Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 395
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Inflammopharmacology ; 26(3): 805-815, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29353412

RESUMO

Spleen tyrosine kinase (Syk) has emerged recently as a major effector of proinflammatory genes expression induced by LPS-elicited TLR4 activation, and manifested by the up-amplification in the production of inflammatory mediators, including PGE2 and NO. Here, we investigated the nature of factors involved in the recruitment and interaction of Syk with TLR4 in gastric mucosa in response to H. pylori LPS. We show that stimulation of gastric mucosal cells with the LPS leads to localization of Syk with the membrane-associated TLR4 complex and its activation through phosphorylation on Tyr. Furthermore, we reveal that the membrane translocation of Syk upon the LPS stimulation occurs with the involvement of protein kinase Cδ (PKCδ)-mediated phosphorylation of Syk on Ser. Thus, our findings demonstrate that H. pylori LPS-induced up-regulation in Syk activity proceeds through the stage of PKCδ-mediated Syk phosphorylation on Ser, required for its recruitment to the membrane-anchored TLR4, followed by the kinase activation through phosphorylation on Tyr. Hence, the phase of PKCδ-mediated Syk phosphorylation on Ser affects the extent of amplification in gastric mucosal inflammatory response to H. pylori.


Assuntos
Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Lipopolissacarídeos/toxicidade , Proteína Quinase C-delta/metabolismo , Quinase Syk/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Humanos , Ratos
2.
Inflammopharmacology ; 25(4): 415-429, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28516374

RESUMO

Infection with Helicobacter pylori is a primary culprit in the etiology of gastric disease, and its cell-wall lipopolysaccharide (LPS) is recognized as a potent endotoxin responsible for triggering a pattern of the mucosal inflammatory responses. The engagement by the LPS of gastric mucosal Toll-like receptor 4 (TLR4) leads to initiation of signal transduction events characterized by the activation of mitogen-activated protein kinase (MAPK) cascade, induction of phosphoinositide-specific phospholipase C (PLC)/protein kinase C (PKC)/phosphatidylinositol 3-kinase (PI3K) pathway, and up-regulation in Src/Akt. These signaling events in turn exert their influence over H. pylori-elicited excessive generation of NO and PGE2 caused by the disturbances in nitric oxide synthase and cyclooxygenase isozyme systems, increase in epidermal growth factor receptor transactivation, and the induction in matrix metalloproteinase-9 (MMP-9) release. Interestingly, the extent of gastric mucosal inflammatory response to H. pylori is influenced by a peptide hormone, ghrelin, the action of which relays on the growth hormone secretagogue receptor type 1a (GHS-R1a)-mediated mobilization of G-protein dependent transduction pathways. Yet, the signals triggered by TLR-4 activation as well as those arising through GHS-R1a stimulation converge at MAPK and PLC/PKC/PI3K pathways that form a key integration node for proinflammatory signals generated by H. pylori LPS as well as for those involved in modulation of inflammation by ghrelin. Hence, therapeutic targeting these signals' convergence and integration node could provide a novel and attractive opportunities for developing more effective treatments of H. pylori-related gastric disease.


Assuntos
Mucosa Gástrica/efeitos dos fármacos , Grelina/uso terapêutico , Helicobacter pylori/efeitos dos fármacos , Mediadores da Inflamação/antagonistas & inibidores , Lipopolissacarídeos/toxicidade , Animais , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Grelina/metabolismo , Grelina/farmacologia , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/metabolismo , Helicobacter pylori/isolamento & purificação , Humanos , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo
3.
Inflammopharmacology ; 24(5): 197-205, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27613723

RESUMO

Regulation of matrix metalloproteinase-9 (MMP-9) secretion in response to proinflammatory challenge remains under a strict control of factors that affect the stability dynamics of the major cytoskeleton polymeric structures, microtubules (MTs). In this study, we report that H. pylori LPS-elicited induction gastric mucosal MMP-9 secretion is accompanied by the enhancement in MT stabilization as evidenced by the increase in α-tubulin acetylation and detyrosination while the modulatory influence of hormone, ghrelin, is associated with MT destabilization and reflected in a decrease in α-tubulin acetylation and detyrosination. Further, we reveal that the LPS-induced enhancement in MT stabilization and up-regulation in MMP-9 secretion as well as the modulatory influence of ghrelin occur with the involvement of PKCδ and SFK. The LPS effect is reflected in a marked increase in PKCδ-mediated α-tubulin phosphorylation on Ser, while the modulatory effect of ghrelin on MT dynamics and MMP-9 secretion is manifested by the SFK-dependent phosphorylation of α-tubulin on Tyr. Moreover, the changes in α-tubulin phosphorylation and MT stabilization dynamics occur in concert with the Golgi recruitment and activation of PKD2 and Arf-GEF. The findings demonstrate that the enhancement in gastric mucosal MMP-9 secretion in response to H. pylori and its modulation by ghrelin are the result of changes in MT dynamics conferred by PKCδ/SFK- mediated α-tubulin Ser/Tyr phosphorylation.


Assuntos
Mucosa Gástrica/metabolismo , Grelina/farmacologia , Helicobacter pylori , Metaloproteinase 9 da Matriz/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Animais , Mucosa Gástrica/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Ratos , Serina/metabolismo , Tirosina/metabolismo
4.
Inflammopharmacology ; 24(2-3): 119-26, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27209313

RESUMO

Matrix metalloproteinas-9 (MMP-9) is a glycosylated endopeptidase associated with host reaction to microbial endotoxins and also characterizes gastric mucosal inflammatory response to H. pylori infection. Here, we report on the factors involved in gastric mucosal MMP-9 secretion in response to H. pylori LPS, and the effect of hormone, ghrelin. We show that both the LPS-elicited induction in MMP-9 secretion and also the modulatory influence of ghrelin occur at the level of MMP-9 processing between the endoplasmic reticulum (ER) and Golgi. Further, we demonstrate that the LPS effect is associated with up-regulation in the activation of Arf1, a small GTPase of the ADP-ribosylation factor family, and the recruitment and phosphorylation of protein kinase D2 (PKD2), involved in the secretory cargo processing in the Golgi. Moreover, we reveal that the LPS-induced up-regulation in MMP-9 secretion is reflected in a marked increase in PKCδ-mediated PKD2 phosphorylation on Ser, while the modulatory effect of ghrelin is manifested by the SFK-PTKs-dependent phosphorylation of PKD2 on Tyr. Thus, our findings demonstrate the role of Arf1/PKD2 in mediation of H. pylori LPS-induced up-regulation in gastric mucosal MMP-9 secretion and suggest the modulatory mechanism of ghrelin action.


Assuntos
Mucosa Gástrica/metabolismo , Grelina/farmacologia , Helicobacter pylori , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Quinases/metabolismo , Tirosina/metabolismo , Animais , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/microbiologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteína Quinase D2 , Ratos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
5.
Inflammopharmacology ; 24(2-3): 87-95, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26886372

RESUMO

Matrix metalloproteinases (MMPs) are a family of endopeptidases implicated in a wide rage of degenerative and inflammatory diseases, including Helicobacter pylori-associated gastritis, and gastric and duodenal ulcer. As gastric mucosal inflammatory responses to H. pylori are characterized by the rise in MMP-9 production, as well as the induction in mitogen-activated protein kinase (MAPK) and Rac1 activation, we investigated the role of Rac1/MAPK in the processes associated with the release of MMP-9. We show that H. pylori LPS-elicited induction in gastric mucosal MMP-9 release is associated with MAPK, ERK and p38 activation, and occurs with the involvement of Rac1 and cytosolic phospholipase A2 (cPLA2). Further, we demonstrate that the LPS-induced MMP-9 release requires ERK-mediated phosphorylation of cPLA2 on Ser(505) that is essential for its membrane localization with Rac1, and that this process necessitates p38 participation. Moreover, we reveal that the activation and membrane translocation of p38 to the Rac1-GTP complex plays a pivotal role in cPLA2-dependent enhancement in MMP-9 release. Hence, our findings provide a strong evidence for the role of ERK/cPLA2 and Rac1/p38/cPLA2 cascade in H. pylori LPS-induced up-regulation in gastric mucosal MMP-9 release.


Assuntos
Mucosa Gástrica/metabolismo , Fosfolipases A2 do Grupo IV/metabolismo , Helicobacter pylori , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/microbiologia , Fosfolipases A2 do Grupo IV/antagonistas & inibidores , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores
6.
Inflammopharmacology ; 24(1): 23-31, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26658844

RESUMO

Infection of gastric mucosa by H. pylori triggers a pattern of inflammatory responses characterized by the rise in proinflammatory cytokine production, up-regulation in mitogen-activated protein kinase (MAPK) cascade, and the induction in epidermal growth factor receptor (EGFR) activation. In this study, we report on the role of MAPK/p38 and Rac1 in the regulation of H. pylori LPS-induced TGF-α ectodomain shedding and EGFR transactivation. We show that stimulation of gastric mucosal cells with the LPS, reflected in p38 phosphorylation, guanine nucleotide exchange factor Dock180 activation and the rise in Rac1-GTP level, is accompanied by the activation of membrane-associated metalloprotease, (TACE) also known as ADAM17, responsible for soluble TGF-α release. Further, we reveal that the LPS-induced TGF-α shedding and EGFR transactivation involves the TACE activation through phosphorylation by p38 that requires Rac1 participation. Moreover, we demonstrate that up-regulation in H. pylori LPS-elicited Rac1-GTP membrane translocation plays a pivotal role in recruitment of the activated p38 to the membrane for TACE activation through phosphorylation on Thr(735). Taken together, our findings provide strong evidence as to the essential function of Rac1 in TACE activation, TGF-α ectodomain shedding, and the EGFR transactivation.


Assuntos
Mucosa Gástrica/metabolismo , Helicobacter pylori/metabolismo , Lipopolissacarídeos/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animais , Receptores ErbB/metabolismo , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Ratos , Ativação Transcricional , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
7.
Inflammopharmacology ; 23(2-3): 111-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25957600

RESUMO

A small GTPase, Rac1, is recognized as an important modulator of the inflammatory responses to bacterial lipopolysaccharide (LPS) by affecting the processes of phospholipase C activation. The activation of Rac1 involves the exchange of GDP for GTP and is catalyzed by the guanine nucleotide exchange factors (GEFs). Here, we report on the gastric mucosal GEF, Dock180, activation in response to H. pylori PS, and the hormone, ghrelin. We show that stimulation of gastric mucosal cells with the LPS leads to up-regulation in Dock180 phosphorylation on Tyr and Ser that is accompanied by a massive rise in Rac1-GTP level, while the effect of ghrelin, manifested by a drop in Dock180 phosphorylation on Ser, is associated with a decrease in Rac1-GTP formation. Furthermore, we demonstrate that phosphorylation on Tyr remains under the control of the Src family protein tyrosine kinases (SFK-PTKs), and is accompanied by Dock180 membrane translocation, while phosphorylation of the membrane-localized Dock180 on Ser represents the stimulatory contribution of protein kinase Cδ (PKCδ) to Dock180 activation. Moreover, we reveal that the interaction between Dock180 and PKCδ is dependent on Dock180 Tyr phosphorylation as well as the activity of PKCδ. Thus, our findings point to the involvement of PKCδ in the LPS-induced up-regulation of Dock180 activation, and suggest the modulatory mechanism of ghrelin influence on the gastric mucosal inflammatory responses to H. pylori.


Assuntos
Mucosa Gástrica/metabolismo , Grelina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/microbiologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C-delta/metabolismo , Ratos , Serina/metabolismo , Tirosina/metabolismo , Regulação para Cima/efeitos dos fármacos , Quinases da Família src/metabolismo
8.
Inflammopharmacology ; 23(2-3): 101-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25796615

RESUMO

Membrane recruitment followed by targeted phosphorylation of specific Tyr and Ser residues and the interaction with Rac GTPases are the crucial parts of an elaborate mechanism of PLCγ2 activation essential for its role in linking the specific receptor responses to a variety of hormones and bacterial endotoxins with the intended intracellular targets. Here, we explored the involvement of Rac in mediation of PLCγ2 activation associated with gastric mucosal inflammatory responses to H. pylori LPS and the hormone, ghrelin. We show that stimulation of gastric mucosal cells with the LPS leads to the membrane translocation of Rac1 as well as PLCγ2, while the effect of ghrelin is manifested by elevation in the membrane PLCγ2 activation and suppression in Rac1 translocation. However, blocking the LPS-induced Rac1 translocation, while detrimental to the PLCγ2 activation, has no effect on its membrane translocation. We reveal further that PLCγ2, localized in the membrane in association with Rac1 following the LPS stimulation, exhibits a marked increase in phosphorylation on Ser, while the modulatory effect of ghrelin, manifested by a drop in Rac1 translocation, is associated with a distinct decrease in PLCγ2 phosphorylation on Ser. Thus, the results suggest that H. pylori-elicited increase in gastric mucosal PLCγ2 phosphorylation on Ser serves as an essential platform for Rac1 colocalization and amplification in PLCγ2 activation.


Assuntos
Mucosa Gástrica/metabolismo , Grelina/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Fosfolipase C gama/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/microbiologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Fosforilação/efeitos dos fármacos , Ratos
9.
Inflammopharmacology ; 23(1): 37-45, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25362585

RESUMO

Phosphoinositide-specific phospholipase C (PLC) enzymes are crucial elements of signal transduction pathways that provide a common link of communication integrating specific receptor responses to a variety of hormones, growth factors, and bacterial endotoxins with the intended intracellular targets. Here, we examined the involvement of PLC in modulation of gastric mucosal inflammatory responses to Helicobacter pylori LPS by peptide hormone, ghrelin. We show that stimulation of gastric mucosal cells with the LPS leads to the activation and membrane translocation of the γ2 isoform of PLC, phosphorylated on Tyr as well as Ser, while the effect of ghrelin is reflected in the translocation and phosphorylation of membrane-associated PLCγ2 on Tyr mainly. Moreover, we demonstrate that PLCγ2 phosphorylation on Tyr remains under the control of the Src family protein tyrosine kinases (SFK-PTKs), and is intimately linked to PLCγ2 membrane localization, while the LPS-induced phosphorylation of membrane-recruited PLCγ2 on Ser displays dependence on protein kinase Cδ (PKCδ) and leads to the amplification in PLCγ2 activation. Thus, our findings link the extent of H. pylori-elicited gastric mucosal inflammatory involvement to the PKCδ-mediated amplification in PLCγ2 activation through phosphorylation on Ser.


Assuntos
Mucosa Gástrica/enzimologia , Grelina/farmacologia , Helicobacter pylori/enzimologia , Fosfolipase C gama/metabolismo , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Mucosa Gástrica/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Ratos
10.
Inflammopharmacology ; 22(4): 251-62, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24840386

RESUMO

A peptide hormone, ghrelin, plays a key role in modulation of gastric mucosal inflammatory responses to Helicobacter pylori by controlling the activation of constitutive nitric oxide synthase via Src/Akt-dependent phosphorylation that requires phosphatidylinositol 3-kinase (PI3K) participation. Here, we examined the relationship among PI3K; its upstream effector, protein kinase C (PKC); and cSrc. We show that stimulation of gastric mucosal cells with H. pylori LPS leads to the activation and membrane translocation of Ser-phosphorylated PKCδ, while the effect of ghrelin is reflected in the phosphorylation of membrane-associated PKCδ on Tyr. Further, we demonstrate that in response to the LPS-induced PKCδ activation both PI3K and Src show a marked increase in their Ser phosphorylation, while the effect of ghrelin is manifested in the phosphorylation of PI3K and cSrc at Tyr. Moreover, whereas Tyr phosphorylation of PKCδ exhibited susceptibility to cSrc inhibitor (PP2), the inhibitor of PKC (GF109203X) but not that of cSrc (PP2) blocked the Tyr phosphorylation of PI3K, while ghrelin-induced cSrc phosphorylation at Tyr was subject to inhibition by the inhibitors of PKC and PI3K. Thus, our findings stipulate the prerequisite of PKCδ in the activation of PI3K as well as cSrc, and imply that PI3K activation provides an essential platform for ghrelin-induced cSrc activation through autophosphorylation at Tyr(416). We also reveal that ghrelin-elicited up-regulation in PKCδ activation by Tyr phosphorylation shows dependence on cSrc activity.


Assuntos
Mucosa Gástrica/patologia , Grelina/metabolismo , Infecções por Helicobacter/patologia , Proteína Quinase C-delta/metabolismo , Animais , Mucosa Gástrica/microbiologia , Helicobacter pylori/isolamento & purificação , Indóis/farmacologia , Inflamação/microbiologia , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Maleimidas/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Ratos , Regulação para Cima
11.
Inflammopharmacology ; 22(3): 169-77, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24057979

RESUMO

A peptide hormone, ghrelin, is recognized as an important modulator of gastric mucosal inflammatory responses to Helicobacter pylori through the regulation of Src/Akt-dependent activation of constitutive nitric oxide synthase (cNOS) by phosphorylation. In this study, we report on the role of phosphatidylinositol 3-kinase (PI3K) in the processes of Src/Akt activation in gastric mucosal cells exposed to H. pylori LPS. We demonstrate that cNOS activation through phosphorylation induced by ghrelin is associated with PI3K activation which occurs upstream of cSrc, and that PI3K is required for cSrc activation of Akt. We show further that ghrelin-induced activation of PI3K, as well as that of Src and Akt, was susceptible to suppression by the inhibitors of phospholipase C (U73122) and protein kinase C (BIM). Both these inhibitors also blocked the ghrelin-induced membrane translocation of PI3K and cSrc, whereas the inhibitor of PI3K (LY294002) blocked only the membrane translocation of cSrc. Collectively, our findings suggest that the modulatory influence of ghrelin in countering gastric mucosal responses to H. pylori LPS relies on PI3K activation that depends on PLC/PKC signaling pathway, and that PI3K activity is required for the induction of cSrc/Akt activation.


Assuntos
Mucosa Gástrica/enzimologia , Grelina/farmacologia , Helicobacter pylori/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Animais , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Helicobacter pylori/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/enzimologia , Ratos
12.
Inflammopharmacology ; 21(3): 241-51, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23563696

RESUMO

Among the key factors defining the extent of gastric mucosal inflammatory involvement in response to Helicobacter pylori is the excessive generation of prostaglandin (PGE2) and nitric oxide (NO), caused by the overexpression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS), and triggered by the activation of mitogen-activated protein kinase (MAPK)/c-Jun N-terminal kinase, p38 and ERK, and nuclear translocation of the cognate transcription factors. In this study, we report on the role of MAPK/ERK in the regulation of H. pylori LPS-induced gastric mucosal expression of COX-2 and iNOS. We show that ERK activation by the LPS leads to phosphorylation of the inhibitory κB kinase-ß (IKK-ß) and cytosolic phospholipase A2 (cPLA2), and is reflected in the upsurge in NF-κB nuclear translocation, induction in COX-2 and iNOS expression, and up-regulation in cPLA2 activity. The modulatory effect of peptide hormone, ghrelin, on the LPS-induced changes, although associated with further enhancement in ERK, IKK-ß, and cPLA2 phosphorylation, was reflected in the suppression of IKK-ß and cPLA2 activity through S-nitrosylation. While the effect of ghrelin on S-nitrosylation was susceptible to suppression by the inhibitors of Src/Akt pathway, the inhibition of ERK activation caused the blockage in IKK-ß and cPLA2 phosphorylation as well as S-nitrosylation. Taken together, our data show that H. pylori-induced ERK activation plays a critical role in up-regulation of gastric mucosal PGE2 and NO generation at the level of IKK-ß and cPLA2 activation, and that ghrelin counters these proinflammatory consequences of the LPS through Src/Akt-dependent S-nitrosylation.


Assuntos
Mucosa Gástrica/patologia , Grelina/metabolismo , Infecções por Helicobacter/fisiopatologia , Inflamação/patologia , Animais , Dinoprostona/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/isolamento & purificação , Quinase I-kappa B/metabolismo , Inflamação/microbiologia , Lipopolissacarídeos/toxicidade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óxido Nítrico/metabolismo , Fosfolipases A2 Citosólicas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Regulação para Cima , Quinases da Família src/metabolismo
13.
Inflammopharmacology ; 21(1): 67-78, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22669511

RESUMO

A peptide hormone, ghrelin, plays an important role in modulation of gastric mucosal inflammatory responses to Helicobacter pylori infection by controlling the cross-talk between nitric oxide synthase (NOS) and cyclooxygenase (COX) enzyme systems. In this study, we report that H. pylori LPS-elicited induction in gastric mucosal COX-2 and inducible (i) iNOS protein expression, and the impairment in constitutive (c) cNOS phosphorylation, was associated with mitogen-activated protein kinase, c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase and p38 activation, and occurred with the involvement of transcription factors, CCATT/enhancer-binding protein (C/EBP) δ, cAMP response element-binding protein, activator protein-1 (AP-1), and NF-κB. The modulatory effect of ghrelin on the LPS-induced changes was manifested in the inhibition of nuclear translocation of p65 NF-κB and C/EBPδ, and suppression in AP-1 activation, and the inhibition in phosphorylation of JNK and p38, as well as their respective downstream targets, c-Jun and ATF-2. However, only the inhibition of p38-mediated ATF-2 phosphorylation was reflected in the reduced expression of COX-2 protein. Further, the effect of ghrelin of the LPS-induced changes was reflected in the increase in Src/Akt-dependent cNOS activation through phosphorylation and the inhibition of cNOS-mediated IKK-ß S-nitrosylation. Our findings indicate ghrelin counters the proinflammatory consequences of H. pylori by interfering with the p38/ATF-2-induced AP-1 activation in association with concurrent up-regulation in Src/Akt-dependent cNOS phosphorylation.


Assuntos
Grelina/metabolismo , Inflamação/patologia , Fator de Transcrição AP-1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Regulação da Expressão Gênica , Infecções por Helicobacter/fisiopatologia , Helicobacter pylori/isolamento & purificação , Inflamação/microbiologia , Lipopolissacarídeos , NF-kappa B/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação , Ratos , Regulação para Cima
14.
Inflammopharmacology ; 19(4): 197-204, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21516493

RESUMO

A peptide hormone, ghrelin, is recognized as an important modulator of gastric mucosal inflammatory responses to H. pylori through the regulation of nitric oxide synthase (NOS) system. As cSrc kinase plays a major role in transduction of signals that regulate the activity of NOS isozyme system, we investigated the influence of H. pylori LPS on the processes associated with Src activation in gastric mucosal cells. The LPS-induced drop in constitutive (c) cNOS activity and up-regulation in inducible (i) iNOS was associated with the suppression in cSrc kinase activity that was reflected in a decrease in its phosphorylation at Tyr4¹6. Further, the countering effect of ghrelin on the LPS-induced changes in cSrc activity and the extent of its phosphorylation was accompanied by a marked reduction in the activity of iNOS and an increase in cNOS activation through phosphorylation at Ser¹¹79. Moreover, the effect of ghrelin on cSrc activation and its Tyr4¹6 phosphorylation was associated with the kinase S-nitrosylation that was susceptible to the blockage by cNOS inhibition. Our findings suggest that up-regulation in iNOS with H. pylori infection leads to disturbances in cNOS phosphorylation that exerts the detrimental effect on the processes of cSrc activation through cNOS-mediated S-nitrosylation. We also show that ghrelin attenuation of H. pylori-induced gastric mucosal inflammatory responses involves the enhancement in cSrc activation, elicited by the kinase S-nitrosylation and the increase in its phosphorylation at Tyr4¹6.


Assuntos
Mucosa Gástrica/metabolismo , Grelina/metabolismo , Helicobacter pylori/imunologia , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Animais , Anticorpos Fosfo-Específicos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos , Mucosa Gástrica/citologia , Mucosa Gástrica/imunologia , Infecções por Helicobacter/imunologia , Imunidade nas Mucosas , Lipopolissacarídeos/toxicidade , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/química , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Nitrosação/efeitos dos fármacos , Concentração Osmolar , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas pp60(c-src)/química , Ratos , Regulação para Cima/efeitos dos fármacos
15.
Inflammopharmacology ; 19(2): 89-97, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21279549

RESUMO

Loss of mucus coat integrity and the impairment in its mucin component as well as the disturbance in nitric oxide (NO) generation are well-recognized features of gastric disease associated with H. pylori infection. As ghrelin plays a major role in the regulation of nitric oxide synthase system, we investigated the influence of this hormone on H. pylori LPS-induced interference with gastric mucin synthesis. The results revealed that the LPS-induced impairment in mucin synthesis and accompanied induction in inducible nitric oxide synthase (iNOS) expression, were associated with the suppression in Akt kinase activity and the impairment in constitutive nitric oxide synthase (cNOS) phosphorylation. The LPS effect on Akt inactivation was manifested in the kinase protein S-nitrosylation and a decrease in its phosphorylation at Ser(473). Further, we show that the countering effect of ghrelin, on the LPS-induced impairment in mucin synthesis was reflected in the suppression of iNOS and the increase in Akt activation, associated with the loss in S-nitrosylation and the increase in phosphorylation, as well as cNOS activation through phosphorylation. Our findings demonstrate that up-regulation in iNOS with H. pylori infection and subsequent Akt kinase inactivation through S-nitrosylation exerts the detrimental effect on the processes dependent on Akt activation, including that of cNOS activation and mucin synthesis. We also show that ghrelin protection against H. pylori-induced impairment in mucin synthesis is intimately linked to the events of Akt activation and reflected in a decrease in the kinase S-nitrosylation and the increase in its phosphorylation.


Assuntos
Cisteína/análogos & derivados , Mucinas Gástricas/biossíntese , Grelina/farmacologia , Lipopolissacarídeos/farmacologia , Processamento de Proteína Pós-Traducional/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , S-Nitrosotióis/metabolismo , Animais , Ácido Ascórbico/farmacologia , Células Cultivadas , Cisteína/metabolismo , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Mucosa Gástrica/citologia , NF-kappa B/antagonistas & inibidores , Naftoquinonas/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Ratos , Ratos Endogâmicos , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
16.
Inflammopharmacology ; 18(5): 233-40, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20596895

RESUMO

Infection with H. pylori is a primary factor in the etiology of gastric disease, and the excessive NO generation and a massive rise in apoptosis are well recognized features that characterize the mucosal inflammatory responses to the bacterium and its lipopolysaccharide (LPS). Here, we report that H. pylori LPS-induced enhancement in gastric mucosal cell apoptosis and NO generation was associated with the suppression in constitutive nitric oxide synthase (cNOS) activity and a marked up-regulation in the activity of inducible nitric oxide synthase (iNOS). Further, we demonstrate that the detrimental effect of the LPS on cNOS was manifested in the enzyme protein S-nitrosylation, that was susceptible to suppression by iNOS inhibitor, 1400W. Moreover, we show that the countering effect of peptide hormone, ghrelin, on the LPS-induced changes in apoptosis and cNOS activity was reflected in the loss in cNOS S-nitrosylation and the increase in the enzyme phosphorylation. These findings demonstrate that the disturbances in gastric mucosal NO generation system caused by H. pylori result from the iNOS-derived NO suppression of cNOS activation through S-nitrosylation. We also report that ghrelin protection against H. pylori-induced gastric mucosal proapoptotic events involves cNOS activation manifested by the increase in enzyme protein phosphorylation and a decrease in its S-nitrosylation.


Assuntos
Apoptose/efeitos dos fármacos , Mucosa Gástrica/citologia , Grelina/metabolismo , Helicobacter pylori/fisiologia , Óxido Nítrico Sintase/metabolismo , Amidinas/farmacologia , Animais , Benzilaminas/farmacologia , Células Cultivadas , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Grelina/farmacologia , Lipopolissacarídeos/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação , Ratos
17.
Inflammopharmacology ; 18(3): 119-25, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20155328

RESUMO

Recent advances in identifying the salivary constituents capable of influencing the oral mucosal inflammatory responses have brought to focus the importance of a peptide hormone, ghrelin. Here, we report on the involvement of ghrelin in controlling the apoptotic processes induced in sublingual salivary gland acinar cells by the lipopolysaccharide (LPS) of a periodontopathic bacterium, Porphyromonas gingivalis. We show that the countering effect of ghrelin on the LPS-induced acinar cell apoptosis was associated with the increase in constitutive nitric oxide synthase (cNOS) activity, and the reduction in caspase-3 and inducible nitric oxide synthase (iNOS). The loss in countering effect of ghrelin on the LPS-induced changes in apoptosis and caspase-3 activity was attained with Src kinase inhibitor, PP2, as well as Akt inhibitor, SH-5, and cNOS inhibitor, L-NAME, but not the iNOS inhibitor, 1400W. The effect of ghrelin on the LPS-induced changes in cNOS activity, moreover, was reflected in the increased cNOS phosphorylation that was sensitive to PP2 as well as SH-5. Furthermore, the ghrelin-induced up-regulation in cNOS activity was associated with the increase in caspase-3 S-nitrosylation that was susceptible to the blockage by SH-5 and L-NAME. The findings point to the involvement of ghrelin in Src/Akt kinase-mediated cNOS activation and the apoptogenic signal inhibition through the NO-induced caspase-3 S-nitrosylation.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Cisteína/análogos & derivados , Grelina/farmacologia , Lipopolissacarídeos/farmacologia , Óxido Nítrico Sintase/metabolismo , S-Nitrosotióis/metabolismo , Glândula Sublingual/citologia , Amidinas/farmacologia , Animais , Ácido Ascórbico/farmacologia , Benzilaminas/farmacologia , Células Cultivadas , Cisteína/metabolismo , Inibidores Enzimáticos/farmacologia , Fosfatos de Inositol/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação/efeitos dos fármacos , Porphyromonas gingivalis/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinas/farmacologia , Ratos , Ratos Endogâmicos , Quinases da Família src/antagonistas & inibidores
18.
Inflammopharmacology ; 17(5): 245-53, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19757089

RESUMO

Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor, is an important regulator of nitric oxide synthase (NOS) and cyclooxygenase (COX) enzyme systems, the products of which are of major significance to the processes of gastric mucosal defense and repair. Here, using primary culture of rat gastric mucosal cells, we report on the mechanism of ghrelin protection against ethanol cytotoxicity. We show that the protective effect of ghrelin was associated with the increase in NO and PGE2 production, and characterized by a marked up-regulation in cytosolic phospholipase A(2) (cPLA(2)) activity and arachidonic acid (AA) release. The loss in countering effect of ghrelin on the ethanol cytotoxicity was attained with constitutive NOS (cNOS) inhibitor, L-NAME, as well as indomethacin and a specific COX-1 inhibitor, SC-560, while specific COX-2 inhibitor, NS-398, and a selective inducible NOS (iNOS) inhibitor, 1400W, had no effect. The effect of L-NAME was reflected in the inhibition of ghrelin-induced mucosal cell capacity for NO production, cPLA(2) activation, and PGE2 generation, whereas indomethacin caused only the inhibition in PGE2 generation. Moreover, the ghrelin-induced up-regulation in AA release was reflected in the cPLA(2) enzyme protein phosphorylation and S-nitrosylation. Preincubation with L-NAME resulted in the inhibition of the ghrelin-induced S-nitrosylation, whereas the ERK inhibitor, PD98059, caused the blockage in cPLA(2) protein phosphorylation as well as S-nitrosylation. The findings demonstrate that ghrelin protection of gastric mucosa against ethanol cytotoxicity involves cNOS-derived NO induction of cPLA(2) activation for the increase in PGE2 synthesis. This activation process apparently includes the cPLA(2) phosphorylation followed by S-nitrosylation.


Assuntos
Etanol/toxicidade , Mucosa Gástrica/efeitos dos fármacos , Grelina/metabolismo , Óxido Nítrico Sintase/metabolismo , Animais , Ácido Araquidônico/metabolismo , Depressores do Sistema Nervoso Central/toxicidade , Dinoprostona/biossíntese , Mucosa Gástrica/metabolismo , Óxido Nítrico/metabolismo , Fosfolipases A2 Citosólicas/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Receptores de Grelina/metabolismo , Regulação para Cima
19.
J Physiol Pharmacol ; 60(2): 49-55, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19617645

RESUMO

A pleiotropic hormone, leptin, secreted into saliva by the acinar cells of salivary glands is an important mediator of the processes of oral mucosal defense. Here, we report on the role of epidermal growth factor receptor (EGFR) transactivation in the signaling events that mediate leptin protection of sublingual salivary gland acinar cells against ethanol cytotoxicity. We show that the protective effect of leptin against ethanol cytotoxicity was associated with the increased EGFR protein tyrosine kinase and cytosolic phospholipase A(2) (cPLA(2)) activity, and characterized by a marked increase in matrix metalloproteinase MMP-9 and arachidonic acid (AA) release, and PGE(2) generation. The loss in countering capacity of leptin against ethanol cytotoxicity was attained with JAK inhibitor AG490, Src inhibitor PP2, and EGFR inhibitor AG1478, as well as ERK inhibitor PD98059. Moreover, the agents evoked also the inhibition in leptin-induced up-regulation in cPLA(2) activity, AA release, and PGE(2) generation. The changes caused by leptin in EGFR phosphorylation, MMP-9, and cPLA(2) activation were susceptible to suppression by metalloprotease inhibitor GM6001, but the production of MMP-9 was not affected by EGFR inhibitor AG1478 or PKC inhibitor Ro318220. These findings point to the involvement of MMP-9 in the event of leptin-induced EGFR transactivation that results in the signaling cascade leading to cPLA(2) activation and up-regulation in PGE(2) generation, thus providing new insights into the mechanism of oral mucosal protection against ethanol toxicity.


Assuntos
Receptores ErbB/metabolismo , Etanol/farmacologia , Leptina/fisiologia , Fosfolipases A2 Citosólicas/metabolismo , Glândulas Salivares/metabolismo , Ativação Transcricional , Animais , Ácido Araquidônico/metabolismo , Células Cultivadas , Dinoprostona/metabolismo , Ativação Enzimática , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Técnicas In Vitro , Janus Quinases/antagonistas & inibidores , Leptina/farmacologia , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Ratos , Glândulas Salivares/citologia , Glândulas Salivares/efeitos dos fármacos , Transdução de Sinais , Regulação para Cima , Quinases da Família src/antagonistas & inibidores
20.
Inflammopharmacology ; 17(1): 6-14, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19127349

RESUMO

A pluripotent cytokine, leptin, released locally within the mucosal tissue is an important mediator of the processes of gastric mucosal defense and repair. Here, we report that leptin protection of gastric mucosal cells against ethanol cytotoxicity requires epidermal growth factor receptor (EGFR) participation. We show that the protective effect of leptin against ethanol cytotoxicity was associated with the increased EGFR and cPLA(2) phosphorylation, and characterized by a marked increase in arachidonic acid (AA) release and prostaglandin (PGE(2)) generation. The loss in countering capacity of leptin on the ethanol-induced cytotoxicity was attained with Src kinase inhibitor, PP2, and EGFR kinase inhibitor, AG1478, as well as ERK inhibitor, PD98059. Moreover, all three agents evoked also the inhibition in leptin-induced upregulation in cPLA(2) activity, AA release, and PGE(2) generation. Furthermore, changes caused by leptin in EGFR phosphorylation and cPLA(2) activation were susceptible to suppression by GM6001, a metalloprotease inhibitor of membrane-anchored EGFR ligand cleavage. These findings disclose an important link between leptin-induced and Src kinase-mediated EGFR transactivation and the activation of cytosolic phospholipase A(2) that leads to up-regulation in PGE2 production, thus providing new insights into the mechanism of gastric mucosal protection by leptin.


Assuntos
Receptores ErbB/efeitos dos fármacos , Mucosa Gástrica/efeitos dos fármacos , Leptina/farmacologia , Fosfolipases A2 Citosólicas/efeitos dos fármacos , Animais , Ácido Araquidônico/metabolismo , Dinoprostona/metabolismo , Ativação Enzimática/efeitos dos fármacos , Receptores ErbB/metabolismo , Etanol/toxicidade , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Técnicas In Vitro , Fosfolipases A2 Citosólicas/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Regulação para Cima/efeitos dos fármacos , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA