Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Cancer ; 109(12): 3034-41, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24178758

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Alcoholes Grasos/farmacología , Extractos Vegetales/farmacología , Tamoxifeno/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Alcoholes Grasos/administración & dosificación , Femenino , Humanos , Células MCF-7 , Transducción de Señal , Tamoxifeno/administración & dosificación
2.
Diabetologia ; 55(11): 2999-3009, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22893028

RESUMEN

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.


Asunto(s)
Muerte Celular/fisiología , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , FN-kappa B/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citocinas/farmacología , Diabetes Mellitus Tipo 1/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Inhibidores Enzimáticos/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Interferón gamma/metabolismo , Interferón gamma/farmacología , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas , Fenilbutiratos/farmacología , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tapsigargina/farmacología , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Tunicamicina/farmacología
3.
Diabetologia ; 54(7): 1766-76, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21472432

RESUMEN

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.


Asunto(s)
Citocinas/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Óxido Nítrico/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Células Secretoras de Insulina/citología , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
4.
Diabetologia ; 54(6): 1447-56, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21347625

RESUMEN

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.


Asunto(s)
Grasas de la Dieta/metabolismo , Glucosa/metabolismo , Homeostasis/fisiología , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Proteína Quinasa C-epsilon/deficiencia , Animales , Eliminación de Gen , Insulina/metabolismo , Ratones , Ratones Noqueados , Modelos Animales , Proteína Quinasa C-epsilon/genética , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
5.
Diabetologia ; 54(2): 380-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21103982

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Citocinas/farmacología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Isoenzimas/metabolismo , Proteína Quinasa C-delta/metabolismo , Animales , Apoptosis/genética , Western Blotting , Técnicas In Vitro , Células Secretoras de Insulina/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Islotes Pancreáticos/citología , Isoenzimas/genética , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Proteína Quinasa C-delta/genética , Factor de Necrosis Tumoral alfa/farmacología
6.
Diabetologia ; 52(12): 2616-20, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19809797

RESUMEN

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.


Asunto(s)
Grasas de la Dieta/efectos adversos , Intolerancia a la Glucosa/inducido químicamente , Proteína Quinasa C-delta/deficiencia , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-epsilon/deficiencia , Proteína Quinasa C-epsilon/metabolismo , Animales , Glucemia/metabolismo , Cruzamientos Genéticos , Eliminación de Gen , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Músculo Esquelético/enzimología , Proteína Quinasa C-delta/genética , Proteína Quinasa C-epsilon/genética , Triglicéridos/metabolismo
7.
Diabetologia ; 52(11): 2369-2373, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19727664

RESUMEN

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.


Asunto(s)
Retículo Endoplásmico/fisiología , Células Secretoras de Insulina/fisiología , Glicoproteínas de Membrana/metabolismo , Transporte de Proteínas/fisiología , Proteínas/metabolismo , Estrés Fisiológico/fisiología , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular Tumoral , Cicloheximida/farmacología , Genes Reporteros , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma , Ratones , Ácido Palmítico/farmacología , Transporte de Proteínas/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Transfección
8.
Diabetologia ; 50(8): 1732-42, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17593346

RESUMEN

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.


Asunto(s)
Músculo Esquelético/metabolismo , Ácidos Fosfatidicos/metabolismo , Fosfoproteínas/metabolismo , Animales , Células Cultivadas , Diacilglicerol Quinasa/metabolismo , Immunoblotting , Proteínas Sustrato del Receptor de Insulina , Ácido Linoleico/farmacología , Espectrometría de Masas , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Tirosina/metabolismo
9.
Diabetologia ; 50(4): 752-63, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17268797

RESUMEN

AIMS/HYPOTHESIS: Increased lipid supply causes beta cell death, which may contribute to reduced beta cell mass in type 2 diabetes. We investigated whether endoplasmic reticulum (ER) stress is necessary for lipid-induced apoptosis in beta cells and also whether ER stress is present in islets of an animal model of diabetes and of humans with type 2 diabetes. METHODS: Expression of genes involved in ER stress was evaluated in insulin-secreting MIN6 cells exposed to elevated lipids, in islets isolated from db/db mice and in pancreas sections of humans with type 2 diabetes. Overproduction of the ER chaperone heat shock 70 kDa protein 5 (HSPA5, previously known as immunoglobulin heavy chain binding protein [BIP]) was performed to assess whether attenuation of ER stress affected lipid-induced apoptosis. RESULTS: We demonstrated that the pro-apoptotic fatty acid palmitate triggers a comprehensive ER stress response in MIN6 cells, which was virtually absent using non-apoptotic fatty acid oleate. Time-dependent increases in mRNA levels for activating transcription factor 4 (Atf4), DNA-damage inducible transcript 3 (Ddit3, previously known as C/EBP homologous protein [Chop]) and DnaJ homologue (HSP40) C3 (Dnajc3, previously known as p58) correlated with increased apoptosis in palmitate- but not in oleate-treated MIN6 cells. Attenuation of ER stress by overproduction of HSPA5 in MIN6 cells significantly protected against lipid-induced apoptosis. In islets of db/db mice, a variety of marker genes of ER stress were also upregulated. Increased processing (activation) of X-box binding protein 1 (Xbp1) mRNA was also observed, confirming the existence of ER stress. Finally, we observed increased islet protein production of HSPA5, DDIT3, DNAJC3 and BCL2-associated X protein in human pancreas sections of type 2 diabetes subjects. CONCLUSIONS/INTERPRETATION: Our results provide evidence that ER stress occurs in type 2 diabetes and is required for aspects of the underlying beta cell failure.


Asunto(s)
Apoptosis , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Animales , Western Blotting , Línea Celular , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/fisiología , Humanos , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Chaperonas Moleculares/genética , Chaperonas Moleculares/fisiología , Estrés Oxidativo , ARN Mensajero/metabolismo
10.
Cell Death Differ ; 10(3): 269-77, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12700627

RESUMEN

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.


Asunto(s)
Apoptosis , Células Epiteliales/metabolismo , Proteína Quinasa C/fisiología , Glándulas Salivales/metabolismo , Adenoviridae/genética , Animales , Caspasa 3 , Caspasas/metabolismo , Línea Celular , Separación Celular , Células Cultivadas , ADN/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Fase G1 , Glutatión Transferasa/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Glándula Parótida/citología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteína Quinasa C-alfa , Proteína Quinasa C-delta , Ratas , Factores de Tiempo
11.
Am J Physiol Endocrinol Metab ; 282(6): E1204-13, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12006349

RESUMEN

We have previously shown that glycogen synthesis is reduced in lipid-treated C(2)C(12) skeletal muscle myotubes and that this is independent of changes in glucose uptake. Here, we tested whether mitochondrial metabolism of these lipids is necessary for this inhibition and whether the activation of specific protein kinase C (PKC) isoforms is involved. C(2)C(12) myotubes were pretreated with fatty acids and subsequently stimulated with insulin for the determination of glycogen synthesis. The carnitine palmitoyltransferase-1 inhibitor etomoxir, an inhibitor of beta-oxidation of acyl-CoA, did not protect against the inhibition of glycogen synthesis caused by the unsaturated fatty acid oleate. In addition, although oleate caused translocation, indicating activation, of individual PKC isoforms, inhibition of PKC by pharmacological agents or adenovirus-mediated overexpression of dominant negative PKC-alpha, -epsilon, or -theta mutants was unable to prevent the inhibitory effects of oleate on glycogen synthesis. We conclude that neither mitochondrial lipid metabolism nor activation of PKC-alpha, -epsilon, or -theta plays a role in the direct inhibition of glycogen synthesis by unsaturated fatty acids.


Asunto(s)
Ácidos Grasos Insaturados/farmacología , Glucógeno/biosíntesis , Isoenzimas/metabolismo , Músculos/efectos de los fármacos , Músculos/metabolismo , Proteína Quinasa C/metabolismo , Animales , Línea Celular , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Compuestos Epoxi/farmacología , Expresión Génica , Immunoblotting , Insulina/farmacología , Isoenzimas/genética , Cinética , Metabolismo de los Lípidos , Ratones , Mitocondrias/metabolismo , Músculos/ultraestructura , Mutación , Ácido Oléico/farmacología , Oxidación-Reducción , Proteína Quinasa C/genética , Proteína Quinasa C-alfa , Proteína Quinasa C-epsilon , Proteína Quinasa C-theta , Transfección
12.
Diabetes ; 50(10): 2210-8, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11574400

RESUMEN

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.


Asunto(s)
Glucógeno/biosíntesis , Isoenzimas/fisiología , Ácido Palmítico/farmacología , Fosfoproteínas Fosfatasas/fisiología , Proteína Quinasa C/fisiología , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Animales , Línea Celular , Ratones , Músculo Esquelético/metabolismo , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2 , Proteínas Proto-Oncogénicas c-akt
13.
Diabetes ; 50(10): 2237-43, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11574404

RESUMEN

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.


Asunto(s)
Glucagón/farmacología , Islotes Pancreáticos/citología , Fragmentos de Péptidos/farmacología , Proteína Quinasa C/metabolismo , Precursores de Proteínas/farmacología , Animales , Transporte Biológico/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular , Núcleo Celular/metabolismo , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Péptido 1 Similar al Glucagón , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Mutación/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos
14.
J Biol Chem ; 276(40): 37341-6, 2001 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-11489909

RESUMEN

Activation of phospholipase C (PLC) in neonatal rat cardiomyocytes (NCM) generates primarily inositol 1,4,5-trisphosphate (Ins(1,4,5)P(3)) in response to rises in intracellular Ca(2+), or inositol 1,4-bisphosphate (Ins(1,4)P(2)) in response to norepinephrine (NE) (Matkovich, S. J. and Woodcock, E. A. (2000) J. Biol. Chem. 275, 10845-10850). To examine the PLC subtype mediating the alpha(1)-adrenergic receptor response, PLC-beta(1) and PLC-beta(3) were overexpressed in NCM using adenoviral infection (Ad-PLC-beta(1) NCM and Ad-PLC-beta(3) NCM, respectively) and PLC responses assessed from [(3)H]inositol phosphate (InsP) generation in the presence of 10 mm LiCl. The [(3)H]InsP response to NE (100 microm) was enhanced in Ad-PLC-beta(1) NCM relative to cells infected with blank virus (Ad-MX NCM), but was reduced in Ad-PLC-beta(3) NCM. In contrast, the [(3)H]InsP response to ATP (100 microm) was not elevated in Ad-PLC-beta(1) NCM, and was enhanced rather than diminished in Ad-PLC-beta(3) NCM, showing that effects of the two PLC-beta isoforms were specific for particular receptor types. PLC-delta(1) overexpression selectively reduced NE-induced [(3)H]InsP responses, without affecting the ATP stimulation. The reduced NE response was associated with a selective loss of PLC-beta(1) expression in Ad-PLC-delta(1) NCM. alpha(1)-Adrenergic receptor activation caused phosphorylation of PLC-beta(1) but not PLC-beta(3), whereas stimulation by ATP induced phosphorylation of PLC-beta(3) but not PLC-beta(1.) Taken together, these studies provide evidence that NE-stimulated InsP generation in NCM is primarily mediated by PLC-beta(1), despite the presence of both PLC-beta(1) and PLC-beta(3) isoforms.


Asunto(s)
Isoenzimas/metabolismo , Miocardio/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Fosfolipasas de Tipo C/metabolismo , Animales , Animales Recién Nacidos , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Fosfatos de Inositol/metabolismo , Fosfolipasa C beta , Fosfolipasa C delta , Fosforilación , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y2
15.
J Biol Chem ; 276(32): 29719-28, 2001 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-11369761

RESUMEN

We report here that the novel protein kinase C isoform, PKCdelta, is required at or prior to the level of the mitochondria for apoptosis induced by a diverse group of cell toxins. We have used adenoviral expression of a kinase-dead (KD) mutant of PKCdelta to explore the requirement for PKCdelta in the mitochondrial-dependent apoptotic pathway. Expression of PKCdeltaKD, but not PKCalphaKD, in salivary epithelial cells resulted in a dose-dependent inhibition of apoptosis induced by etoposide, UV-irradiation, brefeldin A, and paclitaxel. DNA fragmentation was blocked up to 71% in parotid C5 cells infected with the PKCdeltaKD adenovirus, whereas caspase-3 activity was inhibited up to 65%. The activation of caspase-9-like proteases by all agents was also inhibited in parotid C5 cells expressing PKCdeltaKD. The ability of PKCdeltaKD to block the loss of mitochondrial membrane potential was similarly determined. Expression of PKCdeltaKD blocked the decrease in mitochondrial membrane potential observed in cells treated with etoposide, UV, brefeldin A, or paclitaxel in a dose-dependent manner. In contrast to the protective function of PKCdeltaKD, expression of PKCdeltaWT resulted in a potent induction of apoptosis, which could be inhibited by co-infection with PKCdeltaKD. These results suggest that PKCdelta is a common intermediate in mitochondrial-dependent apoptosis in salivary epithelial cells.


Asunto(s)
Apoptosis , Células Epiteliales/metabolismo , Isoenzimas/química , Isoenzimas/fisiología , Mitocondrias/metabolismo , Proteína Quinasa C/química , Proteína Quinasa C/fisiología , Glándulas Salivales/metabolismo , Adenoviridae/genética , Animales , Western Blotting , Brefeldino A/farmacología , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Línea Celular , Separación Celular , Fragmentación del ADN , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Etopósido/farmacología , Citometría de Flujo , Genes Dominantes , Operón Lac , Mutación , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Paclitaxel/farmacología , Isoformas de Proteínas , Proteína Quinasa C-delta , Inhibidores de la Síntesis de la Proteína/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Ratas , Factores de Tiempo , Rayos Ultravioleta
16.
FEBS Lett ; 492(1-2): 101-6, 2001 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-11248245

RESUMEN

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.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Miosinas/metabolismo , Membrana Celular/metabolismo , Humanos , Secreción de Insulina , Fosforilación , Fracciones Subcelulares
17.
J Biol Chem ; 276(22): 19072-7, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11274217

RESUMEN

In pancreatic islets the activation of phospholipase C (PLC) by the muscarinic receptor agonist carbamyolcholine (carbachol) results in the hydrolysis of both phosphatidylinositol 4,5-bisphosphate (PtdInsP(2)) and phosphatidylinositol (PtdIns). Here we tested the hypothesis that PtdIns hydrolysis is mediated by PLCgamma1, which is known to be regulated by activation of tyrosine kinases and PtdIns 3-kinase. PtdIns breakdown was more sensitive than that of PtdInsP(2) to the tyrosine kinase inhibitor, genistein. Conversely, the tyrosine phosphatase inhibitor, vanadate, alone promoted PtdIns hydrolysis and acted non-additively with carbachol. Vanadate did not stimulate PtdInsP(2) breakdown. Carbachol also stimulated a rapid (maximal at 1-2 min) tyrosine phosphorylation of several islet proteins, although not of PLCgamma1 itself. Two structurally unrelated inhibitors of PtdIns 3-kinase, wortmannin and LY294002, more effectively attenuated the hyrolysis of PtdIns compared with PtdInsP(2). Adenovirally mediated overexpression of PLCgamma1 significantly increased carbachol-stimulated PtdIns hydrolysis without affecting that of PtdInsP(2). Conversely overexpression of PLCbeta1 up-regulated the PtdInsP(2), but not PtdIns, response. These results indicate that the hydrolysis of PtdIns and PtdInsP(2) are independently regulated in pancreatic islets and that PLCgamma1 selectively mediates the breakdown of PtdIns. The activation mechanism of PLCgamma involves tyrosine phosphorylation (but not of PLCgamma directly) and PtdIns 3-kinase. Our findings point to a novel bifurcation of signaling pathways downstream of muscarinic receptors and suggest that hydrolysis of PtdIns and PtdInsP(2) might serve different physiological ends.


Asunto(s)
Carbacol/farmacología , Hidrólisis , Islotes Pancreáticos/metabolismo , Isoenzimas/metabolismo , Isoenzimas/fisiología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositoles/metabolismo , Fosfolipasas de Tipo C/metabolismo , Fosfolipasas de Tipo C/fisiología , Adenoviridae/genética , Androstadienos/farmacología , Animales , Agonistas Colinérgicos/farmacología , Cromatografía Líquida de Alta Presión , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Genisteína/farmacología , Fosfatos de Inositol/metabolismo , Potenciales de la Membrana , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfolipasa C gamma , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Ratas , Transducción de Señal , Factores de Tiempo , Tirosina/metabolismo , Vanadatos/farmacología , Wortmanina
18.
J Biol Chem ; 276(7): 5368-74, 2001 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-11087760

RESUMEN

Exposure of pancreatic islets to cytokines such as interleukin (IL)-1beta induces a variety of proinflammatory genes including type II nitric-oxide synthase (iNOS) which produces nitric oxide (NO). NO is thought to be a major cause of islet beta-cell dysfunction and apoptotic beta-cell death, which results in type I diabetes. Since protein kinase C (PKC) mediates some of the actions of cytokines in other cell types, our aim was to assess the role of PKC in IL-1beta-induced iNOS expression in pancreatic beta-cells. PKCdelta, but not PKCalpha, was specifically activated in the rat INS-1 beta-cell line by IL-1beta as assessed by membrane translocation. Moreover, iNOS expression and NO production were significantly attenuated by the PKCdelta specific inhibitor rottlerin and overexpression of a PKCdelta kinase-dead mutant protein. Conversely, overexpression of PKCdelta wild type protein significantly potentiated this response. These results were confirmed at the mRNA level by reverse transcriptase-polymerase chain reaction. However, a role at the level of transcriptional regulation appeared unlikely, since PKCdelta was not required for the activation of NF-kappaB, activating protein 1, and activating transcription factor 2 signaling pathways in response to IL-1beta. There was, however, a significant increase in iNOS mRNA stability mediated by PKCdelta wild type, while PKCdelta kinase-dead acted reciprocally, reducing iNOS mRNA stability. The results indicate that, in addition to transcriptional activation, mRNA stabilization is a key component of the mechanism by which IL-1beta stimulates iNOS expression in beta-cells and that PKCdelta plays an essential role in this process. PKCdelta activation may therefore have significant consequences with regard to cellular function and viability when beta-cells are exposed to IL-1beta and potentially other cytokines.


Asunto(s)
Interleucina-1/farmacología , Islotes Pancreáticos/enzimología , Isoenzimas/fisiología , Óxido Nítrico Sintasa/genética , Proteína Quinasa C/fisiología , Estabilidad del ARN , Animales , Línea Celular , Células Cultivadas , Activación Enzimática , Islotes Pancreáticos/efectos de los fármacos , Isoenzimas/genética , Isoenzimas/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Mutación , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo II , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteína Quinasa C-delta , Transporte de Proteínas , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
19.
Am J Respir Cell Mol Biol ; 23(4): 555-9, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11017922

RESUMEN

We hypothesized that an atypical isoform of protein kinase (PK) C, PKC-zeta, is essential for proliferation of human airway smooth muscle (HASM) cells in primary culture. Recombinant replication-deficient E1-deleted adenoviruses (100 plaque-forming units [pfu]/cell) expressing the antisense of PKC-zeta and the wild-type PKC-zeta (Ad-CMV-PKC-zeta) were added to actively growing cells that were subsequently incubated for 48 h in platelet-derived growth factor (PDGF) 40 ng/mL or 10% fetal bovine serum (FBS). Expression of the antisense at a virus concentration of 100 pfu/cell produced a significant (n = 3, P<0.05) decrease in the mean manual cell count in the presence of PDGF to 37+/-5% relative to that in cells with no virus (100%), whereas in cells infected with virus containing no construct, this figure was 102+/-13%. The increase in cell number in response to FBS, however, was not affected by the presence of the antisense. Corresponding values for cells in 10% FBS were 100+/-22%, 85+/-22%, and 122+/-18%. Western blotting revealed decreased levels of PKC-zeta protein, but not PKC-alpha or PKC-epsilon protein, in cells infected with the antisense when compared with levels in control cells. Thus, in HASM cells, PKC-zeta is involved in proliferation in response to PDGF, but not in response to FBS, for which alternate signal transduction pathways independent of PKC-zeta must exist.


Asunto(s)
División Celular/efectos de los fármacos , Isoenzimas/genética , Músculo Liso/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Proteína Quinasa C/genética , Tráquea/efectos de los fármacos , Adenoviridae/genética , Animales , Bovinos , Línea Celular , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Músculo Liso/citología , Tráquea/citología , Regulación hacia Arriba
20.
Am J Physiol Endocrinol Metab ; 279(5): E1196-201, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11052977

RESUMEN

Muscle insulin resistance in the chronic high-fat-fed rat is associated with increased membrane translocation and activation of the novel, lipid-responsive, protein kinase C (nPKC) isozymes PKC-theta and -epsilon. Surprisingly, fat-induced insulin resistance can be readily reversed by one high-glucose low-fat meal, but the underlying mechanism is unclear. Here, we have used this model to determine whether changes in the translocation of PKC-theta and -epsilon are associated with the acute reversal of insulin resistance. We measured cytosol and particulate PKC-alpha and nPKC-theta and -epsilon in muscle in control chow-fed Wistar rats (C) and 3-wk high-fat-fed rats with (HF-G) or without (HF-F) a single high-glucose meal. PKC-theta and -epsilon were translocated to the membrane in muscle of insulin-resistant HF-F rats. However, only membrane PKC-theta was reduced to the level of chow-fed controls when insulin resistance was reversed in HF-G rats [% PKC-theta at membrane, 23.0 +/- 4.4% (C); 39.7 +/- 3.4% (HF-F, P < 0.01 vs. C); 22.5 +/- 2.7% (HF-G, P < 0.01 vs. HF-F), by ANOVA]. We conclude that, although muscle localization of both PKC-epsilon and PKC-theta are influenced by chronic dietary lipid oversupply, PKC-epsilon and PKC-theta localization are differentially influenced by acute withdrawal of dietary lipid. These results provide further support for an association between PKC-theta muscle cellular localization and lipid-induced muscle insulin resistance and stress the labile nature of high-fat diet-induced insulin resistance in the rat.


Asunto(s)
Grasas de la Dieta/farmacología , Resistencia a la Insulina , Isoenzimas/análisis , Músculo Esquelético/enzimología , Proteína Quinasa C/análisis , Tejido Adiposo , Animales , Glucemia/metabolismo , Composición Corporal , Citosol/enzimología , Carbohidratos de la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Glucosa/administración & dosificación , Técnica de Clampeo de la Glucosa , Insulina/sangre , Masculino , Fibras Musculares de Contracción Rápida/efectos de los fármacos , Fibras Musculares de Contracción Rápida/enzimología , Fibras Musculares de Contracción Rápida/ultraestructura , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/ultraestructura , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...