Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomed Res Int ; 2014: 312847, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25162007

RESUMO

BACKGROUND: Glioma is the most malignant tumor of the central nervous system. Efforts on the development of new chemotherapy are mandatory. Andrographolide (AND), a diterpenoid lactone isolated from the Andrographis paniculata, has been shown to have antitumor activities in several types of cancer cells. Whether AND can exert its antitumor activity in glioblastoma cells remains unknown. This study examined the anticancer effects of AND, both in vitro and in vivo. METHODS: Cell apoptosis was assayed by flow cytometry and nuclear staining. The signaling pathway for AND was determined by western blotting. The effects of AND on tumor growth was evaluated in a mouse model. RESULTS AND CONCLUSION: In vitro, with application of specific inhibitors and siRNA, AND-induced apoptosis was proven through ROS-ERK-P53-caspase 7-PARP signaling pathway. In vivo, AND significantly retarded tumor growth and caused regression of well-formed tumors in vivo. Furthermore, AND did not induce apoptosis or activate ERK and p53 in primary cultured astrocyte cells, and it may serve as a potential therapeutic candidate for the treatment of glioma.


Assuntos
Caspase 7/biossíntese , Diterpenos/administração & dosagem , Glioma/tratamento farmacológico , Proteína Supressora de Tumor p53/biossíntese , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Glioma/genética , Glioma/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , RNA Interferente Pequeno/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
2.
Toxicology ; 314(1): 11-21, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-23973256

RESUMO

Lysophosphatidylcholine (LPC) is a potent pro-arrhythmic derivative of the membrane phosphotidylcholine, which is accumulated in heart tissues during cardiac ischemia. However, the cellular mechanism underlying LPC-induced cardiomyocyte damage remains to be elucidated. This study focuses on the effects of LPC on cardiomyocyte gap junction. At 30µM, LPC decreased the spontaneous contraction rates of cardiomyocytes, and caused arrhythmic contraction without affecting cell viability. Connexin43 (Cx43) was seen as large plaques at cell junctions in control cells, whereas upon LPC treatment, the intensity of Cx43 staining was decreased in a concentration-sensitive manner and Cx43 staining appeared as tiny dots at cell junctions with a corresponding increase in cytoplasmic punctate staining. This distributional change of Cx43 was accompanied by an impairment of the gap junction intercellular communication (GJIC). Further, LPC treatment induced protein kinase C (PKC) activation, and PKC-dependent Cx43 phosphorylation at serine (Ser) 368. Pre-treatment with a specific PKCɛ inhibitor, eV1-2, prevented the LPC-induced Cx43 phosphorylation at Ser368 and the loss of Cx43 from gap junctions, both of which may disturb GJIC functions. Furthermore, siRNA knockdown of PKCɛ in H9c2 cells prevented LPC-induced serine phosphorylation of Cx43, confirming the role of PKCɛ in Cx43 serine phosphorylation. Double labeling immunofluorescence showed that LPC increased the colocalization of Cx43 with ubiquitin, and pretreatment with MG132 effectively prevented LPC-induced gap junction disassembly. LPC increased the ubiquitination of Cx43, which was blocked by eV1-2 pretreatment, suggesting that LPC accelerated the intracellular degradation of Cx43 via the ubiquitin-proteasomal pathway. It can be concluded that LPC destroyed the structure and function of gap junctions via PKCɛ-mediated serine phosphorylation of Cx43. PKCɛ inhibitors might therefore be effective in prevention of LPC-related diseases.


Assuntos
Conexina 43/metabolismo , Lisofosfatidilcolinas/farmacologia , Miócitos Cardíacos/metabolismo , Proteína Quinase C-épsilon/metabolismo , Serina/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Corantes , Feminino , Imunofluorescência , Frequência Cardíaca/efeitos dos fármacos , Imunoprecipitação , Indóis , Masculino , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Sais de Tetrazólio , Tiazóis
3.
Toxicology ; 306: 176-84, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23466501

RESUMO

Higher serum levels of p-cresol in chronic kidney disease populations have been associated with increased cardiovascular mortality. However, studies on how p-cresol affects intercellular junctions between cardiomyocytes were limited. This study investigated the effect of p-cresol on adherens junction (AJ) of neonatal cultured cardiomyocytes and its underlying mechanism. A loss of N-cadherin and p120-catenin (p120ctn) immunostaining from cell-cell contact sites was noted by p-cresol treatment. In addition, p-cresol disrupted AJs by inducing formation of intercellular gaps. Our previous study has revealed that p-cresol increased intracellular calcium levels and activated protein kinase Cα (PKCα) by phosphorylation. The PKCα activation was involved in the p-cresol-mediated AJ disassembly, since pharmacological inhibition of PKCα abolished the above-mentioned p-cresol effect. This PKCα activation also led to the serine dephosphorylation of p120ctn and caused the dissociation of p120ctn from N-cadherin. This hypothesis was further confirmed in H9c2 cells by siRNA approach. SiRNA knockdown of PKCα prevented p-cresol-induced serine dephosphorylation of p120ctn and splitting of AJ. In conclusion, p-cresol caused PKCα-dependent AJ disassembly of cardiomyocytes, which might be related to asychronized contraction.


Assuntos
Junções Aderentes/efeitos dos fármacos , Cresóis/toxicidade , Miocárdio/citologia , Miócitos Cardíacos/efeitos dos fármacos , Junções Aderentes/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Caderinas/metabolismo , Carbazóis/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Feminino , Masculino , Microscopia de Fluorescência , Miocárdio/enzimologia , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteína Quinase C-alfa/metabolismo , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/farmacologia , Ratos
4.
Stem Cells Dev ; 22(9): 1408-18, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23252631

RESUMO

Peripheral arterial diseases, the major complication of diabetes, can result in lower limb amputation. Since endothelial progenitor cells (EPCs) are involved in neovascularization, the aim of this study was to examine whether EPCs isolated from Wharton's jelly (WJ-EPCs) of the umbilical cord, a rich source of mesenchymal stem cells, could reduce ischemia-induced hind limb injury in diabetic mice. We evaluated the effects of WJ-EPC transplantation on hind limb injury caused by femoral artery ligation in mice with streptozotocin (STZ)-induced diabetes. We found that the ischemic hind limb in mice with STZ-induced diabetes showed decreased blood flow and capillary density and increased cell apoptosis and that these effects were significantly inhibited by an injection of WJ-EPCs. In addition, hypoxia-inducible factor-1α (HIF-1α) and interleukin-8 (IL-8) were highly expressed in transplanted WJ-EPCs in the ischemic skeletal tissues and were present at high levels in hypoxia-treated cultured WJ-EPCs. Moreover, incubation of the NOR skeletal muscle cell line under hypoxic conditions in conditioned medium from EPCs cultured for 16 h under hypoxic conditions resulted in decreased expression of pro-apoptotic proteins and increased expression of anti-apoptotic proteins. The inhibition of HIF-1α or IL-8 expression by EPCs using HIF-1α siRNA or IL-8 siRNA, respectively, prevented this change in expression of apoptotic-related proteins. Wharton's jelly in the umbilical cord is a valuable source of EPCs, and transplantation of these EPCs represents an innovative therapeutic strategy for treating diabetic ischemic tissues. The HIF-1α/IL-8 signaling pathway plays a critical role in the protective effects of EPCs in the ischemic hind limb of diabetic mice.


Assuntos
Membro Posterior/irrigação sanguínea , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-8/metabolismo , Isquemia/terapia , Doença Arterial Periférica/terapia , Transplante de Células-Tronco , Animais , Apoptose , Hipóxia Celular , Movimento Celular , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Artéria Femoral/patologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Interleucina-8/genética , Masculino , Camundongos , Camundongos Endogâmicos ICR , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/patologia , Neovascularização Fisiológica , Fluxo Sanguíneo Regional , Transdução de Sinais , Células-Tronco/fisiologia , Ativação Transcricional , Geleia de Wharton/citologia
5.
J Biomed Sci ; 19: 80, 2012 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-22931352

RESUMO

BACKGROUND: Daidzein, a phytoestrogen found in isoflavone, is known to exert neurotrophic and neuroprotective effects on the nervous system. Using primary rat dorsal root ganglion (DRG) neuronal cultures, we have examined the potential neurite outgrowth effect of daidzein. METHODS: Dissociated dorsal root ganglia (DRG) cultures were used to study the signaling mechanism of daidzein-induced neuritogenesis by immunocytochemistry and Western blotting. RESULTS: In response to daidzein treatment, DRG neurons showed a significant increase in total neurite length and in tip number per neuron. The neuritogenic effect of daidzein was significantly hampered by specific blockers for Src, protein kinase C delta (PKCδ) and mitogen-activated protein kinase/extracellular signal-regulated kinase kinases (MEK/ERK), but not by those for estrogen receptor (ER). Moreover, daidzein induced phosphorylation of Src, PKCδ and ERK. The activation of PKCδ by daidzein was attenuated in the presence of a Src kinase inhibitor, and that of ERK by daidzein was diminished in the presence of either a Src or PKCδ inhibitor. CONCLUSION: Daidzein may stimulate neurite outgrowth of DRG neurons depending on Src kinase, PKCδ and ERK signaling pathway.


Assuntos
Gânglios Espinais , Isoflavonas/farmacologia , Neuritos , Fármacos Neuroprotetores/farmacologia , Animais , Células Cultivadas , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/crescimento & desenvolvimento , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Proteína Oncogênica pp60(v-src)/antagonistas & inibidores , Proteína Oncogênica pp60(v-src)/metabolismo , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais
6.
Toxicology ; 302(1): 11-7, 2012 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-22813906

RESUMO

High serum levels of p-cresol have been associated with cardiovascular diseases. This study investigated the effects of p-cresol on gap junctions in neonatal cultured cardiomyocytes. p-Cresol reduced the spontaneous contraction rates of cardiomyocytes, and caused irregular cardiomyocyte beating. Junctional connexin 43 (Cx43) plaques became smaller in size and the gap junction intercellular communication (GJIC) impaired. Moreover, p-cresol increased intracellular Ca(+2) levels, and induced Ca(+2)-dependent protein kinase Cα (PKCα) activation. p-Cresol decreased P1 and P2 Cx43 levels, and increased non-phosphorylated S368-Cx43 levels. The above changes as well as Cx43 disassembly and GJIC decrease induced by p-cresol were prevented by the BAPTA-AM or PKCα inhibitor Gö6976. These results suggest that PKCα mediates p-cresol-induced gap junction disassembly and GJIC dysfunction via S368-Cx43 serine dephosphorylation. This hypothesis was further confirmed in H9c2 cells by siRNA approach. SiRNA knockdown of PKCα prevented p-cresol-induced increase in nonphosphorylated Cx43. This finding supports the association of p-cresol and cardiovascular diseases.


Assuntos
Cálcio/metabolismo , Cresóis/toxicidade , Junções Comunicantes/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Comunicação Celular , Linhagem Celular , Células Cultivadas , Junções Comunicantes/metabolismo , Técnicas de Silenciamento de Genes , Miócitos Cardíacos/metabolismo , Fosforilação , Proteína Quinase C-alfa/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley
7.
J Cell Biochem ; 113(3): 1034-43, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22213462

RESUMO

Uremic patients have a much higher risk of cardiovascular diseases and death. Uremic toxins are probably involved in the development of vascular endothelial dysfunction. Indoxyl sulfate (IS) is a uremic toxin that accumulates with deterioration of renal function. This study explored the effects of IS on the adherens junctions of vascular endothelial cells and revealed the underlying mechanism. Bovine pulmonary artery endothelial cells (BPAECs) were treated with IS, and the distribution of vascular endothelial cadherin (VE-cadherin), p120-catenin, ß-catenin, and stress fibers was examined by immunofluorescence. IS treatment resulted in disruption of intercellular contacts between BPAECs with prominent parallel-oriented intracellular stress fiber formation. Intracellular free radical levels which measured by flow cytometry increased after IS treatment. The antioxidant, MnTMPyP, and an ERK pathway inhibitor, U0126, both significantly prevented IS-induced disruption of intercellular contacts. Western blotting analyses demonstrated that IS-induced phosphorylation of myosin light chain kinase (MLCK) and myosin light chains (MLC) as well as activation of extracellular-signal-regulated protein kinase (ERK1/ERK2). Pretreatment with MnTMPyP prevented ERK1/2 phosphorylation. U0126 prevented the IS-induced MLCK and MLC phosphorylation. MEK-ERK acted as the upstream regulator of the MLCK-MLC pathway. These findings suggest that the superoxide anion-MEK-ERK-MLCK-MLC signaling mediates IS-induced junctional dispersal of BPAECs.


Assuntos
Junções Aderentes/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Indicã/farmacologia , Sistema de Sinalização das MAP Quinases , Junções Aderentes/ultraestrutura , Animais , Butadienos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/ultraestrutura , Células Endoteliais/enzimologia , Células Endoteliais/ultraestrutura , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Nitrilas/farmacologia , Superóxidos/metabolismo
8.
J Sci Food Agric ; 91(4): 694-702, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21302324

RESUMO

BACKGROUND: Schisandra chinensis, commonly used in Asia for tea material and traditional Chinese medicine, is presumed to enhance mental and intellectual functions. In this study, the effects and signalling mechanisms of a purified compound schisandrin, one of the lignan of Schisandra chinensis, on primary cultured hippocampal neurons were investigated. RESULTS: Schisandrin treatment enhanced total dendritic length and branching complexity, both of which were significantly suppressed in the presence of specific blockers for calmodulin-dependent kinase II (CaMKII), protein kinase C epsilon (PKCε), and mitogen activated protein kinase kinase (MEK). Moreover, schisandrin induced calcium influx, and phosphorylation of CaMKII, PKCε, and MEK. Inhibition of CAMKII and PKCε attenuated the schisandrin-induced phosphorylation of PKCε and MEK, and the phosphorylation of MEK, respectively. Moreover, schisandrin also stimulated the phosphorylation of cyclic AMP responsive-element binding protein (CREB) at Ser-133, an effect that was blocked by KN93. In addition to its neuritogenic effects, schisandrin increased the numbers of postsynaptic density-95-positive and FM1-43-positive puncta in dendrites and synaptic boutons, respectively. CONCLUSION: In hippocampal neurons, schisandrin exhibits neurotrophic properties that are mediated by the CaMKII-PKCε-MEK pathway.


Assuntos
Ciclo-Octanos/farmacologia , Dendritos/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Lignanas/farmacologia , Neurogênese/efeitos dos fármacos , Extratos Vegetais/farmacologia , Compostos Policíclicos/farmacologia , Densidade Pós-Sináptica/efeitos dos fármacos , Schisandra/química , Animais , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/metabolismo , Dendritos/metabolismo , Frutas , Hipocampo/crescimento & desenvolvimento , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , Densidade Pós-Sináptica/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Wistar
9.
FEBS Lett ; 585(4): 705-10, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21251911

RESUMO

An acidic microenvironment induces disruption of adherens junctions (AJs) of hepatoma cells. This study investigated the impact of an acidic extracellular pH (pHe) on p120-catenin (p120-ctn) serine phosphorylation. pH 6.6 treatment increased intracellular calcium levels, activated protein kinase C (PKC)α and PKCδ, and decreased serine phosphorylation of p120-ctn. Further knockdown of PKCα and δ by small interference RNA (siRNA) prevented the pH 6.6-induced downregulation of p120-ctn at AJ and the serine dephosphorylation of p120-ctn. Moreover, PP2 pretreatment and siRNA of c-Src abrogated the pH 6.6-induced PKCδ activation. Together, the c-Src-PKCδ cascade and PKCα regulate the acidic pHe-induced AJ disruption.


Assuntos
Junções Aderentes/metabolismo , Cateninas/metabolismo , Proteína Quinase C-delta/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Inibidores Enzimáticos , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Fosforilação , Proteína Quinase C-alfa/genética , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/genética , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno , Serina/metabolismo , Fatores de Tempo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética , delta Catenina
10.
J Agric Food Chem ; 58(22): 11645-52, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-20961042

RESUMO

Cordycepin, a nucleoside isolated from Cordyceps sinensis, is an inhibitor of polyadenylation and has an antitumor effect. We used CGTH W-2, a follicular thyroid carcinoma cell line, to study the mechanism of the anticancer effect of cordycepin. Cordycepin decreased cell viability and resulted in apoptosis but not necrosis. Cordycepin increased intracellular calcium levels triggering calpain activation, which led to apoptosis. BAPTA/AM and calpeptin inhibited the cordycepin-induced cleavage of caspase 7 and poly (ADP-ribose) polymerase (PARP), implying an upstream role of calcium and calpain. CGTH W-2 cells expressed four subtypes of adenosine receptors (AR), A1AR, A2AAR, A2BAR, and A3AR. Specific antagonists to AR subtypes all blocked cordycepin-induced apoptosis to different degrees. Small interfering RNA for A1AR and A3AR abrogated cordycepin-induced apoptosis. In conclusion, the cordycepin-induced apoptosis of CGTH W-2 cells is mediated by the calcium-calpain-caspase 7-PARP pathway, and ARs are involved in the apoptotic effect of cordycepin.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Calpaína/metabolismo , Caspase 7/metabolismo , Desoxiadenosinas/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Calpaína/genética , Caspase 7/genética , Linhagem Celular Tumoral , Cordyceps/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Poli(ADP-Ribose) Polimerases/genética , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/fisiopatologia
11.
FEBS Lett ; 584(21): 4442-8, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20937274

RESUMO

Adenosine (Ado), an endogenous nucleoside, can stimulate corticosterone synthesis in adrenal cells via the A(2A)/A(2B) adenosine receptors (ARs). This study evaluated the contribution of protein kinase C (PKC) isoforms in Ado-induced steroidogenesis. The PKC inhibitor calphostin c blocked Ado-induced steroidogenesis, the mitogen-activated protein kinase (MEK)-extracellular signal-related regulated kinase (ERK)-cyclic AMP responsive element-binding protein cascade, and the mRNA expression of steroidogenic acute regulatory protein and CYP11B1. Further analyses revealed that PKCµ was indeed activated by Ado. Moreover, downregulation of PKCµ by small interfering RNA (siRNA) inhibited Ado-stimulated steroidogenesis and ERK phosphorylation. Finally, inhibition of either A(2A)AR or A(2B)AR led to the suppression of PKCµ phosphorylation. Together, these findings suggest that A(2)AR-PKCµ-MEK signaling mediates Ado-stimulated adrenal steroidogenesis.


Assuntos
Adenosina/farmacologia , Glândulas Suprarrenais/citologia , Proteína Quinase C/metabolismo , Esteroides/biossíntese , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosfoproteínas/genética , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptor A2B de Adenosina/metabolismo
12.
J Cell Biochem ; 110(2): 428-37, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20225271

RESUMO

This study examined the effect of rottlerin on the focal adhesion-mediated cell migration of CGTH W-2 human follicular thyroid carcinoma cells. Rottlerin (10 microM) resulted in decreased adhesion of CGTH W-2 cells to matrix substance, which was correlated with metastatic potential. Rottlerin treatment also resulted in a marked reduction in the migration of CGTH W-2 cells. Protein levels of integrin beta1, FAK, and paxillin were decreased by rottlerin. Consistent with this, immunostaining of FAK, vinculin, and paxillin revealed disassembly of the focal adhesions. Disruption of actin stress fibers was noted, which was compatible with reduced expression levels and activities of Rac-1 and Rho. The effect of rottlerin on cell migration was not attributable to inhibition of PKCdelta activity since siRNA knockdown of PKCdelta did not recapitulate the effects of rottlerin on cell adhesion and migration. Furthermore, activation of PKCdelta by phorbol esters failed to restore the rottlerin-inhibited migratory ability. The mitochondrial uncoupler, carbonylcyanide-4-(trifluoromethoxy)-phenylhydrazone, was able to mimic several rottlerin's effects. In summary, we demonstrated that rottlerin inhibits the migration of CGTH W-2 cells by disassembly of focal adhesion complexes in a PKCdelta-independent manner, and might play as a mitochondrial uncoupler role in these events.


Assuntos
Acetofenonas/farmacologia , Benzopiranos/farmacologia , Movimento Celular/efeitos dos fármacos , Adesões Focais , Proteína Quinase C-delta/metabolismo , Neoplasias da Glândula Tireoide/patologia , Apoptose , Western Blotting , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Microscopia de Fluorescência , Proteína Quinase C-delta/genética , RNA Interferente Pequeno , Neoplasias da Glândula Tireoide/enzimologia
13.
Int J Biochem Cell Biol ; 42(5): 762-70, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20093193

RESUMO

Astrocytes play a crucial role in maintaining the homeostasis of the brain. Changes to gap junctional intercellular communication (GJIC) in astrocytes and excessive inflammation may trigger brain damage and neurodegenerative diseases. In this study, we investigated the effect of lipopolysaccharide (LPS) on connexin43 (Cx43) gap junctions in rat primary astrocytes. Following LPS treatment, dose- and time-dependent inhibition of Cx43 expression was seen. Moreover, LPS induced a reduction in Cx43 immunoreactivity at cell-cell contacts and significantly inhibited GJIC, as revealed by the fluorescent dye scrape loading assay. Toll-like receptor 4 (TLR4) protein expression was increased 2-3-fold following LPS treatment. To study the pathways underlying these LPS-induced effects, we examined downstream effectors of TLR4 signaling and found that LPS induced a significant increase in phosphorylated extracellular signal-regulated kinase (pERK) levels up to 6 h, followed by signal attenuation and downregulation of caveolin-3 expression. Interestingly, LPS treatment also induced a dramatic increase in inducible nitric oxide synthase (iNOS) levels at 6 h, which were sustained up to 18-24 h. The LPS-induced downregulation of Cx43 and caveolin-3 was prevented by co-treatment of astrocytes with the iNOS cofactor inhibitor 1400W, but not the ERK inhibitor PD98059. Specific knockdown of caveolin-3 using siRNA had a significant inhibitory effect on GJIC and resulted in a downregulation of Cx43. Our results suggest that long-term LPS treatment of astrocytes leads to inhibition of Cx43 gap junction communication by the activation of iNOS and downregulation of caveolin-3 via a TLR4-mediated signaling pathway.


Assuntos
Astrócitos/fisiologia , Caveolina 3/metabolismo , Comunicação Celular , Conexina 43/metabolismo , Regulação para Baixo , Junções Comunicantes/fisiologia , Lipopolissacarídeos/toxicidade , Animais , Animais Recém-Nascidos , Astrócitos/patologia , Caveolina 3/genética , Células Cultivadas , Conexina 43/genética , Ativação Enzimática , Corantes Fluorescentes , Junções Comunicantes/patologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Ratos , Ratos Wistar , Transdução de Sinais , Fatores de Tempo , Receptor 4 Toll-Like/metabolismo , Regulação para Cima
14.
J Cell Biochem ; 108(4): 851-9, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19711372

RESUMO

We have previously shown that culturing HepG2 cells in pH 6.6 culture medium increases the c-Src-dependent tyrosine phosphorylation of beta-catenin and induces disassembly of adherens junctions (AJs). Here, we investigated the upstream mechanism leading to this pH 6.6-induced modification of E-cadherin. In control cells cultured at pH 7.4, E-cadherin staining was linear and continuous at cell-cell contact sites. Culturing cells at pH 6.6 was not cytotoxic, and resulted in weak and discontinuous junctional E-cadherin staining, consistent with the decreased levels of E-cadherin in membrane fractions. pH 6.6 treatment activated c-Src and Fyn kinase and induced tyrosine phosphorylation of p120 catenin (p120ctn) and E-cadherin. Inhibition of Src family kinases by PP2 attenuated the pH 6.6-induced tyrosine phosphorylation of E-cadherin and p120ctn, and prevented the loss of these proteins from AJs. In addition, E-cadherin was bound to Hakai and ubiquitinated. Furthermore, pH 6.6-induced detachment of E-cadherin from AJs was blocked by pretreatment with MG132 or NH(4)Cl, indicating the involvement of ubiquitin-proteasomal/lysosomal degradation of E-cadherin. An early loss of p120ctn prior to E-cadherin detachment from AJs was noted, concomitant with a decreased association between p120ctn and E-cadherin at pH 6.6. PP2 pretreatment prevented the dissociation of these two proteins. In conclusion, pH 6.6 activated Src kinases, resulting in tyrosine phosphorylation of E-cadherin and p120ctn and a weakening of the association of E-cadherin with p120ctn and contributing to the instability of E-cadherin at AJs.


Assuntos
Junções Aderentes/metabolismo , Caderinas/química , Cateninas/química , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Quinases da Família src/metabolismo , Membrana Celular/metabolismo , Inibidores Enzimáticos/farmacologia , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Lisossomos/química , Fosforilação , Complexo de Endopeptidases do Proteassoma/química , Ubiquitina/química , delta Catenina
15.
Int J Biochem Cell Biol ; 41(11): 2323-33, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19523531

RESUMO

Caveolin-3, the major caveolin isoform in cardiomyocytes, plays an important role in the rapid signaling pathways initiated by stimulation of the membrane-associated molecules. To examine the role of caveolin-3 in regulating estrogen receptor alpha in cardiomyocytes, we investigate whether the membrane estrogen receptor alpha associates with caveolin-3 and whether this association is linked to the 17beta-estradiol-mediated signals. In control cardiomyocytes, following discontinuous sucrose gradient centrifugation, caveolin-3 was found predominantly in the lipid raft buoyant fractions, whereas it was distributed to both the buoyant and non-lipid raft heavy fractions following metabolic inhibition treatment. Confocal microscopy showed that estrogen receptor alpha co-localized with caveolin-3 on the plasma membrane of neonatal and adult rat cardiomyocytes. This membrane labeling of estrogen receptor alpha was not seen following treatment with the cholesterol-depleting agent methyl-beta-cyclodextrin (5mM), whereas metabolic inhibition had little effect on the membrane distribution of estrogen receptor alpha. Metabolic inhibition induced tyrosine phosphorylation of caveolin-3 and decreased its association with estrogen receptor alpha, both effects being mediated via a Src activation mechanism, since they were inhibited by the selective tyrosine kinase inhibitor PP2. Metabolic inhibition also induced tyrosine phosphorylation of connexin43 and increased its association with c-Src, both effects being prevented by 17beta-estradiol (200 nM). The effect of 17beta-estradiol on metabolic inhibition-induced tyrosine phosphorylation of connexin43 was inhibited by the specific estrogen receptor antagonist ICI182780. These data identify cardiac caveolin-3 as juxtamembrane scaffolding for estrogen receptor alpha docking at caveolae, which provide a unique compartment for conveying 17beta-estradiol-elicited, rapid signaling to regulate connexin43 phosphorylation during ischemia.


Assuntos
Caveolina 3/metabolismo , Conexina 43/metabolismo , Receptor alfa de Estrogênio/metabolismo , Miócitos Cardíacos/metabolismo , Envelhecimento/patologia , Animais , Centrifugação com Gradiente de Concentração , Ativação Enzimática/efeitos dos fármacos , Estradiol/farmacologia , Imunofluorescência , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/enzimologia , Microscopia Confocal , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Pirimidinas/farmacologia , Ratos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , beta-Ciclodextrinas/farmacologia , Quinases da Família src/metabolismo
16.
Int J Biochem Cell Biol ; 41(7): 1536-46, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19166962

RESUMO

Oleic acid (OA) affects assembly of gap junctions in neonatal cardiomyocytes. Adherens junction (AJ) regulates the stability of gap junction integrity; however, the effect of OA on AJ remains largely unexplored. The distribution of N-cadherin and catenins at cell-cell junction was decreased by OA. OA induced activation of protein kinase C(PKC)-alpha and -epsilon and Src family kinase, and all three kinases were involved in the oleic acid-induced disassembly of the adherens junction, since it was blocked by pretreatment with Gö6976 (a PKCalpha inhibitor), epsilonV1-2 (a PKCepsilon inhibitor), or PP2 (a Src family kinase inhibitor). Src family kinase appeared to be the downstream of PKC-alpha and -epsilon, as blockade of either PKC-alpha or -epsilon activity prevented the OA-induced activation of Src family kinase. Immunoprecipitation analyses showed that OA activated Fyn and Fer. OA promoted the association of p120 catenin/beta-catenin with Fyn and Fer and caused increased tyrosine phosphorylation of p120 catenin and beta-catenin, resulting in decreased binding of the former to N-cadherin and of the latter to alpha-catenin. Pretreatment with PP2 abrogated this OA-induced tyrosine phosphorylation of p120 catenin and beta-catenin and restored the association of N-cadherin with p120 catenin and that of beta-catenin with alpha-catenin. In conclusion, these results show that OA activates the PKC-Fyn signaling pathway, leading to the disassembly of the AJ. Therefore, inhibitors of PKC-alpha/-epsilon and Src family kinase are potential candidates as cardioprotection agents against OA-induced heart injury during ischemia-reperfusion.


Assuntos
Junções Aderentes/efeitos dos fármacos , Junções Aderentes/enzimologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Ácido Oleico/farmacologia , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Cateninas , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Modelos Biológicos , Miócitos Cardíacos/citologia , Necrose/enzimologia , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-épsilon/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar , beta Catenina/metabolismo , Quinases da Família src/metabolismo , delta Catenina
17.
Cell Biol Int ; 32(11): 1371-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18789391

RESUMO

18Beta-glycyrrhetinic acid (18beta-GA) regulates serine/threonine dephosphorylation of connexin43 (Cx43). Phospho-specific antibodies were used here to determine the effect of 18beta-GA on serine 368-phosphorylated Cx43 (pSer368Cx43) in cultured rat neonatal cardiomyocytes by immunofluorescence microscopy and immunoblot analyses. 18beta-GA caused a time-dependent increase in pSer368Cx43 levels and induced gap junction disassembly, shown by a change in pSer368Cx43 immunostaining from large aggregates to dispersed punctates at cell-cell contact areas. 18beta-GA also induced a time-dependent increase in the levels of serine 729-phosphorylated PKCepsilon, the active form of PKCepsilon. The 18beta-GA-induced increase in pSer368Cx43 levels and changes in pSer368Cx43 staining pattern were abolished by the PKC inhibitor, chelerythrine. Furthermore, 18beta-GA increased the co-immunoprecipitation of Cx43 with PKCepsilon. However, the 18beta-GA-induced increase in pSer368Cx43 levels and increased association of Cx43 with PKCepsilon were inhibited by co-treatment with the protein phosphatase type 1 and type 2A inhibitor, calyculin A. We conclude that 18beta-GA induces Ser368 phosphorylation of Cx43 via PKCepsilon.


Assuntos
Conexina 43/metabolismo , Junções Comunicantes/metabolismo , Ácido Glicirretínico/farmacologia , Miócitos Cardíacos/metabolismo , Proteína Quinase C-épsilon/efeitos dos fármacos , Sequência de Aminoácidos/efeitos dos fármacos , Sequência de Aminoácidos/fisiologia , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/farmacologia , Antineoplásicos/farmacologia , Benzofenantridinas/farmacologia , Células Cultivadas , Conexina 43/química , Conexina 43/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/ultraestrutura , Imuno-Histoquímica , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/ultraestrutura , Fosforilação/efeitos dos fármacos , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Wistar , Serina/química , Serina/metabolismo , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
18.
Int J Biochem Cell Biol ; 40(12): 2815-25, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18582595

RESUMO

Adenosine promotes adrenal steroidogenesis in vitro and in vivo. However, the underlying signaling mechanisms of this event and the function of the adenosine receptor subtypes in adrenal cells remain to be elucidated. Expression of A1, A2A, A2B, and A3 adenosine receptor mRNA in rat adrenal cells was shown by reverse transcription-polymerase chain reaction. Adenosine increased corticosterone production in a time- and dose-dependent manner, and this adenosine effect was mediated by the A2 adenosine receptors, since the antagonists specific for the A2A and A2B adenosine receptors, and specific silencing the A2A adenosine receptor expression with small interfering RNA significantly blocked the adenosine-induced steroidogenesis. Using pharmacological approaches, we further demonstrated that Janus kinase 2 was the downstream molecule next to the A2A and A2B adenosine receptors. Inhibition of Janus kinase 2 prevented the adenosine-induced steroidogenesis and phosphorylation of mitogen-activated protein kinase kinase 1/2 and extracellular signal-regulated kinase 1/2, demonstrating that Janus kinase 2 was the upstream effector of the mitogen-activated protein kinase kinase pathway. Pretreatment with A2 adenosine receptor, Janus kinase 2, or mitogen-activated protein kinase kinase inhibitors significantly decreased the adenosine-induced phosphorylation of 3',5'-cyclic adenosine monophosphate responsive element binding protein. In conclusion, these data show that adenosine-stimulated steroidogenesis is mediated via the A2A and A2B adenosine receptors, activation of which triggers the Janus kinase 2-mitogen-activated protein kinase kinase-extracellular signal-regulated kinase cascade and 3',5'-cyclic adenosine monophosphate responsive element binding protein phosphorylation. Based on its stimulatory effect on glucocorticoid production, adenosine is a potential candidate as anti-inflammatory agent.


Assuntos
Glândulas Suprarrenais/metabolismo , Janus Quinase 2/metabolismo , Receptor A2A de Adenosina/metabolismo , Receptor A2B de Adenosina/metabolismo , Esteroides/biossíntese , Adenosina/farmacologia , Glândulas Suprarrenais/citologia , Animais , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Ratos , Ratos Wistar
19.
Int J Cancer ; 123(4): 801-9, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18506685

RESUMO

Lysophosphatidic acid (LPA), known as the "ovarian cancer activating factor," is a natural phospholipid involved in important biological functions, such as cell proliferation, wound healing and neurite retraction. LPA causes colony dispersal in various carcinoma cell lines by inducing morphological changes, including membrane ruffling, lamellipodia formation, cell-cell dissociation and single cell migration. However, its effects on cell-cell dissociation and cell-cell adhesion of ovarian cancer cells have not been studied. In our study, we showed that LPA induced sequential events of intercellular junction dispersal and "half-junction" formation in ovarian cancer SKOV3 cells and that Src-family kinases were involved in both processes, since the effects were abolished by the selective tyrosine kinase inhibitor PP2. LPA induced rapid and transient activation of Src family kinases, which were recruited to cell-cell junctions by increasing the association with the adherens junction protein p120-catenin. We identified the Src family kinase, Fyn, as the key component associated with p120-catenin after LPA stimulation in SKOV3 cells. Our study provides evidence that LPA induces junction dispersal in ovarian cancer SKOV3 cells by activating the Src family kinase Fyn and increasing its association with p120-catenin at the cell-cell junction.


Assuntos
Junções Aderentes/efeitos dos fármacos , Junções Aderentes/enzimologia , Moléculas de Adesão Celular/metabolismo , Lisofosfolipídeos/farmacologia , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Cateninas , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-fyn/antagonistas & inibidores , Pirimidinas/farmacologia , alfa Catenina/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo , delta Catenina
20.
Cancer Lett ; 267(1): 37-48, 2008 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-18423982

RESUMO

Little attention has been paid to the role of adherens junctions (AJs) in acidic extracellular pH (pHe)-induced cell invasion. Incubation of HepG2 cells in acidic medium (pH 6.6) induced cell dispersion from tight cell clusters, and this change was accompanied by downregulation of beta-catenin at cell junctions and a rapid activation of c-Src. Pretreatment with PP2 prevented the acidic pH-induced downregulation of beta-catenin at AJ and in the membrane fractions. The acidic pHe-induced c-Src activation increased tyrosine phosphorylation of beta-catenin and decreased the amount of beta-catenin-associated E-cadherin. The depletion of membrane-bound beta-catenin coincided with enhanced cell migration and invasion, and this acidic pHe-increased cell migration and invasion was prevented by PP2. In conclusion, this study characterizes a novel signaling pathway responsible for acidic microenvironment-promoted migration and invasive behaviors of cancer cells.


Assuntos
Junções Aderentes/metabolismo , Concentração de Íons de Hidrogênio , beta Catenina/metabolismo , Quinases da Família src/farmacologia , Junções Aderentes/fisiologia , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Humanos , Fosforilação , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...