhances BK(Ca) currents through the protein kinase G pathway but not the protein kinase A pathway, and is involved in cell injury through BK(Ca) channels.


Subject(s)
Humans , Apoptosis , Blotting, Western , Cyclic AMP-Dependent Protein Kinases , Cyclic GMP-Dependent Protein Kinases , Fibroblasts , Guanosine , Guanylate Cyclase , Hydrogen Peroxide , Hydrogen , Oxidative Stress , Phosphotransferases , Potassium Channels, Calcium-Activated , Protein Kinases
3.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 264-273, 2015.
Article in English | WPRIM | ID: wpr-812148

ABSTRACT

Scutellarin (SCU), a flavonoid from a traditional Chinese medicinal plant. Our previous study has demonstrated that SCU relaxes mouse aortic arteries mainly in an endothelium-depend-ent manner. In the present study, we investigated the vasoprotective effects of SCU against HR-induced endothelial dysfunction (ED) in isolated rat CA and the possible mechanisms involving cyclic guanosine monophosphate (cGMP) dependent protein kinase (PKG). The isolated endothelium-intact and endothelium-denuded rat CA rings were treated with HR injury. Evaluation of endothelium-dependent and -independent vasodilation relaxation of the CA rings were performed using wire myography and the protein expressions were assayed by Western blotting. SCU (10-1 000 μmol·L(-1)) could relax the endothelium-intact CA rings but not endothelium-denuded ones. In the intact CA rings, the PKG inhibitor, Rp-8-Br-cGMPS (PKGI-rp, 4 μmol·L(-1)), significantly blocked SCU (10-1 000 μmol·L(-1))-induced relaxation. The NO synthase (NOS) inhibitor, NO-nitro-L-arginine methylester (L-NAME, 100 μmol·L(-1)), did not significantly change the effects of SCU (10-1 000 μmol·L(-1)). HR treatment significantly impaired ACh-induced relaxation, which was reversed by pre-incubation with SCU (500 μmol·L(-1)), while HR treatment did not altered NTG-induced vasodilation. PKGI-rp (4 μmol·L(-1)) blocked the protective effects of SCU in HR-treated CA rings. Additionally, HR treatment reduced phosphorylated vasodilator-stimulated phosphoprotein (p-VASP, phosphorylated product of PKG), which was reversed by SCU pre-incubation, suggesting that SCU activated PKG phosphorylation against HR injury. SCU induces CA vasodilation in an endothelium-dependent manner to and repairs HR-induced impairment via activation of PKG signaling pathway.


Subject(s)
Animals , Rats , Apigenin , Pharmacology , Cell Adhesion Molecules , Cell Hypoxia , Coronary Vessels , Cyclic GMP , Metabolism , Pharmacology , Cyclic GMP-Dependent Protein Kinases , Glucuronates , Pharmacology , Microfilament Proteins , NG-Nitroarginine Methyl Ester , Metabolism , Pharmacology , Phosphoproteins , Rats, Sprague-Dawley , Reperfusion Injury , Signal Transduction , Thionucleotides , Metabolism , Pharmacology , Vasodilation , Physiology
4.
Braz. j. med. biol. res ; 47(10): 876-885, 10/2014. tab, graf
Article in English | LILACS | ID: lil-722165

ABSTRACT

The aim of the present study was to determine the mechanisms underlying the relaxant effect of adrenomedullin (AM) in rat cavernosal smooth muscle (CSM) and the expression of AM system components in this tissue. Functional assays using standard muscle bath procedures were performed in CSM isolated from male Wistar rats. Protein and mRNA levels of pre-pro-AM, calcitonin receptor-like receptor (CRLR), and Subtypes 1, 2 and 3 of the receptor activity-modifying protein (RAMP) family were assessed by Western immunoblotting and quantitative real-time polymerase chain reaction, respectively. Nitrate and 6-keto-prostaglandin F1α (6-keto-PGF1α; a stable product of prostacyclin) levels were determined using commercially available kits. Protein and mRNA of AM, CRLR, and RAMP 1, -2, and -3 were detected in rat CSM. Immunohistochemical assays demonstrated that AM and CRLR were expressed in rat CSM. AM relaxed CSM strips in a concentration-dependent manner. AM22-52, a selective antagonist for AM receptors, reduced the relaxation induced by AM. Conversely, CGRP8-37, a selective antagonist for calcitonin gene-related peptide receptors, did not affect AM-induced relaxation. Preincubation of CSM strips with NG-nitro-L-arginine-methyl-ester (L-NAME, nitric oxide synthase inhibitor), 1H-(1,2,4)oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, quanylyl cyclase inhibitor), Rp-8-Br-PET-cGMPS (cGMP-dependent protein kinase inhibitor), SC560 [5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-trifluoromethyl pyrazole, selective cyclooxygenase-1 inhibitor], and 4-aminopyridine (voltage-dependent K+ channel blocker) reduced AM-induced relaxation. On the other hand, 7-nitroindazole (selective neuronal nitric oxide synthase inhibitor), wortmannin (phosphatidylinositol 3-kinase inhibitor), H89 (protein kinase A inhibitor), SQ22536 [9-(tetrahydro-2-furanyl)-9H-purin-6-amine, adenylate cyclase inhibitor], glibenclamide (selective blocker of ATP-sensitive K+ channels), and apamin (Ca2+-activated channel blocker) did not affect AM-induced relaxation. AM increased nitrate levels and 6-keto-PGF1α in rat CSM. The major new contribution of this research is that it demonstrated expression of AM and its receptor in rat CSM. Moreover, we provided evidence that AM-induced relaxation in this tissue is mediated by AM receptors by a mechanism that involves the nitric oxide-cGMP pathway, a vasodilator prostanoid, and the opening of voltage-dependent K+ channels.


Subject(s)
Animals , Male , Adrenomedullin/pharmacology , Calcitonin Receptor-Like Protein/analysis , Muscle, Smooth/drug effects , Parasympatholytics/pharmacology , Penis/drug effects , Vasodilator Agents/pharmacology , /pharmacology , /analysis , Adrenomedullin/genetics , Adrenomedullin/metabolism , Blotting, Western , Calcitonin Receptor-Like Protein/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Indazoles/pharmacology , Muscle Relaxation , Muscle, Smooth/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/analysis , Nitric Oxide/analogs & derivatives , Penis/metabolism , Potassium Channels, Voltage-Gated/metabolism , Rats, Wistar , Real-Time Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptor Activity-Modifying Protein 1/genetics , Receptor Activity-Modifying Protein 1/metabolism , /metabolism , /genetics , /metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism
5.
Acta cir. bras ; 28(7): 537-542, July 2013. ilus, graf
Article in English | LILACS | ID: lil-679087

ABSTRACT

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.


Subject(s)
Animals , Male , Rats , Calcium/metabolism , Cyclic GMP-Dependent Protein Kinases/physiology , Mesenteric Artery, Superior/metabolism , Shock, Hemorrhagic/metabolism , Blotting, Western , Calcium/analysis , Cyclic GMP-Dependent Protein Kinases/analysis , Enzyme-Linked Immunosorbent Assay , Muscle Contraction , Mesenteric Artery, Superior/physiopathology , Random Allocation , Rats, Wistar , Shock, Hemorrhagic/physiopathology
6.
Experimental & Molecular Medicine ; : e65-2013.
Article in English | WPRIM | ID: wpr-152459

ABSTRACT

Vascular smooth muscle cells (VSMCs) undergo phenotypic changes in response to vascular injury such as angioplasty. Protein kinase G (PKG) has an important role in the process of VSMC phenotype switching. In this study, we examined whether rosiglitazone, a peroxisome proliferator-activated receptor (PPAR)-gamma agonist, could modulate VSMC phenotype through the PKG pathway to reduce neointimal hyperplasia after angioplasty. In vitro experiments showed that rosiglitazone inhibited the phenotype change of VSMCs from a contractile to a synthetic form. The platelet-derived growth factor (PDGF)-induced reduction of PKG level was reversed by rosiglitazone treatment, resulting in increased PKG activity. This increased activity of PKG resulted in phosphorylation of vasodilator-stimulated phosphoprotein at serine 239, leading to inhibited proliferation of VSMCs. Interestingly, rosiglitazone did not change the level of nitric oxide (NO) or cyclic guanosine monophosphate (cGMP), which are upstream of PKG, suggesting that rosiglitazone influences PKG itself. Chromatin immunoprecipitation assays for the PKG promoter showed that the activation of PKG by rosiglitazone was mediated by the increased binding of Sp1 on the promoter region of PKG. In vivo experiments showed that rosiglitazone significantly inhibited neointimal formation after balloon injury. Immunohistochemistry staining for calponin and thrombospondin showed that this effect of rosiglitazone was mediated by modulating VSMC phenotype. Our findings demonstrate that rosiglitazone is a potent modulator of VSMC phenotype, which is regulated by PKG. This activation of PKG by rosiglitazone results in reduced neointimal hyperplasia after angioplasty. These results provide important mechanistic insight into the cardiovascular-protective effect of PPARgamma.


Subject(s)
Animals , Rats , Aorta/injuries , Calcium-Binding Proteins/genetics , Cell Proliferation , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Hyperplasia/metabolism , Microfilament Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Nitric Oxide/metabolism , PPAR gamma/agonists , Promoter Regions, Genetic , Rats, Sprague-Dawley , Sp1 Transcription Factor/metabolism , Thiazolidinediones/pharmacology , Thrombospondins/genetics , Tunica Intima/metabolism , Vascular System Injuries/metabolism
7.
Experimental & Molecular Medicine ; : 363-368, 2012.
Article in English | WPRIM | ID: wpr-57564

ABSTRACT

Dendroaspis natriuretic peptide (DNP), a new member of the natriuretic peptide family, is structurally similar to atrial, brain, and C-type natriuretic peptides. However, the effects of DNP on the cardiac function are poorly defined. In the present study, we examined the effect of DNP on the cardiac L-type Ca2+ channels in rabbit ventricular myocytes. DNP inhibited the L-type Ca2+ current (ICa,L) in a concentration dependent manner with a IC50 of 25.5 nM, which was blocked by an inhibitor of protein kinase G (PKG), KT5823 (1 microM). DNP did not affect the voltage dependence of activation and inactivation of ICa,L. The alpha1c subunit of cardiac L-type Ca2+ channel proteins was phosphorylated by the treatment of DNP (1 microM), which was completely blocked by KT5823 (1 microM). Finally, DNP also caused the shortening of action potential duration in rabbit ventricular tissue by 22.3 +/- 4.2% of the control (n = 6), which was completely blocked by KT5823 (1 microM). These results clearly indicate that DNP inhibits the L-type Ca2+ channel activity by phosphorylating the Ca2+ channel protein via PKG activation.


Subject(s)
Animals , Rabbits , Action Potentials/drug effects , Biological Transport/drug effects , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Carbazoles/pharmacology , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Elapid Venoms/metabolism , Enzyme Activation , Heart , Heart Ventricles/drug effects , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Peptides/metabolism , Phosphorylation/drug effects
8.
Acta Physiologica Sinica ; (6): 563-576, 2012.
Article in English | WPRIM | ID: wpr-333166

ABSTRACT

Injury or inflammation affecting sensory neurons in the dorsal root ganglia (DRG) causes hyperexcitability of DRG neurons that can lead to spinal central sensitization and neuropathic pain. Recent studies have indicated that, following chronic compression of DRG (CCD) or acute dissociation of DRG (ADD) treatment, both hyperexcitability of neurons in intact DRG and behaviorally expressed hyperalgesia are maintained by activity in cGMP-PKG signaling pathway. Here, we provide evidence supporting the idea that CCD or ADD treatment activates cGMP-PKA signaling pathway in the DRG neurons. The results showed that CCD or ADD results in increase of levels of cGMP concentration and expression of PKG-I mRNA, as well as PKG-I protein in DRG. CCD or ADD treated-DRG neurons become hyperexcitable and exhibit increased responsiveness to the activators of cGMP-PKG pathway, 8-Br-cGMP and Sp-cGMP. Hyperexcitability of the injured neurons is inhibited by cGMP-PKG pathway inhibitors, ODQ and Rp-8-pCPT-cGMPS. In vivo delivery of Rp-8-pCPT-cGMPS into the compressed ganglion within the intervertebral foramen suppresses CCD-induced thermal hyperalgesia. These findings indicate that the in vivo CCD or in vitro ADD treatment can activate the cGMP-PKG signaling pathway, and that continuing activation of cGMP-PKG pathway is required to maintain DRG neuronal hyperexcitability and/or hyperalgesia after these two dissimilar forms of injury-related stress.


Subject(s)
Animals , Rats , Cyclic GMP , Metabolism , Cyclic GMP-Dependent Protein Kinases , Metabolism , Ganglia, Spinal , Hyperalgesia , Rats, Sprague-Dawley , Signal Transduction , Thionucleotides , Metabolism
9.
Chinese Journal of Cardiology ; (12): 858-863, 2012.
Article in Chinese | WPRIM | ID: wpr-326404

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the underlying mechanism of the protective effects of resveratrol on oxidant-induced mitochondrial damage in embryonic rat cardiomyocytes.</p><p><b>METHODS</b>H9c2 cells, a permanent cell line derived from embryonic rat cardiac tissue, and then randomly divided into control group [PBS, cells exposed to H2O2 (600 µmol/L) for 20 min to induce mitochondrial oxidant damage], resveratrol group (0.01, 0.1, 1, 5, 10 and 20 µmol/L for 20 min at 20 min before exposing to H2O2), resveratrol plus inhibitor group (1 µmol/L KT5823 for 10 min at 10 min before 5 µmol/L resveratrol treatment) and inhibitor group (1 µmol/L KT5823 for 10 min). Mitochondrial membrane potential (ΔΨm) was measured by staining cells with tetramethylrhodamine ethyl ester (TMRE) and the mitochondrial permeability transition pore (mPTP) opening was evaluated by measuring the decrease of TMRE fluorescence intensity. Immunofluorescence assay was used to observe GSK-3β phosphorylation. The phosphorylation of GSK-3β and VASP were determined by Western blot. To detect intracellular NO, cells were loaded with DAF-FM DA (specific fluorescent dye of NO) and imaged with confocal microscopy.</p><p><b>RESULTS</b>Compared to the control group, resveratrol (0.01-5 µmol/L) attenuated H2O2-induced mitochondrial damage reflected by attenuating the H2O2-induced TMRE fluorescence intensity decrease in a dose-dependent manner and the efficacy of 10 and 20 µmol/L resveratrol was significantly lower than that of 5 µmol/L resveratrol. Resveratrol also significantly upregulated the protein expression of VASP and increased GSK-3β Ser(9) phosphorylation, which could lead the inactivation of GSK-3β. These effects of resveratrol could be significantly abolished by protein kinase G inhibitor KT5823, while KT5823 alone did not affect GSK-3β and VASP phosphorylation. Confocal microscopy showed that DAF-FM (specific NO indicator) was similar between resveratrol and control group, suggesting that resveratrol did not produce NO.</p><p><b>CONCLUSIONS</b>Resveratrol could attenuate oxidant-induced mitochondrial damage in embryonic rat cardiomyocytes by inactivating GSK-3β via cGMP/PKG signaling pathway independent of NO-related mechanism.</p>


Subject(s)
Animals , Rats , Carbazoles , Pharmacology , Cell Line , Cyclic GMP , Metabolism , Cyclic GMP-Dependent Protein Kinases , Metabolism , Glycogen Synthase Kinase 3 , Metabolism , Glycogen Synthase Kinase 3 beta , Hydrogen Peroxide , Metabolism , Mitochondria, Heart , Metabolism , Myocytes, Cardiac , Cell Biology , Oxidants , Metabolism , Signal Transduction , Stilbenes , Pharmacology
10.
The Korean Journal of Physiology and Pharmacology ; : 211-217, 2012.
Article in English | WPRIM | ID: wpr-728098

ABSTRACT

Recent studies have demonstrated that nitric oxide (NO) activates transient receptor potential vanilloid subtype 1 (TRPV1) via S-nitrosylation of the channel protein. NO also modulates various cellular functions via activation of the soluble guanylyl cyclase (sGC)/protein kinase G (PKG) pathway and the direct modification of proteins. Thus, in the present study, we investigated whether NO could indirectly modulate the activity of TRPV1 via a cGMP/PKG-dependent pathway in cultured rat dorsal root ganglion (DRG) neurons. NO donors, sodium nitroprusside (SNP) and S-nitro-N-acetylpenicillamine (SNAP), decreased capsaicin-evoked currents (Icap). NO scavengers, hemoglobin and 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (CPTIO), prevented the inhibitory effect of SNP on Icap. Membrane-permeable cGMP analogs, 8-bromoguanosine 3', 5'-cyclic monophosphate (8bromo-cGMP) and 8-(4chlorophenylthio)-guanosine 3',5'-cyclic monophosphate (8-pCPT-cGMP), and the guanylyl cyclase stimulator YC-1 mimicked the effect of SNP on Icap. The PKG inhibitor KT5823 prevented the inhibition of Icap by SNP. These results suggest that NO can downregulate the function of TRPV1 through activation of the cGMP/PKG pathway in peripheral sensory neurons.


Subject(s)
Animals , Humans , Rats , Benzoates , Carbazoles , Cyclic GMP-Dependent Protein Kinases , Ganglia, Spinal , Guanosine , Guanylate Cyclase , Hemoglobins , Imidazoles , Neurons , Nitric Oxide , Nitroprusside , Penicillamine , Phosphotransferases , Proteins , Receptors, Cytoplasmic and Nuclear , Sensory Receptor Cells , Spinal Nerve Roots , Tissue Donors
11.
Experimental & Molecular Medicine ; : 749-755, 2012.
Article in English | WPRIM | ID: wpr-110117

ABSTRACT

Cinnamyl alcohol (CAL) is known as an antipyretic, and a recent study showed its vasodilatory activity without explaining the mechanism. Here we demonstrate the vasodilatory effect and the mechanism of action of CAL in rat thoracic aorta. The change of tension in aortic strips treated with CAL was measured in an organ bath system. In addition, vascular strips or human umbilical vein endothelial cells (HUVECs) were used for biochemical experiments such as Western blot and nitrite and cyclic guanosine monophosphate (cGMP) measurements. CAL attenuated the vasoconstriction of phenylephrine (PE, 1 microM)-precontracted aortic strips in an endothelium-dependent manner. CAL-induced vasorelaxation was inhibited by pretreatment with NG-nitro-L-arginine methyl ester (L-NAME; 10(-4) M), methylene blue (MB; 10(-5) M) and 1 H-[1,2,4]-oxadiazolole-[4,3-a] quinoxalin-10one, (ODQ; 10(-6) or 10(-7) M) in the endothelium-intact aortic strips. Atrial natriuretic peptide (ANP; 10(-8) or 10(-9) M) did not affect the vasodilatory effect of CAL. The phosphorylation of endothelial nitric oxide synthase (eNOS) and generation of nitric oxide (NO) were stimulated by CAL treatment in HUVECs and inhibited by treatment with L-NAME. In addition, cGMP and PKG1 activation in aortic strips treated with CAL were also significantly inhibited by L-NAME. Furthermore, CAL relaxed Rho-kinase activator calpeptin-precontracted aortic strips, and the vasodilatory effect of CAL was inhibited by the ATP-sensitive K+ channel inhibitor glibenclamide (Gli; 10(-5) M) and the voltage-dependent K+ channel inhibitor 4-aminopyridine (4-AP; 2 x 10(-4) M). These results suggest that CAL induces vasorelaxation by activating K+ channels via the NO-cGMP-PKG pathway and the inhibition of Rho-kinase.


Subject(s)
Animals , Humans , Male , Rats , Aorta/drug effects , Atrial Natriuretic Factor/pharmacology , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Dipeptides/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Methylene Blue/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Oxadiazoles/pharmacology , Phenylephrine/pharmacology , Phosphorylation , Potassium Channel Blockers/pharmacology , Potassium Channels/agonists , Propanols/pharmacology , Quinoxalines/pharmacology , Rats, Sprague-Dawley , Signal Transduction , Vasoconstriction/drug effects , Vasodilation/drug effects , rho-Associated Kinases/antagonists & inhibitors
12.
An. acad. bras. ciênc ; 83(2): 637-648, June 2011. ilus
Article in English | LILACS | ID: lil-589920

ABSTRACT

Schistosomes are trematode parasites and of worldwide medical importance for humans and animals. Growth and development of these parasites require a specific host environment, but also permanent communication processes between the two genders. Accumulating molecular evidence indicates that the responsible interactions are mediated by signal transduction processes. Conserved signaling molecules were identified, and first approaches made for their characterization. However, no representative of the conserved family of cGMP-dependent protein kinases (cGKs) has been described in this parasite yet. Within the Schistosoma mansoni genome data-set we identified cGK homologs, of which one was investigated in more detail in this study. We present the cloning of SmcGK1, whose sequence shows homology to cGKs of higher eukaryotes. SmcGK1 was found to be gender-independently transcribed in adult schistosomes. The occurrence of SmcGK1 sense and antisense transcripts suggests that the expression of this gene is controlled at the post-transcriptional level. In situ hybridization experiments demonstrated a gonad-preferential expression profile in both genders indicating a role of SmcGK1, at least during sexual development of schistosomes. Using a cGK-specific inhibitor to treat adult schistosomes in vitro finally resulted in a multifaceted phenotype including slow motion, oocyte congestion, and reduced egg production.


Esquistossomos são parasitas trematodos de importância médica em todo o mundo para o homem e os animais. O crescimento e o desenvolvimento destes parasitas requerem um ambiente específico do hospedeiro, mas também um processo de comunicação permanente entre parasitas dos dois sexos. Evidência molecular tem se acumulado e indica que as interações são mediadas por processos de transdução de sinal. Moléculas sinalizadoras conservadas foram identificadas, e as primeiras abordagens têm sido feitas para sua caracterização. Contudo, não foi ainda descrito nenhum representante da família conservada das proteína-quinases dependentes de cGMP (cGKs) neste parasita. Analisando o genoma do Schistosoma mansoni nós identificamos homólogos de cGK, dos quais um foi investigado em mais detalhe no presente estudo. Aqui apresentamos a clonagem do gene SmcGK1, cuja sequência mostra homologia com cGKs de eucariotos superiores. Smc- GK1 foi detectada como sendo transcrita de forma gêneroindependente em esquistossomos adultos. A ocorrência de transcritos de SmcGK1 senso e antisenso sugere que a expressão deste gene é controlada em nível pos-transcricional. Experimentos de hibridização in situ demonstraram uma expressão preferencial nas gônadas em ambos os gêneros, indicando um papel para SmcGK1, pelo menos durante o desenvolvimento de esquistossomos. Usando um inibidor específico de cGK para tratamento de esquistossomos adultos in vitro finalmente resultou em um fenótipo multifacetado, incluindo movimentos lentos, congestão dos oócitos, e redução da produção de ovos.


Subject(s)
Animals , Female , Male , Cyclic GMP-Dependent Protein Kinases/genetics , Gonads/metabolism , Oocytes/metabolism , Schistosoma mansoni/enzymology , Base Sequence , Cloning, Molecular , Cyclic GMP-Dependent Protein Kinases/metabolism , DNA, Complementary/genetics , In Situ Hybridization , Molecular Sequence Data , Schistosoma mansoni/genetics , Signal Transduction/genetics
13.
The Korean Journal of Physiology and Pharmacology ; : 37-43, 2010.
Article in English | WPRIM | ID: wpr-727343

ABSTRACT

The serine/threonine kinase Akt has been shown to play a role of multiple cellular signaling pathways and act as a transducer of many functions initiated by growth factor receptors that activate phosphatidylinositol 3-kinase (PI3K). It has been reported that phosphorylated Akt activates eNOS resulting in the production of NO and that NO stimulates soluble guanylate cyclase (sGC), which results in accumulation of cGMP and subsequent activation of the protein kinase G (PKG). It has been also reported that PKG activates PI3K/Akt signaling. Therefore, it is possible that PI3K, Akt, eNOS, sGC, and PKG form a loop to exert enhanced and sustained activation of Akt. However, the existence of this loop in eNOS-expressing cells, such as endothelial cells or astrocytes, has not been reported. Thus, we examined a possibility that Akt phosphorylation might be enhanced via eNOS/sGC/PKG/PI3K pathway in astrocytes in vivo and in vitro. Phosphorylation of Akt was detected in astrocytes after KA treatment and was maintained up to 72 h in mouse hippocampus. 2 weeks after KA treatment, astrocytic Akt phosphorylation was normalized to control. The inhibition of eNOS, sGC, and PKG significantly decreased Akt and eNOS phosphorylation induced by KA in astrocytes. In contrast, the decreased phosphorylation of Akt and eNOS by eNOS inhibition was significantly reversed with PKG activation. The above findings in mouse hippocampus were also observed in primary astrocytes. These data suggest that Akt/eNOS/sGC/PKG/PI3K pathway may constitute a loop, resulting in enhanced and sustained Akt activation in astrocytes.


Subject(s)
Animals , Mice , Astrocytes , Cyclic GMP-Dependent Protein Kinases , Endothelial Cells , Guanylate Cyclase , Hippocampus , Kainic Acid , Nitric Oxide , Phosphatidylinositol 3-Kinase , Phosphorylation , Phosphotransferases , Receptors, Growth Factor , Transducers
14.
Journal of Veterinary Science ; : 103-106, 2010.
Article in English | WPRIM | ID: wpr-221274

ABSTRACT

Sildenafil increases the cyclic guanosine monophosphate (cGMP) by inhibition of a phosphodiesterase 5, thereby leading to an antinociceptive effect. The increased cGMP may exert the effect on an L-type calcium channel through the activation of protein kinase G (PKG). The purpose of this study was to examine the possible involvement of a PKG-L-type calcium channel on the effect of sildenafil at the spinal level. Catheters were inserted into the intrathecal space of male SD rats. Pain was induced by applying 50 microliter of a 5% formalin solution to the hindpaw. The sildenafil-induced effect was examined after an intrathecal pretreatment of a PKG inhibitor (KT 5823), or a L-type calcium channel activator (FPL 64176). Intrathecal sildenafil produced an antinociceptive effect during phase 1 (0~10 min interval) and phase 2 (10~60 min interval) in the formalin test. Intrathecal KT 5823 and FPL 64176 attenuated the antinociceptive effect of sildenafil during both phases. Sildenafil is effective against both acute pain and the facilitated pain state at the spinal level. In addition, the inhibition of an L-type calcium channel by activation of the PKG may contribute to the antinocieptive mechanism of sildenafil in the spinal cord.


Subject(s)
Animals , Male , Rats , Calcium Channel Agonists/pharmacology , Calcium Channels, L-Type/physiology , Carbazoles/pharmacology , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Pain/drug therapy , Pain Measurement , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyrroles/pharmacology , Rats, Sprague-Dawley , Sulfones/pharmacology
15.
Braz. j. med. biol. res ; 42(1): 87-93, Jan. 2009. ilus, tab, graf
Article in English | LILACS | ID: lil-505424

ABSTRACT

Nitric oxide (NO) donors produce NO-related activity when applied to biological systems. Among its diverse functions, NO has been implicated in vascular smooth muscle relaxation. Despite the great importance of NO in biological systems, its pharmacological and physiological studies have been limited due to its high reactivity and short half-life. In this review we will focus on our recent investigations of nitrosyl ruthenium complexes as NO-delivery agents and their effects on vascular smooth muscle cell relaxation. The high affinity of ruthenium for NO is a marked feature of its chemistry. The main signaling pathway responsible for the vascular relaxation induced by NO involves the activation of soluble guanylyl-cyclase, with subsequent accumulation of cGMP and activation of cGMP-dependent protein kinase. This in turn can activate several proteins such as K+ channels as well as induce vasodilatation by a decrease in cytosolic Ca2+. Oxidative stress and associated oxidative damage are mediators of vascular damage in several cardiovascular diseases, including hypertension. The increased production of the superoxide anion (O2-) by the vascular wall has been observed in different animal models of hypertension. Vascular relaxation to the endogenous NO-related response or to NO released from NO deliverers is impaired in vessels from renal hypertensive (2K-1C) rats. A growing amount of evidence supports the possibility that increased NO inactivation by excess O2- may account for the decreased NO bioavailability and vascular dysfunction in hypertension.


Subject(s)
Animals , Rats , Cyclic GMP-Dependent Protein Kinases/drug effects , Muscle, Smooth, Vascular/drug effects , Nitric Oxide Donors/pharmacology , Ruthenium/pharmacology , Aorta/drug effects , Calcium Channels/drug effects , Calcium Channels/physiology , Cyclic GMP-Dependent Protein Kinases/metabolism , Hypertension, Renal/physiopathology , Muscle Relaxation , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide/metabolism , Potassium Channels/drug effects , Potassium Channels/physiology , Ruthenium/chemistry , Signal Transduction/drug effects , Time Factors , Vasodilation/drug effects , Vasodilation/physiology
16.
Korean Journal of Anesthesiology ; : 716-722, 2008.
Article in Korean | WPRIM | ID: wpr-159722

ABSTRACT

BACKGROUND: This experiments investigated the signaling cascade responsible for anti-infarct effect by an A2 adenosine receptor (AR) agonist 5'-N-Ethylcarboxaminidoadenosine (NECA). METHODS: Langendorff perfused isolated rat hearts were subjected to 30 minutes of regional ischemia and 120 minutes of reperfusion. Drugs were perfused for a period of 5 minutes before and 60 minutes after reperfusion. For comparison of cardioprotection among groups, area at necrosis (AN) and area at risk (AAR) were measured by triphenyltetrazolium chloride staining. RESULTS: NECA significantly attenuated AN/AAR (14.1 +/- 1.9%, P < 0.001) compared with control hearts (30.7 +/- 2.8%). Anti-infarct effect by NECA was attenuated by an A(2A)AR antagonist 8-(3-chlorostyryl)caffeine (23.7 +/- 3.4%, P < 0.05) and an A(2B)AR antagonist MRS1706 (29.9 +/- 3.3%, P < 0.001). Cardioprotection by NECA was blocked by a guanylyl cyclase inhibitor (23.1 +/- 2.9%, P < 0.05) and a protein kinase G (PKG) inhibitor KT5823 (30.3 +/- 3.2%, P < 0.001). Glycogen synthase kinase-3beta (GSK-3beta) inhibitor SB216763 attenuated the AN/AAR in both NECA with MRS (17.8 +/- 2.7%, P < 0.01 vs. control) and NECA with KT5823 treated hearts (8.2 +/- 1.8%, P < 0.001 vs. control). The mitochondrial permeability transition pore (mPTP) opener atractyloside also aborted NECA's anti-infarct effect (24.7 +/- 2.4% P < 0.05). CONCLUSIONS: The signaling pathway by NECA administered at reperfusion involves the activation of both A2AAR and A2BAR and cGMP/PKG pathway, which in turn depends on inactivation of GSK-3beta and inhibition of mPTP opening.


Subject(s)
Animals , Rats , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Adenosine , Adenosine-5'-(N-ethylcarboxamide) , Atractyloside , Caffeine , Carbazoles , Cyclic GMP-Dependent Protein Kinases , Glycogen Synthase , Glycogen Synthase Kinase 3 , Guanylate Cyclase , Heart , Indoles , Ischemia , Maleimides , Mitochondria , Mitochondrial Membrane Transport Proteins , Myocardial Infarction , Myocardial Reperfusion , Myocardial Reperfusion Injury , Necrosis , Permeability , Purines , Receptors, Purinergic P1 , Reperfusion , Reperfusion Injury , Tetrazolium Salts
17.
Asian Journal of Andrology ; (6): 675-684, 2006.
Article in English | WPRIM | ID: wpr-253778

ABSTRACT

The pathophysiology of diabetes is multifactorial and no single etiology is at the forefront. The proposed mechanisms of erectile dysfunction (ED) in diabetic patients includes elevated advanced glycation end-products (AGEs) and increased levels of oxygen free radicals, impaired nitric oxide (NO) synthesis, increased endothelin B receptor binding sites and ultrastructural changes, upregulated RhoA/Rho-kinase pathway, NO-dependent selective nitrergic nerve degeneration and impaired cyclic guanosine monophosphate (cGMP)-dependent kinase-1 (PKG-1). The treatment of diabetic ED is multimodal. Treatment of the underlying hyperglycemia and comorbidities is of utmost importance to prevent or halt the progression of the disease. The peripherally acting oral phosphodiesterase type 5 (PDE5) inhibitors are the mainstay of oral medical treatment of ED in diabetics. Vacuum erection devices are an additional treatment as a non-invasive treatment option. Local administration of vasoactive medication via urethral suppository or intracorporal injection can be effective with minimal side-effects. Patients with irreversible damage of the erectile mechanism are candidates for penile implantation. Future strategies in the evolution of the treatment of ED are aimed at correcting or treating the underlying mechanisms of ED. With an appropriate vector, researchers have been able to transfect diabetic animals with agents such as neurotrophic factors and nitric oxide synthase (NOS). Further studies in gene therapy are needed to fully ascertain its safety and utility in humans.


Subject(s)
Humans , Male , Alprostadil , Therapeutic Uses , Cyclic GMP-Dependent Protein Kinases , Physiology , Diabetes Complications , Diabetic Neuropathies , Erectile Dysfunction , Therapeutics , Glycation End Products, Advanced , Physiology , Nitric Oxide , Physiology , Penile Erection , Penile Prosthesis , Penis , Phosphodiesterase Inhibitors , Therapeutic Uses , Receptor, Endothelin B , Physiology , Suppositories , Vacuum
18.
Gac. méd. Méx ; 141(6): 513-526, nov.-dic. 2005. ilus
Article in Spanish | LILACS | ID: lil-632126

ABSTRACT

Los procesos neurofisiológicos, bioquímicos y moleculares descritos en la integración de la memoria, más que estar relacionados con la actividad colinérgica involucran fundamentalmente a neurotransmisores como la serotonina y el glutamato, así como a diversos canales iónicos como los del calcio y los del potasio. De hecho, los receptores de estos neurotransmisores están ligados directamente con la activación de la potenciación a largo plazo (LTP), mecanismo que contribuye a la preservación de la memoria. De esta forma que la activación del receptor 5HT desencadena una señal de transducción que al influenciar bioquímicamente al núcleo produce diversos cambios presinápticos con los que se expulsa al magnesio del área postsináptica, despolarizando a la neurona y activando simultáneamente a los receptores N metilD Aspartato dependientes (NMDAR), contribuyendo en esta forma a perpetuar el mecanismo de LTP en sus distintas fases: LTP1 que depende de la activación de proteincinasas; LTP2 ligada con la traslación genética; y LTP3 relacionada con la transcripción. A este poderoso mecanismo de activación neuronal, se contrapone el fenómeno de depresión a largo plazo (LTD), que se inicia cuando la neurona pre sináptica activa al inhibidor 1 en el momento en que detecta una reducción en el influjo de calcio, promoviendo en esta forma la defosforilación de una proteincinasa tipo II calcio calmodulin dependiente, lo que detiene el desarrollo del proceso de autofosforilación y con ello, el mecanismo de LTP. No obstante lo difundido de la hipótesis colinérgica en la enfermedad de Alzheimer, la integración de la memoria depende fundamentalmente de la intervención de otros sistemas de neurotransmisión como lo son el serotonérgico y el glutamatérgico, los que no han sido debidamente considerados en el tratamiento de esta enfermedad; sin embargo más allá de estos sistemas, se encuentran los mecanismos de autofosforilación de distintas proteincinasas cuyo control, además de repercutir sobre la expresión genética, podría restituir algunos de los trastornos que afectan la función cognoscitiva.


Neurophysiological, biochemical and molecular processes described in the integration of memory are closely related with neurotransmitters such as glutamate and serotonin (5HT) and with the function of calcium and potassium ion channels more than with cholinergic activity. In fact, glutamate and 5 HT receptors are closely related with Long-Term Potentiation (LTP) processes, the mechanism by which memory is preserved throughout time. That is, the activation of the 5 HT4 receptor triggers a transduction signal that after influencing nuclear cell activity, provokes several presynaptic changes, which leads to the displacement of magnesium from the postsynaptic area depolarizing the neuron and leading to the activation of N methyl -D-aspartate receptors (NMDA). As a whole, this process contributes to the support and perpetuation of LTP, which consists of the following processes: LTP1 that depends on protein kinase activity; LTP2 linked to translation of genes; and LTP3 closely related to genes transcription. On the opposite side but in perfect balance, we find the mechanism of Long Term depression (LTD), which is triggered instead when the Ca++ flow decreases in the presynaptic neuron activating the inhibitor 1 enzyme that promotes the dephosphorylation of a calmodulin dependent protein kinase II and as a result, the inhibition of autophosphorylation and consequently of LTP too. Despite the widespread dissemination of the cholinergic hypothesis in Alzheimer's disease, memory build up rather than involving acetylcholine essentially depends on the participation of other neurotransmitters such as 5 HT and glutamate, which have not been adequately considered in the treatment of this disease. However, beyond neurotransmission, it is the cellular mechanism of autophosphorylation of several protein kinases, the process susceptible of being activated or controlled by the action of distinct substances. In such a case, it would be possible to exert some influence on gene expression improving perhaps, some of the physiopathological deficits that characterize memory disruption.


Subject(s)
Humans , Memory/physiology , Signal Transduction/physiology , Calmodulin/physiology , Cyclic GMP-Dependent Protein Kinases/physiology , Intracellular Signaling Peptides and Proteins/physiology , Nucleotides, Cyclic/physiology , Receptors, Cholinergic/physiology , Receptors, Neurotransmitter/physiology
19.
Mem. Inst. Oswaldo Cruz ; 98(4): 499-500, June 2003. graf
Article in English | LILACS | ID: lil-344242

ABSTRACT

Although there are some data concerning the nitric oxide and the cyclic 3'-5'guanosine monophosphate (cGMP) signaling pathway in trypanosomatids, there is no report about the cGMP-dependent enzymatic activity identification. In this sense, a cGMP dependent activity was detected on soluble fraction from Leishmania amazonensis promastigotes with a high metacyclic level. This information is valuable in order to explore the metabolic pathway of G kinase protein in this parasite


Subject(s)
Animals , Cyclic GMP-Dependent Protein Kinases , Leishmania mexicana , Subcellular Fractions
20.
Journal of the Korean Surgical Society ; : 194-200, 2003.
Article in Korean | WPRIM | ID: wpr-125361

ABSTRACT

PURPOSE: The purpose of this study was to investigate the effect of exogenous nitric oxide (NO) on the proliferation of gastric carcinoma cells and the signaling pathways that regulate these responses. METHODS: MKN-28 cells were obtained from the Korean Cell Line Bank (KCLB) and maintained in DMEM culture media. The effect of sodium nitroprusside (SNP), a NO donor, on the proliferation of a serum-starved gastric carcinoma cell line, MKN-28, was examined by [3H]thymidine incorporation. Western blot was performed to analyze the translocation of protein kinase C (PKC)-deltafrom the cytosol to the plasma membrane of the MKN-28 cells. RESULTS: The proliferation of MKN-28 cells was significantly increased by SNP. It was also found that the proliferation was significantly inhibited by the protein kinase A (PKA) inhibitor, KT5720, and the protein kinase G inhibitor (PKG), KT5823, in SNP-treated cells. The SNP-induced proliferation was also inhibited by the PKC-deltaspecific inhibitor, rottlerin (1mu), but was increased by the PKC-beta inhibitor, Go6976 (1muM). The amount of translocated PKC-deltaprotein in the plasma membrane from the cytosol increased time-dependently after treating the cells with SNP, suggesting that NO activates PKC-delta Anti-inflammatory drugs, including dexamethasone, aspirin, indomethacin, mephenamic acid, and acetaminophen inhibited the SNP-induced proliferation of the cells and blocked of PKC-deltaactivation. CONCLUSION: NO stimulates the proliferation of serum- starved gastric cancer cells. The NO-induced proliferation may be mediated by PKC-delta The inhibitory effect of anti-inflammatory drugs on cell proliferation may be related to the inhibition of PKC-deltaactivity.


Subject(s)
Humans , Acetaminophen , Aspirin , Blotting, Western , Cell Line , Cell Membrane , Cell Proliferation , Culture Media , Cyclic AMP-Dependent Protein Kinases , Cyclic GMP-Dependent Protein Kinases , Cytosol , Dexamethasone , Indomethacin , Nitric Oxide , Nitroprusside , Protein Kinase C , Protein Kinase C-delta , Protein Kinases , Stomach Neoplasms , Tissue Donors
SELECTION OF CITATIONS
SEARCH DETAIL