Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Br J Cancer ; 109(12): 3034-41, 2013 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24178758

RESUMO

BACKGROUND: Persin is a plant toxin that displays synergistic cytotoxicity with tamoxifen in human breast cancer cell lines. Here, we examined the ability of persin to circumvent tamoxifen resistance and delineated the intracellular signalling pathways involved. METHODS: The induction of apoptosis in tamoxifen-resistant and -sensitive breast cancer cells was measured by flow cytometry following treatment with persin±tamoxifen. Markers of endoplasmic reticulum stress (ERS) were analysed following treatment, and their causal role in mediating persin-induced apoptosis was determined using chemical inhibitors and RNA interference. RESULTS: Cells that were resistant to an apoptotic concentration of tamoxifen maintained an apoptotic response to persin. Persin-induced apoptosis was associated with an increase in markers of ERS, that is, CHOP expression and XBP-1 splicing and was decreased by CHOP siRNA. The CASP-4 inhibitor Z-YVAD-FMK markedly inhibited persin-induced apoptosis in both tamoxifen-sensitive and -resistant cells. CONCLUSION: The cytotoxic effects of persin are CASP-4 dependent and mediated by CHOP-dependent and -independent ERS signalling cascades. Increased ERS signalling contributes to persin-induced reversal of tamoxifen resistance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Álcoois Graxos/farmacologia , Extratos Vegetais/farmacologia , Tamoxifeno/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Álcoois Graxos/administração & dosagem , Feminino , Humanos , Células MCF-7 , Transdução de Sinais , Tamoxifeno/administração & dosagem
2.
Diabetologia ; 55(11): 2999-3009, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22893028

RESUMO

AIMS/HYPOTHESIS: Pancreatic beta cell destruction in type 1 diabetes may be mediated by cytokines such as IL-1ß, IFN-γ and TNF-α. Endoplasmic reticulum (ER) stress and nuclear factor-κB (NFκB) signalling are activated by cytokines, but their significance in beta cells remains unclear. Here, we investigated the role of cytokine-induced ER stress and NFκB signalling in beta cell destruction. METHODS: Isolated mouse islets and MIN6 beta cells were incubated with IL-1ß, IFN-γ and TNF-α. The chemical chaperone 4-phenylbutyric acid (PBA) was used to inhibit ER stress. Protein production and gene expression were assessed by western blot and real-time RT-PCR. RESULTS: We found in beta cells that inhibition of cytokine-induced ER stress with PBA unexpectedly potentiated cell death and NFκB-regulated gene expression. These responses were dependent on NFκB activation and were associated with a prolonged decrease in the inhibitor of κB-α (IκBα) protein, resulting from increased IκBα protein degradation. Cytokine-mediated NFκB-regulated gene expression was also potentiated after pre-induction of ER stress with thapsigargin, but not tunicamycin. Both PBA and thapsigargin treatments led to preferential upregulation of ER degradation genes over ER-resident chaperones as part of the adaptive unfolded protein response (UPR). In contrast, tunicamycin activated a balanced adaptive UPR in association with the maintenance of Xbp1 splicing. CONCLUSIONS/INTERPRETATION: These data suggest a novel mechanism by which cytokine-mediated ER stress interacts with NFκB signalling in beta cells, by regulating IκBα degradation. The cross-talk between the UPR and NFκB signalling pathways may be important in the regulation of cytokine-mediated beta cell death.


Assuntos
Morte Celular/fisiologia , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , NF-kappa B/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Animais , Antineoplásicos/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Citocinas/farmacologia , Diabetes Mellitus Tipo 1/patologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/fisiologia , Inibidores Enzimáticos/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Interferon gama/metabolismo , Interferon gama/farmacologia , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas , Fenilbutiratos/farmacologia , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tapsigargina/farmacologia , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Tunicamicina/farmacologia
3.
Diabetologia ; 55(10): 2682-2692, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22820510

RESUMO

AIMS/HYPOTHESIS: Activation of the G protein-coupled receptor (GPR)40 by long-chain fatty acids potentiates glucose-stimulated insulin secretion (GSIS) from pancreatic beta cells, and GPR40 agonists are in clinical development for type 2 diabetes therapy. GPR40 couples to the G protein subunit Gα(q/11) but the signalling cascade activated downstream is unknown. This study aimed to determine the mechanisms of GPR40-dependent potentiation of GSIS by fatty acids. METHODS: Insulin secretion in response to glucose, oleate or diacylglycerol (DAG) was assessed in dynamic perifusions and static incubations in islets from wild-type (WT) and Gpr40 (-/-) mice. Depolymerisation of filamentous actin (F-actin) was visualised by phalloidin staining and epifluorescence. Pharmacological and molecular approaches were used to ascertain the roles of protein kinase D (PKD) and protein kinase C delta in GPR40-mediated potentiation of GSIS. RESULTS: Oleate potentiates the second phase of GSIS, and this effect is largely dependent upon GPR40. Accordingly, oleate induces rapid F-actin remodelling in WT but not in Gpr40 (-/-) islets. Exogenous DAG potentiates GSIS in both WT and Gpr40 (-/-) islets. Oleate induces PKD phosphorylation at residues Ser-744/748 and Ser-916 in WT but not Gpr40 (-/-) islets. Importantly, oleate-induced F-actin depolymerisation and potentiation of GSIS are lost upon pharmacological inhibition of PKD1 or deletion of Prkd1. CONCLUSIONS/INTERPRETATION: We conclude that the signalling cascade downstream of GPR40 activation by fatty acids involves activation of PKD1, F-actin depolymerisation and potentiation of second-phase insulin secretion. These results provide important information on the mechanisms of action of GPR40, a novel drug target for type 2 diabetes.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteína Quinase C/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Actinas/metabolismo , Animais , Células Cultivadas , Diglicerídeos/farmacologia , Glucose/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Knockout , Modelos Animais , Ácido Oleico/farmacologia , Proteína Quinase C-delta/deficiência , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/fisiologia , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/fisiologia
4.
Diabetologia ; 54(7): 1766-76, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21472432

RESUMO

AIMS/HYPOTHESIS: Pro-inflammatory cytokines such as IL-1ß, IFN-γ and TNF-α may contribute to pancreatic beta cell destruction in type 1 diabetes. A mechanism requiring nitric oxide, which is generated by inducible nitric oxide synthase (iNOS), in cytokine-induced endoplasmic reticulum (ER) stress and apoptosis has been proposed. Here, we tested the role of nitric oxide in cytokine-induced ER stress and the subsequent unfolded protein response (UPR) in beta cells. METHODS: Isolated islets from wild-type and iNos (also known as Nos2) knockout (iNos ( -/- )) mice, and MIN6 beta cells were incubated with IL-1ß, IFN-γ and TNF-α for 24-48 h. N (G)-methyl-L: -arginine was used to inhibit nitric oxide production in MIN6 cells. Protein levels and gene expression were assessed by western blot and real-time RT-PCR. RESULTS: In islets and MIN6 cells, inhibition of nitric oxide production had no effect on the generation of ER stress by cytokines, as evidenced by downregulation of Serca2b (also known as Atp2a2) mRNA and increased phosphorylation of PKR-like ER kinase, Jun N-terminal kinase (JNK) and eukaryotic translation initiation factor 2 α subunit. However, nitric oxide regulated the pattern of UPR signalling, which delineates the cellular decision to adapt to ER stress or to undergo apoptosis. Inhibition of nitric oxide production led to reduced expression of pro-apoptotic UPR markers, Chop (also known as Ddit3), Atf3 and Trib3. In contrast, adaptive UPR markers (chaperones, foldases and degradation enhancers) were increased. Further analysis of mouse islets showed that cytokine-induced Chop and Atf3 expression was also dependent on JNK activity. CONCLUSIONS/INTERPRETATION: The mechanism by which cytokines induce ER stress in mouse beta cells is independent of nitric oxide production. However, nitric oxide may regulate the switch between adaptive and apoptotic UPR signalling.


Assuntos
Citocinas/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Óxido Nítrico/metabolismo , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Células Secretoras de Insulina/citologia , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
5.
Diabetologia ; 54(2): 380-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21103982

RESUMO

AIMS/HYPOTHESIS: Proinflammatory cytokines contribute to beta cell destruction in type 1 diabetes, but the mechanisms are incompletely understood. The aim of the current study was to address the role of the protein kinase C (PKC) isoform PKCδ, a diverse regulator of cell death, in cytokine-stimulated apoptosis in primary beta cells. METHODS: Islets isolated from wild-type or Prkcd(-/-) mice were treated with IL-1ß, TNF-α and IFNγ and assayed for apoptosis, nitric oxide (NO) generation and insulin secretion. Activation of signalling pathways, apoptosis and endoplasmic reticulum (ER) stress were determined by immunoblotting. Stabilisation of mRNA transcripts was measured by RT-PCR following transcriptional arrest. Mice were injected with multiple low doses of streptozotocin (MLD-STZ) and fasting blood glucose monitored. RESULTS: Deletion of Prkcd inhibited apoptosis and NO generation in islets stimulated ex vivo with cytokines. It also delayed the onset of hyperglycaemia in MLD-STZ-treated mice. Activation of ERK, p38, JNK, AKT1, the ER stress markers DDIT3 and phospho-EIF2α and the intrinsic apoptotic markers BCL2 and MCL1 was not different between genotypes. However, deletion of Prkcd destabilised mRNA transcripts for Nos2, and for multiple components of the toll-like receptor 2 (TLR2) signalling complex, which resulted in disrupted TLR2 signalling. CONCLUSIONS/INTERPRETATION: Loss of PKCδ partially protects against hyperglycaemia in the MLD-STZ model in vivo, and against cytokine-mediated apoptosis in vitro. This is accompanied by reduced NO generation and destabilisation of Nos2 and components of the TLR2 signalling pathway. The results highlight a mechanism for regulating proinflammatory gene expression in beta cells independently of transcription.


Assuntos
Apoptose/efeitos dos fármacos , Citocinas/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Isoenzimas/metabolismo , Proteína Quinase C-delta/metabolismo , Animais , Apoptose/genética , Western Blotting , Técnicas In Vitro , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Ilhotas Pancreáticas/citologia , Isoenzimas/genética , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Reação em Cadeia da Polimerase , Proteína Quinase C-delta/genética , Fator de Necrose Tumoral alfa/farmacologia
6.
Diabetologia ; 54(6): 1447-56, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21347625

RESUMO

AIMS/HYPOTHESIS: We examined the time-dependent effects of deletion of the gene encoding protein kinase C epsilon (Prkce) on glucose homeostasis, insulin secretion and hepatic lipid metabolism in fat-fed mice. METHODS: Prkce(-/-) and wild-type (WT) mice were fed a high-fat diet for 1 to 16 weeks and subjected to i.p. glucose tolerance tests (ipGTT) and indirect calorimetry. We also investigated gene expression and protein levels by RT-PCR, quantitative protein profiling (isobaric tag for relative and absolute quantification; iTRAQ) and immunoblotting. Lipid levels, mitochondrial oxidative capacity and lipid metabolism were assessed in liver and primary hepatocytes. RESULTS: While fat-fed WT mice became glucose intolerant after 1 week, Prkce(-/-) mice exhibited normal glucose and insulin levels. iTRAQ suggested differences in lipid metabolism and oxidative phosphorylation between fat-fed WT and Prkce(-/-) animals. Liver triacylglycerols were increased in fat-fed Prkce(-/-) mice, resulting from altered lipid partitioning which promoted esterification of fatty acids in hepatocytes. In WT mice, fat feeding elevated oxygen consumption in vivo and in isolated liver mitochondria, but these increases were not seen in Prkce(-/-) mice. Prkce(-/-) hepatocytes also exhibited reduced production of reactive oxygen species (ROS) in the presence of palmitate. After 16 weeks of fat feeding, however, the improved glucose tolerance in fat-fed Prkce(-/-) mice was instead associated with increased insulin secretion during ipGTT, as we have previously reported. CONCLUSIONS/INTERPRETATION: Prkce deletion ameliorates diet-induced glucose intolerance via two temporally distinct phenotypes. Protection against insulin resistance is associated with changes in hepatic lipid partitioning, which may reduce the acute inhibitory effects of fatty acid catabolism, such as ROS generation. In the longer term, enhancement of glucose-stimulated insulin secretion prevails.


Assuntos
Gorduras na Dieta/metabolismo , Glucose/metabolismo , Homeostase/fisiologia , Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , Proteína Quinase C-épsilon/deficiência , Animais , Deleção de Genes , Insulina/metabolismo , Camundongos , Camundongos Knockout , Modelos Animais , Proteína Quinase C-épsilon/genética , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
7.
Diabetologia ; 52(11): 2369-2373, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19727664

RESUMO

AIMS/HYPOTHESIS: Saturated fatty acids augment endoplasmic reticulum (ER) stress in pancreatic beta cells and this is implicated in the loss of beta cell mass that accompanies type 2 diabetes. However, the mechanisms underlying the induction of ER stress are unclear. Our aim was to establish whether saturated fatty acids cause defects in ER-to-Golgi protein trafficking, which may thereby contribute to ER stress via protein overload. METHODS: Cells of the mouse insulinoma cell line MIN6 were transfected with temperature-sensitive vesicular stomatitis virus G protein (VSVG) tagged with green fluorescent protein to quantify the rate of ER-to-Golgi protein trafficking. I14 antibody, which detects only correctly folded VSVG, was employed to probe the folding environment of the ER. ER stress markers were monitored by western blotting. RESULTS: Pretreatment with palmitate, but not oleate, significantly reduced the rate of ER-to-Golgi protein trafficking assessed using VSVG. This was not secondary to ER stress, since thapsigargin, which compromises chaperone function by depletion of ER calcium, markedly inhibited VSVG folding and promoted strong ER stress but only slightly reduced protein trafficking. Blockade of ER-to-Golgi protein trafficking with brefeldin A (BFA) was sufficient to trigger ER stress, but neither BFA nor palmitate compromised VSVG folding. CONCLUSIONS/INTERPRETATION: Reductions in ER-to-Golgi protein trafficking potentially contribute to ER stress during lipoapoptosis. In this case ER stress would be triggered by protein overload, rather than a disruption of the protein-folding capacity of the ER.


Assuntos
Retículo Endoplasmático/fisiologia , Células Secretoras de Insulina/fisiologia , Glicoproteínas de Membrana/metabolismo , Transporte Proteico/fisiologia , Proteínas/metabolismo , Estresse Fisiológico/fisiologia , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular Tumoral , Cicloeximida/farmacologia , Genes Reporter , Células Secretoras de Insulina/efeitos dos fármacos , Insulinoma , Camundongos , Ácido Palmítico/farmacologia , Transporte Proteico/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Transfecção
8.
Diabetologia ; 52(12): 2616-20, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19809797

RESUMO

AIMS/HYPOTHESIS: This study aimed to determine whether protein kinase C (PKC) delta plays a role in the glucose intolerance caused by a high-fat diet, and whether it could compensate for loss of PKCepsilon in the generation of insulin resistance in skeletal muscle. METHODS: Prkcd (-/-), Prkce (-/-) and wild-type mice were fed high-fat diets and subjected to glucose tolerance tests. Blood glucose levels and insulin responses were determined during the tests. Insulin signalling in liver and muscle was assessed after acute in vivo insulin stimulation by immunoblotting with phospho-specific antibodies. Activation of PKC isoforms in muscle from Prkce (-/-) mice was assessed by determining intracellular distribution. Tissues and plasma were assayed for triacylglycerol accumulation, and hepatic production of lipogenic enzymes was determined by immunoblotting. RESULTS: Both Prkcd (-/-) and Prkce (-/-) mice were protected against high-fat-diet-induced glucose intolerance. In Prkce (-/-) mice this was mediated through enhanced insulin availability, while in Prkcd (-/-) mice the reversal occurred in the absence of elevated insulin. Neither the high-fat diet nor Prkcd deletion affected maximal insulin signalling. The activation of PKCdelta in muscle from fat-fed mice was enhanced by Prkce deletion. PKCdelta-deficient mice exhibited reduced liver triacylglycerol accumulation and diminished production of lipogenic enzymes. CONCLUSIONS/INTERPRETATION: Deletion of genes encoding isoforms of PKC can improve glucose intolerance, either by enhancing insulin availability in the case of Prkce, or by reducing lipid accumulation in the case of Prkcd. The absence of PKCepsilon in muscle may be compensated by increased activation of PKCdelta in fat-fed mice, suggesting that an additional role for PKCepsilon in this tissue is masked.


Assuntos
Gorduras na Dieta/efeitos adversos , Intolerância à Glucose/induzido quimicamente , Proteína Quinase C-delta/deficiência , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/deficiência , Proteína Quinase C-épsilon/metabolismo , Animais , Glicemia/metabolismo , Cruzamentos Genéticos , Deleção de Genes , Intolerância à Glucose/sangue , Intolerância à Glucose/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Músculo Esquelético/enzimologia , Proteína Quinase C-delta/genética , Proteína Quinase C-épsilon/genética , Triglicerídeos/metabolismo
9.
Cell Death Differ ; 10(3): 269-77, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12700627

RESUMO

We have used expression of a kinase dead mutant of PKCalpha (PKCalphaKD) to explore the role of this isoform in salivary epithelial cell apoptosis. Expression of PKCalphaKD by adenovirus-mediated transduction results in a dose-dependent induction of apoptosis in salivary epithelial cells as measured by the accumulation of sub-G1 DNA, activation of caspase-3, and cleavage of PKCdelta and PKCzeta, known caspase substrates. Induction of apoptosis is accompanied by nine-fold activation of c-Jun-N-terminal kinase, and an approximately two to three-fold increase in activated mitogen-activated protein kinase (MAPK) as well as total MAPK protein. Previous studies from our laboratory have shown that PKCdelta activity is essential for the apoptotic response of salivary epithelial cells to a variety of cell toxins. To explore the contribution of PKCdelta to PKCalphaKD-induced apoptosis, salivary epithelial cells were cotransduced with PKCalphaKD and PKCdeltaKD expression vectors. Inhibition of endogenous PKCdelta blocked the ability of PKCalphaKD to induce apoptosis as indicated by cell morphology, DNA fragmentation, and caspase-3 activation, indicating that PKCdelta activity is required for the apoptotic program induced under conditions where PKCalpha is inhibited. These findings indicate that PKCalpha functions as a survival factor in salivary epithelial cells, while PKCdelta functions to regulate entry into the apoptotic pathway.


Assuntos
Apoptose , Células Epiteliais/metabolismo , Proteína Quinase C/fisiologia , Glândulas Salivares/metabolismo , Adenoviridae/genética , Animais , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Separação Celular , Células Cultivadas , DNA/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Fase G1 , Glutationa Transferase/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Glândula Parótida/citologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Proteína Quinase C-delta , Ratos , Fatores de Tempo
10.
Diabetes ; 48(12): 2383-9, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10580427

RESUMO

Although cytoskeletal proteins such as myosin II are implicated in the control of insulin secretion, their precise role is poorly understood. In other secretory cells, myosin II is predominantly regulated via the phosphorylation of the regulatory light chains (RLC). The current study was aimed at investigating RLC phosphorylation in beta-cells. In both the insulin-secreting cell line RINm5F and rat pancreatic islets, the RLC was basally phosphorylated on the myosin light chain kinase sites (Ser19/Thr18). Phosphorylation at these sites was not consistently increased by either metabolic stimuli (glyceraldehyde/glucose) or the depolarizing agent KCl. The RLC sites phosphorylated by protein kinase C (PKC) (Ser1/Ser2) were unphosphorylated in the basal state, not affected by nutrients or KCl, and only slightly increased by the PKC activator phorbol 12-myristate 13-acetate (PMA). Like the other insulin secretagogues, however, PMA did promote serine phosphorylation of the myosin heavy chain (MHC) in RINm5F cells. Phosphopeptide mapping suggested that the same peptide was phosphorylated under both PMA and glyceraldehyde stimulation, which further extends our previous study of the Ca2+-dependent phosphorylation of this protein (Wilson JR, Ludowyke RI, Biden TJ: Nutrient stimulation results in a rapid Ca2+-dependent threonine phosphorylation of myosin heavy chain in rat pancreatic islets and RINm5F cells. J Biol Chem 273:22729-22737, 1998). Overall, our results demonstrate that in RINm5F cells and rat pancreatic islets, MHC phosphorylation correlates better with insulin secretion than phosphorylation of the RLC. We therefore propose that in beta-cells, in contrast to other secretory cells, phosphorylation of the MHC is more important than that of the RLC for regulation of the myosin II protein complex during insulin secretion.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , Cadeias Pesadas de Miosina/metabolismo , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Proteína Quinase C/metabolismo , Animais , Células Cultivadas , Secreção de Insulina , Insulinoma/metabolismo , Insulinoma/fisiopatologia , Ilhotas Pancreáticas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/fisiopatologia , Mapeamento de Peptídeos , Fosfatos/metabolismo , Fosfopeptídeos/análise , Fosforilação , Fosfosserina/metabolismo , Fosfotreonina/metabolismo , Cloreto de Potássio/farmacologia , Ratos , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
11.
Diabetes ; 50(10): 2210-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11574400

RESUMO

We have shown previously that palmitate treatment of C2C12 skeletal muscle myotubes causes inhibition of the protein kinase B (PKB) pathway and hence reduces insulin-stimulated glycogen synthesis through the elevation of intracellular ceramide levels. Ceramide is known to activate both atypical protein kinase C (aPKC) zeta and protein phosphatase (PP) 2A, and each of these effectors has been reported to inhibit PKB. In the present study, palmitate pretreatment was found to elevate PP2A-like activity in myotubes and to prevent its inhibition by insulin. Incubation with the phosphatase inhibitor okadaic acid before insulin stimulation protected against the effect of the fatty acid on PKB phosphorylation. Palmitate was unable to inhibit PKB activity and glycogen synthesis in cells overexpressing the activated PKB mutant (T308D,S473D)-PKBalpha, which is unaffected by phosphatase. In contrast, PKB activity and glycogen synthesis were still inhibited by palmitate in cells overexpressing a membrane-targeted and, hence, activated PKB mutant that retains sensitivity to phosphatase. Although aPKC activity was also increased in palmitate-treated cells, overexpression of wild-type or kinase-dead aPKCzeta did not alter the inhibitory effects of the lipid on either stimulation of PKB or glycogen synthesis by insulin. We conclude that palmitate disrupts insulin signaling in C2C12 myotubes by promoting PP2A-like activity and, therefore, the dephosphorylation of PKB, which in turn reduces the stimulation of glycogen synthesis.


Assuntos
Glicogênio/biossíntese , Isoenzimas/fisiologia , Ácido Palmítico/farmacologia , Fosfoproteínas Fosfatases/fisiologia , Proteína Quinase C/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Animais , Linhagem Celular , Camundongos , Músculo Esquelético/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 2 , Proteínas Proto-Oncogênicas c-akt
12.
Diabetes ; 49(3): 392-8, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10868960

RESUMO

Expression of muscarinic receptors in rat islets, RINm5F cells, and INS-1 cells was established by reverse transcriptase-polymerase chain reaction (RT-PCR) and quantified by RNase protection. Both methods indicated that m3 and m1 receptors were expressed approximately equally in the various cellular preparations and to a much greater extent than the m5 subtype. However, the cell lines, especially RINm5F cells, expressed less of a given receptor subtype than did islets. Immunohistochemistry indicated that m3 receptors were expressed throughout the islet core. Binding studies using the radiolabeled muscarinic receptor antagonist QNB demonstrated a maximal binding capacity of INS-1 cells of 23.0+/-2.9 fmol/mg protein. Functional analyses were undertaken using INS-1 cells stably transfected with either m1 or m3 receptor cDNAs. Overexpression of either receptor did not affect basal responses but markedly enhanced maximal responses to the muscarinic receptor agonist carbachol. Although maximal hydrolysis of phosphatidylinositol 4,5-bisphosphate (Ptd InsP2) was twofold greater in m1-transfectants as compared with m3-transfectants, cell lines overexpressing either receptor gave essentially equivalent secretory responses to a full range of carbachol doses. The results demonstrate that both m1 and m3 muscarinic receptors are well expressed in pancreatic beta-cells, functionally linked to signaling pathways, and capable of initiating insulin secretion with equal potencies.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores Muscarínicos/fisiologia , Animais , Carbacol/farmacologia , Células Cultivadas , Agonistas Colinérgicos/farmacologia , Hidrólise , Imuno-Histoquímica , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Antagonistas Muscarínicos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Quinuclidinil Benzilato/metabolismo , Ratos , Ratos Wistar , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Transfecção
13.
Diabetes ; 50(10): 2237-43, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11574404

RESUMO

Glucagon-like peptide-1 (GLP-1), an insulinotropic and glucoincretin hormone, is a potentially important therapeutic agent in the treatment of diabetes. We previously provided evidence that GLP-1 induces pancreatic beta-cell growth nonadditively with glucose in a phosphatidylinositol-3 kinase (PI-3K)-dependent manner. In the present study, we investigated the downstream effectors of PI-3K to determine the precise signal transduction pathways that mediate the action of GLP-1 on beta-cell proliferation. GLP-1 increased extracellular signal-related kinase 1/2, p38 mitogen-activated protein kinase (MAPK), and protein kinase B activities nonadditively with glucose in pancreatic beta(INS 832/13) cells. GLP-1 also caused nuclear translocation of the atypical protein kinase C (aPKC) zeta isoform in INS as well as in dissociated normal rat beta-cells as shown by immunolocalization and Western immunoblotting analysis. Tritiated thymidine incorporation measurements showed that the p38 MAPK inhibitor SB203580 suppressed GLP-1-induced beta-cell proliferation. Further investigation was performed using isoform-specific pseudosubstrates of classical (alpha, beta, and gamma) or zeta aPKC isoforms. The PKCzeta pseudosubstrate suppressed the proliferative action of GLP-1, whereas the inhibitor of classical PKC isoforms had no effect. Overexpression of a kinase-dead PKCzeta acting as a dominant negative protein suppressed GLP-1-induced proliferation. In addition, ectopic expression of a constitutively active PKCzeta mutant stimulated tritiated thymidine incorporation to the same extent as GLP-1, and the glucoincretin had no growth-promoting action under this condition. The data indicate that GLP-1-induced activation of PKCzeta is implicated in the beta-cell proliferative signal of the insulinotropic hormone. The results are consistent with a model in which GLP-1-induced PI-3K activation results in PKCzeta translocation to the nucleus, which may play a role in the pleiotropic effects (DNA synthesis, metabolic enzymes, and insulin gene expression) of the glucoincretin.


Assuntos
Glucagon/farmacologia , Ilhotas Pancreáticas/citologia , Fragmentos de Peptídeos/farmacologia , Proteína Quinase C/metabolismo , Precursores de Proteínas/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular , Núcleo Celular/metabolismo , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Peptídeo 1 Semelhante ao Glucagon , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Mutação/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno
14.
Diabetes ; 46(2): 169-78, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9000691

RESUMO

We have tested the hypothesis that changes in the levels and cellular location of protein kinase C (PKC) isozymes might be associated with the development of insulin resistance in skeletal muscles from the high-fat-fed rat. Lipid measurements showed that triglyceride and diacylglycerol, an activator of PKC, were elevated four- and twofold, respectively. PKC activity assays indicated that the proportion of membrane-associated calcium-independent PKC was also increased. As determined by immunoblotting, total (particulate plus cytosolic) PKC alpha, epsilon, and zeta levels were not different between control and fat-fed rats. However, the ratio of particulate to cytosolic PKC epsilon in red muscles from fat-fed rats was increased nearly sixfold, suggesting chronic activation. In contrast, the amount of cytosolic PKC theta was downregulated to 45% of control, while the ratio of particulate to cytosolic levels increased, suggesting a combination of chronic activation and downregulation. Interestingly, while insulin infusion in glucose-clamped rats increased the proportion of PKC theta in the particulate fraction of red muscle, this was potentiated by fat-feeding, suggesting that the translocation is a consequence of altered lipid flux rather than a proximal event in insulin signaling. PKC epsilon and theta measurements from individual rats correlated with triglyceride content of red gastrocnemius muscle; they did not correlate with plasma glucose, which was not elevated in fat-fed rats, suggesting that they were not simply a consequence of hyperglycemia. Our results suggest that these specific alterations in PKC epsilon and PKC theta might contribute to the link between increased lipid availability and muscle insulin resistance previously described using high-fat-fed rats.


Assuntos
Gorduras na Dieta/metabolismo , Resistência à Insulina , Isoenzimas/metabolismo , Músculo Esquelético/enzimologia , Proteína Quinase C/metabolismo , Animais , Compartimento Celular , Citosol/enzimologia , Insulina/fisiologia , Membranas/enzimologia , Ratos , Ratos Wistar
15.
Mol Endocrinol ; 6(6): 920-6, 1992 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1323056

RESUMO

Dopamine, a major neurotransmitter in the mammalian nervous system, exerts its physiological effects through receptors of the G-protein-coupled receptor superfamily. Two major classes of dopamine receptor, D1 and D2, are distinguishable by both biochemical and pharmacological criteria. D1 receptors activate adenylyl cyclase, whereas the D2 class of receptors inhibits this second messenger system. Two subtypes of the human dopamine D2 receptor are generated by alternate splicing of the RNA transcript of a single gene. These two forms, termed D2A (long) and D2B (short), differ by the insertion of 29 amino acids within the putative third cytoplasmic loop, an intracellular domain thought to have a role in coupling this class of receptors to particular second messenger systems. We report here that the D2A and D2B structural subtypes are also functionally distinct. Expression of the two subtypes in a fibroblast cell line revealed that while occupation of both receptors leads to an increase in cytosolic free calcium concentration, they differ in their capacity to inhibit cAMP production. At physiological dopamine concentrations, the D2B-mediated inhibition of calcitonin gene-related peptide-stimulated cAMP accumulation is almost double the response mediated by the D2A subtype. Furthermore, the D2B subtype can maximally attenuate cAMP accumulation by up to 85%, whereas the D2A subtype is less effective, maximally inhibiting cAMP accumulation by only 64%. The D2A and D2B subtypes, thus, constitute functionally distinct forms of the dopamine receptor that can couple to multiple intracellular signalling pathways.


Assuntos
Dopamina/metabolismo , Receptores Dopaminérgicos/classificação , Sistemas do Segundo Mensageiro , Animais , Sequência de Bases , Ligação Competitiva , Células CHO/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Cálcio/metabolismo , Linhagem Celular Transformada , Colforsina/farmacologia , Cricetinae , AMP Cíclico/metabolismo , Dopamina/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Humanos , Dados de Sequência Molecular , Receptores Dopaminérgicos/biossíntese , Receptores Dopaminérgicos/genética , Receptores Dopaminérgicos/fisiologia , Proteínas Recombinantes de Fusão/biossíntese , Especificidade da Espécie , Relação Estrutura-Atividade
16.
FEBS Lett ; 492(1-2): 101-6, 2001 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-11248245

RESUMO

The study addressed the functional link between remodelling of the actomyosin cytoskeleton in pancreatic beta-cells and the regulation of insulin secretion. Confocal microscopy revealed that myosin heavy chain (MHC) IIA co-localized very well with filamentous (F)-actin in RINm5F cells but MHCIIB did not. Subcellular localization of MHCIIB was not altered by stimulation with 30 mM KCl (which evokes Ca(2+)-dependent insulin secretion). In contrast MHCIIA redistributed in a manner similar to F-actin, especially towards the apical surface, but also away from peripheral regions towards cell contact points on the basal surface. Finally, Ca(2+)-dependent insulin secretion was inhibited by stabilization of actin filaments with jasplakinolide. The results support a role for the MHCIIA/actin cytoskeleton in regulating insulin secretion.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Miosinas/metabolismo , Membrana Celular/metabolismo , Humanos , Secreção de Insulina , Fosforilação , Frações Subcelulares
17.
Ann N Y Acad Sci ; 488: 317-33, 1986.
Artigo em Inglês | MEDLINE | ID: mdl-3107454

RESUMO

The initial events in signal transduction in insulin-secreting cells are summarized in FIGURE 8. Both nutrient stimuli, such as glucose and amino acids and the muscarinic agonist carbachol (carbamylcholine) raise [Ca2+]i. Although the rise in [Ca2+]i precedes the stimulation of insulin release, it is not a moment-to-moment regulator of release. The metabolizable fuel stimuli cause Ca2+ influx through voltage-dependent Ca2+ channels following depolarization of the membrane potential. In contrast, carbachol, which does not depolarize, elicits Ptd Ins 4,5-P2 hydrolysis, a reaction catalyzed by phospholipase C. The generation of Ins 1,4,5-P3 in this instance is Ca2+ independent, but appears to involve a GTP-binding protein. However, this protein is not a substrate for pertussis toxin. The levels of Ins 1,4,5-P3, which releases Ca2+ from an ATP-dependent Ca2+ pool of the endoplasmic reticulum, are increased prior to the rise in [Ca2+]i. The mitochondria may take up Ca2+ after large increases in [Ca2+]i. A previously proposed second messenger, arachidonic acid, is much less selective than Ins 1,4,5-P3 in that it releases Ca2+ from mitochondria as well as from the endoplasmic reticulum in a slow and irreversible manner. As Ins 1,4,5-P3 is also generated during glucose stimulation of islets, albeit in a Ca2+-dependent manner, this metabolite could mediate not only the action of carbachol but also contribute to amplifying the [Ca2+]i rise in response to glucose.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Aminoácidos/farmacologia , Animais , Ácido Araquidônico , Ácidos Araquidônicos/metabolismo , Cálcio/metabolismo , Carbacol/farmacologia , Glucose/farmacologia , Gliceraldeído/farmacologia , Homeostase , Inositol 1,4,5-Trifosfato , Fosfatos de Inositol/biossíntese , Fosfatos de Inositol/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Potenciais da Membrana
18.
Adv Exp Med Biol ; 441: 263-70, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9781332

RESUMO

Malonyl CoA is a regulator of carnitine palmitoyl transferase 1 (CPT1), the enzyme that controls the transfer of long chain fatty acyl CoA into mitochondria where it is oxidized. Recent studies indicate that in skeletal muscle the concentration of malonyl CoA is acutely (minutes) regulated by changes in its fuel supply and energy expenditure. In response to changes in fuel supply, regulation appears to be due to alterations in the cytosolic concentration of citrate, which is both an allosteric activator of acetyl CoA carboxylase (ACC), the enzyme that catalyzes malonyl CoA synthesis and a source of its precursor, cytosolic acetyl CoA. During exercise and immediately thereafter regulation by citrate appears to be lost and malonyl CoA levels diminish as the result of a decrease in ACC activity secondary to phosphorylation. Sustained increases in the concentration of malonyl CoA have been observed in muscle of a number of insulin-resistant rodents including the Zucker (fa/fa) and GK rats, KKAgy mice, glucose-infused rats and rats in which muscle has been made insulin resistant by denervation. Available data suggest that malonyl CoA could be linked to insulin resistance in these rodents by virtue of its effects on the cytosolic concentration of long chain fatty acyl CoA (LCFA CoA) and one or more protein kinase C isozymes. Whether similar alterations occur in other tissues and contribute to the pathophysiology of the insulin resistance syndrome remains to be determined.


Assuntos
Resistência à Insulina , Malonil Coenzima A/metabolismo , Músculo Esquelético/metabolismo , Animais , Carnitina O-Palmitoiltransferase/metabolismo , Camundongos , Ratos
19.
J Hypertens Suppl ; 12(10): S41-5, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7769490

RESUMO

CONTROL OF CARDIOVASCULAR SYSTEM: Neuropeptide Y has three major activities which are important in the modulation of blood pressure homeostasis. When released from sympathetic neurons innervating the cardiovascular system, this peptide causes direct long-lasting vasoconstriction, inhibits the release of noradrenaline and other neurotransmitters and potentiates the action of noradrenaline and other pressor agents. RECEPTOR SUBTYPE DIVERSITY: At least two major subtypes of neuropeptide Y receptor have been defined by pharmacological criteria, and the major subtype involved in the control of blood pressure (Y1) has been isolated by molecular cloning. Analysis of the cloned DNA sequence has confirmed that the receptor is a member of the G protein-coupled receptor superfamily and when expressed in various cell lines can couple to both the inhibition of adenylate cyclase and the elevation of intracellular calcium. NEUROPEPTIDE Y ANTAGONISTS: A specific neuropeptide Y antagonist has been developed which significantly lowers the dose-dependent neuropeptide Y-induced pressor response in normal rats. The inhibition is specific for the peptide and also selective for the postsynaptic Y1 receptor-mediated vasoconstrictor activity. Administration of this specific and selective inhibitor significantly reduces resting arterial blood pressure, which remains depressed for up to 4 h in normal and spontaneously hypertensive rats. CONCLUSIONS: Inhibition of endogenous neuropeptide Y activity demonstrates that this peptide makes a significant contribution to the control of blood pressure and indicates the therapeutic potential of neuropeptide Y inhibitors as a new class of antihypertensive agent. The molecular cloning of the neuropeptide Y receptor subtype responsible for both the direct vasoconstrictor activity of the peptide and the potentiation of the action of other pressor agents represents an important advance in our understanding of the molecular basis of neuropeptide Y action and will help in the development of selective neuropeptide Y antagonists.


Assuntos
Pressão Sanguínea/fisiologia , Sistema Cardiovascular/metabolismo , Neuropeptídeo Y/fisiologia , Receptores de Neuropeptídeo Y/fisiologia , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Neuropeptídeo Y/química , Neuropeptídeo Y/metabolismo , Ratos , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Receptores de Neuropeptídeo Y/química , Receptores de Neuropeptídeo Y/metabolismo
20.
Diabetologia ; 50(8): 1732-42, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17593346

RESUMO

AIMS/HYPOTHESIS: Insulin resistance in skeletal muscle is strongly associated with lipid oversupply, but the intracellular metabolites and underlying mechanisms are unclear. We therefore sought to identify the lipid intermediates through which the common unsaturated fatty acid linoleate causes defects in IRS-1 signalling in L6 myotubes and mouse skeletal muscle. MATERIALS AND METHODS: Cells were pre-treated with 1 mmol/l linoleate for 24 h. Subsequent insulin-stimulated IRS-1 tyrosine phosphorylation and its association with the p85 subunit of phosphatidylinositol 3-kinase were determined by immunoblotting. Intracellular lipid species and protein kinase C activation were modulated by overexpression of diacylglycerol kinase epsilon, which preferentially converts unsaturated diacylglycerol into phosphatidic acid, or by inhibition of lysophosphatidic acid acyl transferase with lisofylline, which reduces phosphatidic acid synthesis. Phosphatidic acid species in linoleate-treated cells or muscle from insulin-resistant mice fed a safflower oil-based high-fat diet that was rich in linoleate were analysed by mass spectrometry. RESULTS: Linoleate pretreatment reduced IRS-1 tyrosine phosphorylation and p85 association. Overexpression of diacylglycerol kinase epsilon reversed the activation of protein kinase C isoforms by linoleate, but paradoxically further diminished IRS-1 tyrosine phosphorylation. Conversely, lisofylline treatment restored IRS-1 phosphorylation. Mass spectrometry indicated that the dilinoleoyl-phosphatidic acid content increased from undetectable levels to almost 20% of total phosphatidic acid in L6 cells and to 8% of total in the muscle of mice fed a high-fat diet. Micelles containing dilinoleoyl-phosphatidic acid specifically inhibited IRS-1 tyrosine phosphorylation and glycogen synthesis in L6 cells. CONCLUSIONS/INTERPRETATION: These data indicate that linoleate-derived phosphatidic acid is a novel lipid species that contributes independently of protein kinase C to IRS-1 signalling defects in muscle cells in response to lipid oversupply.


Assuntos
Músculo Esquelético/metabolismo , Ácidos Fosfatídicos/metabolismo , Fosfoproteínas/metabolismo , Animais , Células Cultivadas , Diacilglicerol Quinase/metabolismo , Immunoblotting , Proteínas Substratos do Receptor de Insulina , Ácido Linoleico/farmacologia , Espectrometria de Massas , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA