Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 34(27): 3504-13, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25220417

RESUMEN

Obesity and inflammation are both risk factors for a variety of cancers, including breast cancer in postmenopausal women. Intake of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) decreases the risk of breast cancer, and also reduces obesity-associated inflammation and insulin resistance, but whether the two effects are related is currently unknown. We tested this hypothesis in a postmenopausal breast cancer model using ovariectomized, immune-competent female mice orthotopically injected with Py230 mammary tumor cells. Obesity, whether triggered genetically or by high-fat diet (HFD) feeding, increased inflammation in the mammary fat pad and promoted mammary tumorigenesis. The presence of tumor cells in the mammary fat pad further enhanced the local inflammatory milieu. Tumor necrosis factor-alpha (TNF-α) was the most highly upregulated cytokine in the obese mammary fat pad, and we observed that TNF-α dose-dependently stimulated Py230 cell growth in vitro. An ω-3 PUFA-enriched HFD (referred to as fish oil diet, FOD) reduced inflammation in the obese mammary fat pad in the absence of tumor cells and inhibited Py230 tumor growth in vivo. Although some anti-inflammatory effects of ω-3 PUFAs were previously shown to be mediated by the G-protein-coupled receptor 120 (GPR120), the FOD reduced Py230 tumor burden in GPR120-deficient mice to a similar degree as observed in wild-type mice, indicating that the effect of FOD to reduce tumor growth does not require GPR120 in the host mouse. Instead, in vitro studies demonstrated that ω-3 PUFAs act directly on tumor cells to activate c-Jun N-terminal kinase, inhibit proliferation and induce apoptosis. Our results show that obesity promotes mammary tumor progression in this model of postmenopausal breast cancer and that ω-3 PUFAs, independent of GPR120, inhibit mammary tumor progression in obese mice.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Neoplasias Mamarias Experimentales/complicaciones , Neoplasias Mamarias Experimentales/patología , Obesidad/complicaciones , Receptores Acoplados a Proteínas G/fisiología , Animales , Células Cultivadas , Dieta Alta en Grasa , Progresión de la Enfermedad , Femenino , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Ovariectomía , Posmenopausia/fisiología
2.
Biochemistry (Mosc) ; 76(11): 1262-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22117553

RESUMEN

Insulin regulates glucose uptake into fat and skeletal muscle cells by modulating the translocation of GLUT4 between the cell surface and interior. We investigated a role for cortactin, a cortical actin binding protein, in the actin filament organization and translocation of GLUT4 in Chinese hamster ovary (CHO-GLUT4myc) and L6-GLUT4myc myotube cells. Overexpression of wild-type cortactin enhanced insulin-stimulated GLUT4myc translocation but did not alter actin fiber formation. Conversely, cortactin mutants lacking the Src homology 3 (SH3) domain inhibited insulin-stimulated formation of actin stress fibers and GLUT4 translocation similar to the actin depolymerizing agent cytochalasin D. Wortmannin, genistein, and a PP1 analog completely blocked insulin-induced Akt phosphorylation, formation of actin stress fibers, and GLUT4 translocation indicating the involvement of both PI3-K/Akt and the Src family of kinases. The effect of these inhibitors was even more pronounced in the presence of overexpressed cortactin suggesting that the same pathways are involved. Knockdown of cortactin by siRNA did not inhibit insulin-induced Akt phosphorylation but completely inhibited actin stress fiber formation and glucose uptake. These results suggest that the actin binding protein cortactin is required for actin stress fiber formation in muscle cells and that this process is absolutely required for translocation of GLUT4-containing vesicles to the plasma membrane.


Asunto(s)
Actinas/metabolismo , Cortactina/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fibras de Estrés/metabolismo , Citoesqueleto de Actina/metabolismo , Androstadienos/farmacología , Animales , Células CHO , Membrana Celular/metabolismo , Cortactina/genética , Cricetinae , Citocalasina D/farmacología , Técnicas de Silenciamiento del Gen , Transportador de Glucosa de Tipo 4/genética , Humanos , Proteínas de Microfilamentos/genética , Fibras Musculares Esqueléticas/citología , Fosforilación , Transporte de Proteínas , ARN Interferente Pequeño/genética , Transducción de Señal , Wortmanina , Familia-src Quinasas/metabolismo
3.
Neurobiol Aging ; 27(3): 439-45, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16464656

RESUMEN

Prolonged hypoxia, encountered in individuals suffering from various cardiorespiratory diseases, enhances the likelihood of subsequently developing Alzheimer's disease (AD). However, the underlying mechanisms are unknown, as are the mechanisms of neurodegeneration of amyloid beta peptides (AbetaPs), although the latter involves disruption of Ca2+ homeostasis. Here, immunohistochemistry demonstrated that hypoxia increased production of AbetaPs, an effect which was prevented by inhibition of either beta or gamma secretase, the enzymes required for liberation of AbetaP from its precursor protein. Whole-cell patch clamp recordings showed that hypoxia selectively increased functional expression of L-type Ca2+ channels. This was prevented by inhibition of either beta or gamma secretase, indicating that hypoxic channel up-regulation is dependent upon AbetaP formation. Our results indicate for the first time that hypoxia promotes AbetaP formation in central neurons, and show that this leads to abnormally high selective expression of L-type Ca2+ channels whose blockade has previously been shown to be neuroprotective in AD models. These findings provide a cellular basis for understanding the increased incidence of AD following prolonged hypoxia.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Canales de Calcio Tipo L/metabolismo , Cerebelo/metabolismo , Potenciales de la Membrana/fisiología , Neuronas/metabolismo , Oxígeno/metabolismo , Animales , Hipoxia de la Célula/fisiología , Células Cultivadas , Ratas
4.
J Neurochem ; 79(3): 699-712, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11701773

RESUMEN

The effects of amyloid beta protein on voltage-gated K(+) channel currents were studied using the whole-cell patch-clamp technique. The 1-40 amino acid form of amyloid beta protein was applied to primary cultures of rat cerebellar granule and cortical neurones for 24 h. Both the unaggregated and aggregated forms of the peptide, which have differing biological activities, were used. In cerebellar granule neurones, 24-h pre-incubation with 1 microM unaggregated amyloid beta protein resulted in a 60% increase in the 'A'-type component of K(+) current. Increased delayed rectifier activity was Cd(2+)-sensitive and was presumed to be secondary to an increase in voltage-gated Ca(2+) channel current activity. Unaggregated amyloid beta protein had no effect on any component of the K(+) channel current in cortical neurones. One micromolar of aggregated amyloid beta protein had no effect on K(+) channel current in either cell type but reduced cell survival within 24 h as measured using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) assays. The unaggregated form of amyloid beta protein had no neurotoxic effects when applied to either neurone type for up to 72 h. These data indicate that the unaggregated, non-pathological form of amyloid beta protein causes changes in the ion channel function of neurones, possibly reflecting a physiological role for the peptide.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Activación del Canal Iónico/efectos de los fármacos , Neuronas/fisiología , Fragmentos de Péptidos/toxicidad , Canales de Potasio/fisiología , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/química , Animales , Cloruro de Cadmio/farmacología , Células Cultivadas , Cerebelo/citología , Corteza Cerebral/citología , Potenciales de la Membrana/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Fragmentos de Péptidos/química , Potasio/metabolismo , Ratas , Solubilidad
5.
Biochem Biophys Res Commun ; 282(2): 615-20, 2001 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-11401505

RESUMEN

Previously we have shown that the insulin receptor and phospholipase C-gamma1 physically interact in the 3T3-L1 adipocyte cell line. In this study, we investigated the ability of insulin and PDGF to stimulate PLC-gamma1 enzyme activity as measured by PI-(4,5)P(2) hydrolysis. Both insulin and PDGF caused a rapid (<1 min) increase in PLC activity associated with the respective receptor. PDGF treatment resulted in a higher and more sustained stimulation of PLC-gamma1 activity compared to insulin (0.95 pmol/min/mg vs 0.68 pmol/min/mg). Furthermore, insulin and PDGF promoted increases in total cellular DAG, one of the products of PI-(4,5)P(2) hydrolysis. Insulin-stimulated PLC activity appears to be downstream of PI-3Kinase as the DAG increase was partially blocked by Wortmannin and addition of PI-(3,4,5)P(3) activated PLC-gamma1 in vitro. Inhibition of PLC using U73122 or an inhibitory peptide caused a decrease in insulin-stimulated 2-deoxyglucose transport and GLUT4 translocation that was rescued by the addition of OAG, a cell-permeable synthetic DAG.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipocitos/enzimología , Insulina/farmacología , Isoenzimas/metabolismo , Proteínas Musculares , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfolipasas de Tipo C/metabolismo , Células 3T3 , Adipocitos/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Diglicéridos/biosíntesis , Diglicéridos/farmacología , Activación Enzimática/efectos de los fármacos , Estrenos/farmacología , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1 , Transportador de Glucosa de Tipo 4 , Isoenzimas/antagonistas & inhibidores , Ratones , Proteínas de Transporte de Monosacáridos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipasa C gamma , Factor de Crecimiento Derivado de Plaquetas/farmacología , Pirrolidinonas/farmacología , Fosfolipasas de Tipo C/antagonistas & inhibidores
6.
Brain Res Mol Brain Res ; 89(1-2): 50-7, 2001 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-11311975

RESUMEN

The effects of chronic hypoxia (2.5% O(2), 24 h) on [3H]noradrenaline ([3H]NA) release evoked from human neuroblastoma SH-SY5Y cells by depolarisation and by activation of muscarinic receptors was investigated. Depolarization of cells with 100 mM K(+) evoked [3H]NA release, and chronic hypoxia enhanced this release significantly. In fluorimetric studies, the K(+)-evoked rises of [Ca(2+)](i) observed in response to 100 mM K(+) were also significantly enhanced. Muscarine-evoked [3H]NA release was also dramatically enhanced by chronic hypoxia. However, muscarine-induced release of Ca(2+) from intracellular stores and subsequent capacitative Ca(2+) entry was unaffected. The protein kinase C inhibitors GF 109 203X and RO-31-8220 did not prevent the enhancement of muscarine-evoked release caused by chronic hypoxia. These findings indicate that chronic hypoxia increases release of [3H]NA from human neuroblastoma SH-SY5Y cells. Enhancement of K(+)-evoked release was attributable to an enhancement of depolarisation-mediated Ca(2+) influx. In contrast, the larger enhancement of muscarine-evoked [3H]NA release was not due to greater release of Ca(2+) from internal stores, nor due to enhanced Ca(2+) influx. Furthermore, it was not attributable to activation of protein kinase C. These findings suggest that enhancement of sympathetic output, known to occur following prolonged hypoxia, may be mediated in part by enhancement of exocytosis.


Asunto(s)
Hipoxia Encefálica/metabolismo , Neuronas/metabolismo , Norepinefrina/farmacocinética , Calcio/metabolismo , Hipoxia de la Célula/fisiología , Enfermedad Crónica , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Humanos , Indoles/farmacología , Maleimidas/farmacología , Potenciales de la Membrana/fisiología , Muscarina/farmacología , Agonistas Muscarínicos/farmacología , Neuroblastoma , Neuronas/citología , Potasio/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Receptores Muscarínicos/metabolismo , Estimulación Química , Tritio , Células Tumorales Cultivadas
7.
Brain Res Mol Brain Res ; 87(1): 81-91, 2001 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-11223162

RESUMEN

The aim of this study was to obtain further understanding of the mechanism by which activation of muscarinic M(1) receptors inhibits K(+)-evoked noradrenaline (NA) release in the human neuroblastoma SH-SY5Y. Previous studies have found that muscarinic M(1) and M(3) receptors couple to the activation of phospholipase C in SH-SY5Y cells leading to an increase in (a) intracellular calcium ([Ca(2+)](i)) and (b) activation of protein kinase C (PKC). This study used specific inhibitors of PKC and conditions which deplete Ca(2+)(i) stores to examine the role of protein kinase C and changes in [Ca(2+)](i) in mediating the inhibition of K(+)-evoked NA release by muscarine. Our data show that pretreatment of SH-SY5Y cell layers with bisindolylmaleimide I (BIM-I) (i) failed to reverse inhibition of K(+)-evoked NA release by muscarine but (ii) did overcome the attenuation of muscarine inhibition following pretreatment with TPA. Furthermore pretreating cell layers with Ca(2+)-free Hepes buffered saline in the presence of thapsigargin, conditions which prevented muscarine induced increases in [Ca(2+)](i), failed to prevent inhibition of K(+)-evoked NA release by muscarine. The effect of muscarine on K(+)-evoked uptake of Ca(2+)(e) was examined in SH-SY5Y cells loaded with Fura-2. Muscarine inhibited Ca(2+)(e)-uptake by decreasing the rate at which Ca(2+) entered SH-SY5Y cells via voltage sensitive Ca(2+)-channels. Thus this study shows that muscarine inhibits depolarisation-evoked NA release by a mechanism which is not dependent on activation of PKC or release of Ca(2+) from internal stores.


Asunto(s)
Calcio/metabolismo , Neuronas/metabolismo , Norepinefrina/farmacocinética , Receptores Muscarínicos/metabolismo , Venenos Elapídicos/farmacología , Estrenos/farmacología , Humanos , Potenciales de la Membrana/fisiología , Muscarina/farmacología , Agonistas Muscarínicos/farmacología , Neuroblastoma , Forboles/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Potasio/farmacología , Proteína Quinasa C/metabolismo , Pirrolidinonas/farmacología , Receptor Muscarínico M1 , Sistemas de Mensajero Secundario/efectos de los fármacos , Sistemas de Mensajero Secundario/fisiología , Estimulación Química , Tritio , Células Tumorales Cultivadas
8.
Diabetes ; 49(11): 1783-93, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11078444

RESUMEN

Osmotic shock and insulin stimulate GLUT4 translocation and glucose transport via mechanisms that are for the most part distinct yet convergent. In this article, we investigated the effect of osmotic shock and insulin on the activation of the mitogen-activated protein kinase (MAPK) cascades in differentiated 3T3-L1 adipocytes. The MAPKs are activated by phosphorylation on conserved tyrosine and threonine residues. Both sorbitol and insulin strongly stimulated extracellular regulated kinase (ERK) 1 and 2 phosphorylation (8- and 18-fold, respectively). In contrast, c-jun-NH2-terminal kinase (JNK)/stress-activated protein kinase (SAPK) phosphorylation was stimulated only by sorbitol (sevenfold) and not by insulin. Phosphorylation of p38 MAPK was stimulated strongly by sorbitol (22-fold) but weakly by insulin (2.7-fold). Measurement of intrinsic JNK and p38 MAPK activity confirmed the phosphorylation studies. JNK and p38 MAPK were activated only significantly by sorbitol. The MAPKs are phosphorylated by dual-specificity kinases (mitogen-activated ERK-activating kinase [MEK] or MAPK kinase [MKK]). As expected, sorbitol and insulin both stimulated MEK phosphorylation. MKK4 was phosphorylated only in response to sorbitol, and neither of the stimuli caused phosphorylation of MKK3 or 6. To determine the functional significance of the observed activation of p38 MAPK in response to insulin and osmotic shock, we used three pyridinyl imidazole p38 MAPK inhibitors, SB203580, SB202190, and PD169316. Insulin and osmotic shock-stimulated glucose transport was not inhibited by any inhibitor at concentrations that were shown to block p38 MAPK activity. Furthermore, activation of the p38 MAPK pathway by treatment of cells with anisomycin did not stimulate glucose transport. These results suggest that activation of the p38 MAPK pathway is not involved in the stimulation of glucose transport.


Asunto(s)
Adipocitos/metabolismo , Glucosa/metabolismo , Insulina/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Presión Osmótica , Células 3T3 , Animales , Transporte Biológico , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Péptidos y Proteínas de Señalización Intracelular , Cinética , MAP Quinasa Quinasa 4 , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Fosfoserina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Piridinas/farmacología , Sorbitol/farmacología , Fosfolipasas de Tipo C/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos
9.
Metabolism ; 49(8): 962-8, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10954011

RESUMEN

Glycogen synthase (GS) is the rate-limiting enzyme controlling nonoxidative glucose disposal in skeletal muscle. A reduction in GS activity and an impaired insulin responsiveness are characteristic features of skeletal muscle in type 2 diabetes. These properties also exist in human skeletal muscle cell cultures from type 2 diabetic subjects. To determine the effect of an isolated reduction in GS on skeletal muscle insulin action, cultures from nondiabetic subjects were treated with antisense oligonucleotides (ODNs) to GS to interfere with expression of the gene. Treatment with antisense ODNs reduced GS protein expression by 70% compared with control (scrambled) ODNs (P < .01). GS activity measured at 0.01 mmol/L glucose-6-phosphate (G-6-P) was reduced by antisense ODN treatment. The insulin responsiveness of GS was impaired. Insulin also failed to stimulate glucose incorporation into glycogen after antisense ODN treatment. The cellular glycogen content was lower in antisense ODN-treated cells compared with control ODN. The insulin responsiveness of glucose uptake was abolished by antisense ODN treatment. Thus, reductions in GS expression in human skeletal muscle cells lead to impairments in insulin responsiveness and may play an important role in insulin-resistant states.


Asunto(s)
Silenciador del Gen , Glucógeno Sintasa/biosíntesis , Resistencia a la Insulina/fisiología , Proteínas Musculares , Músculo Esquelético/fisiología , Transportadoras de Casetes de Unión a ATP/biosíntesis , Adulto , Sistema de Transporte de Aminoácidos X-AG , Técnicas de Cultivo , Regulación hacia Abajo/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica , Glucosa/farmacocinética , Transportador de Glucosa de Tipo 4 , Glucógeno/biosíntesis , Glucógeno/metabolismo , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Humanos , Insulina/sangre , Resistencia a la Insulina/genética , Proteínas de Transporte de Monosacáridos/biosíntesis , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Oligorribonucleótidos Antisentido/genética , Oligorribonucleótidos Antisentido/farmacología , Fosfatidiletanolaminas/farmacología , ARN Mensajero/genética
10.
Oncogene ; 18(35): 4908-19, 1999 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-10490825

RESUMEN

Nerve growth factor (NGF) treatment of Chinese hamster ovary fibroblast (CHO) cells exogenously expressing 2.5x105 TrkA receptors (CHO/TrkA) results in inhibition of serum and insulin-like growth factor-I (IGF-I) stimulated cell proliferation in a dose-dependent manner. Furthermore, NGF does not stimulate [3H]thymidine incorporation and inhibits IGF-I mediated DNA synthesis in CHO/TrkA cells. NGF and IGF-I induce extracellular-signal regulated kinase 1 (ERK1) and ERK2 activation, but NGF is able to stimulate a higher and more sustained activation of these enzymes compared with IGF-I. Cotreatment with NGF and IGF-I yields an ERK1/2 activity profile similar to that of NGF treatment alone. While pretreatment with mitogen activated protein kinase kinase (MKK) inhibitor PD98059 (30 microM) results in 100% inhibition of IGF-I stimulated MAPK phosphorylation (IC50<1 microM), NGF mediated MAPK phosphorylation is only decreased by 50% (IC50=3 microM). NGF, but not IGF-I, stimulates tyrosine phosphorylation and activation of PLC-gamma1 which can be inhibited in a dose-dependent manner by phosphoinositide-specific phospholipase C (PI-PLC) inhibitor U73122 (IC50=4 microM). Pretreatment with U73122 (IC50=7 microM) results in an 87% inhibition of NGF mediated MAPK phosphorylation, while cotreatment with PD98059 and U73122 results in 97% inhibition. U73122 pretreatment has no effect on NGF stimulated Akt activation. NGF, but not IGF-I, stimulates the tyrosine phosphorylation of Suc1-associated neurotrophic factor-induced tyrosine phosphorylation target (SNT-1)/fibroblast growth factor receptor substrate 2 (FRS2) which can be completely prevented by pretreatment with 10 microM U73122. Finally, inhibition of PI-PLC results in NGF's ability to stimulate DNA synthesis in the absence and presence of IGF-I.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/farmacología , Isoenzimas/antagonistas & inhibidores , Mitógenos/farmacología , Factores de Crecimiento Nervioso/farmacología , Proteínas Serina-Treonina Quinasas , Transducción de Señal/efectos de los fármacos , Fosfolipasas de Tipo C/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Células CHO , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , División Celular/efectos de los fármacos , Cricetinae , ADN/biosíntesis , Activación Enzimática/efectos de los fármacos , Humanos , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Isoenzimas/metabolismo , Proteínas de la Membrana/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Fosfatidilinositol Diacilglicerol-Liasa , Fosfoinositido Fosfolipasa C , Fosfolipasa C gamma , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor trkA , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Fosfolipasas de Tipo C/metabolismo
12.
J Biol Chem ; 273(22): 13808-18, 1998 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-9593725

RESUMEN

Phospholipase C-gamma (PLCgamma) is the isozyme of PLC phosphorylated by multiple tyrosine kinases including epidermal growth factor, platelet-derived growth factor, nerve growth factor receptors, and nonreceptor tyrosine kinases. In this paper, we present evidence for the association of the insulin receptor (IR) with PLCgamma. Precipitation of the IR with glutathione S-transferase fusion proteins derived from PLCgamma and coimmunoprecipitation of the IR and PLCgamma were observed in 3T3-L1 adipocytes. To determine the functional significance of the interaction of PLCgamma and the IR, we used a specific inhibitor of PLC, U73122, or microinjection of SH2 domain glutathione S-transferase fusion proteins derived from PLCgamma to block insulin-stimulated GLUT4 translocation. We demonstrate inhibition of 2-deoxyglucose uptake in isolated primary rat adipocytes and 3T3-L1 adipocytes pretreated with U73122. Antilipolytic effect of insulin in 3T3-L1 adipocytes is unaffected by U73122. U73122 selectively inhibits mitogen-activated protein kinase, leaving the Akt and p70 S6 kinase pathways unperturbed. We conclude that PLCgamma is an active participant in metabolic and perhaps mitogenic signaling by the insulin receptor in 3T3-L1 adipocytes.


Asunto(s)
Adipocitos/metabolismo , Insulina/metabolismo , Isoenzimas/metabolismo , Proteínas Musculares , Receptor de Insulina/metabolismo , Transducción de Señal , Fosfolipasas de Tipo C/metabolismo , Células 3T3 , Adipocitos/enzimología , Animales , Desoxiglucosa/metabolismo , Inhibidores Enzimáticos/farmacología , Estrenos/farmacología , Transportador de Glucosa de Tipo 4 , Isoenzimas/antagonistas & inhibidores , Lipólisis , Masculino , Ratones , Microinyecciones , Proteínas de Transporte de Monosacáridos/metabolismo , Fosfolipasa C gamma , Pirrolidinonas/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/metabolismo , Fosfolipasas de Tipo C/antagonistas & inhibidores , Dominios Homologos src
13.
J Biol Chem ; 273(17): 10331-7, 1998 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-9553088

RESUMEN

The insulin receptor exists as two isoforms, A and B, that result from alternative splicing of exon 11 in the primary transcript. We have shown previously that the alternative splicing is developmentally and hormonally regulated. Consequently, these studies were instigated to identify sequences within the primary RNA transcript that regulate the alternative splicing. Minigenes containing exons 10, 11, and 12 and the intervening introns were constructed and transfected into HepG2 cells, which contain both isoforms of the insulin receptor. The cells were able to splice the minigene transcript to give both A (- exon 11) and B-like (+ exon 11) RNAs. A series of internal deletions within intron 10 were tested for their ability to give A and B RNAs. Intron 10 contained two sequences that modulated exon 11 inclusion; a 48-nucleotide purine-rich sequence at the 5' end of intron 10 that functions as a splicing enhancer and causes an increase in exon 11 inclusion, and a 43-nucleotide sequence at the 3' end of intron 10 upstream of the branch point sequence that favors skipping of exon 11. Increasing the length of the polypyrimidine tract at the 3' end of intron 10 caused exon 11 to be spliced constitutively, indicating that a weak splice site is required for alternative splicing. Finally, point mutations, insertions, and deletions within exon 11 itself were able to regulate inclusion of the exon both positively and negatively.


Asunto(s)
Empalme Alternativo , Exones , Intrones , Precursores del ARN/genética , ARN Mensajero/genética , Receptor de Insulina/genética , Secuencia de Bases , Línea Celular , Elementos de Facilitación Genéticos , Humanos , Datos de Secuencia Molecular , Mutación , Eliminación de Secuencia
14.
Cancer Res ; 58(6): 1159-64, 1998 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-9515800

RESUMEN

Insulin-like growth factor I action has been implicated in the pathogenesis of many different malignancies, including breast cancer. Insulin-like growth factor I receptors (IGF-IRs) are overexpressed in virtually all breast cancer cell lines, in which they are believed to enhance growth and inhibit apoptosis. In this study, the functional activity of IGF-IRs from normal and malignant human breast tissue was assessed. IGF-IR expression was 14-fold higher in malignant breast tissue than in normal breast tissue. IGF-IR autophosphorylation and kinase activity were 2-4-fold higher in purified receptor preparations from malignant breast tissue as compared to normal breast tissue when normalized for receptor number. This increase in receptor function, coupled with the enhanced receptor expression, amounts to a 40-fold elevation in IGF-IR tyrosine kinase activity in malignant breast tissue. The enhanced receptor autophosphorylation and kinase activity were observed in the absence of hormonal stimulation and seem to result from an alteration in the intrinsic activity of the receptor itself. Protein tyrosine phosphatase activity is also increased in malignant breast tissue. These data suggest that the IGF-IR is an important target for breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/enzimología , Mama/enzimología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Somatomedina/metabolismo , Humanos , Ligandos , Fosforilación , Fosfotirosina/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Receptor de Insulina/metabolismo
15.
Biochem Mol Med ; 62(2): 139-50, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9441865

RESUMEN

The Wilms tumor gene WT1 has been implicated in the early development of the kidney. Mutations in WT1 are found in a small fraction of Wilms tumor, a pediatric nephroblastoma, and Denys-Drash syndrome, characterized by genitourinary abnormalities. The WT1 gene product functions as a transcriptional repressor of growth factor-related genes. The kidney is one of the major sites of insulin action in vivo and expresses high levels of insulin receptors (IR). IR expression has been detected during early embryogenesis, suggesting that it may play a role in development. We investigated whether two WT1 splice variants lacking or including a three-amino-acid (KTS) insertion between the third and fourth zinc finger in the DNA-binding domain could repress the IR promoter in vitro. We show that the +KTS variant effectively represses promoter activity under all conditions tested but the -KTS variant was only able to repress in the presence of cotransfected C/EBP beta or a dominant-negative p53 mutation. Deletional mapping indicated that distinct regions of the IR promoter mediated the effects of the two isoforms and DNaseI footprint analysis identified potential WT1 binding sites within these regions.


Asunto(s)
Empalme Alternativo/genética , Proteínas de Unión al ADN/genética , Genes del Tumor de Wilms/fisiología , Regiones Promotoras Genéticas , Receptor de Insulina/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Carcinoma Hepatocelular , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/fisiología , Células HeLa , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Pruebas de Precipitina , Ratas , Eliminación de Secuencia , Factores de Transcripción/biosíntesis , Factores de Transcripción/fisiología , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/fisiología , Proteínas WT1 , Tumor de Wilms/genética , Dedos de Zinc/genética
16.
Cancer Res ; 56(12): 2781-8, 1996 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8665514

RESUMEN

There is strong evidence to suggest that insulin and insulin-like growth factor (IGF)-I may be important for tumor growth. Both the insulin and IGF-I receptors (IGF-IR) are overexpressed in breast cancer, and antibody blockade of the IGF-IR inhibits the growth of some breast cancer cell lines. Furthermore, expression of an insulin receptor (IR) in a normal mammary epithelia] cell line causes insulin-dependent transformation. Functional inactivation of p53 is also very frequent in many tumors. In this paper, we investigated whether inactivation of p53 might be involved in the overexpression of the IR in malignancy, specifically breast cancer. We demonstrate a positive correlation between IR and IGF-IR levels and p53 overexpression in primary human breast malignancies. To examine possible mechanisms by which p53 may regulate IR gene expression, we show that p53 can repress the IR promoter and that a dominant-negative p53 (248Q) can de-repress the promoter in cells containing normal p53. The p53 effect was shown to be mediated by C/EBP and Sp1 transcription factors. We also documented that p53-null mice had elevated levels of Sp1, but not C/EBPalpha, and that insulin binding to liver extracts was increased compared to wild-type controls. These results suggest that p53 inactivation may lead to an up-regulation of genes, such as the IR, that are dependent on these transcription factors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Factor de Transcripción Sp1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Secuencia de Bases , Unión Competitiva , Proteínas Potenciadoras de Unión a CCAAT , Femenino , Genes Reporteros , Humanos , Ratones , Datos de Secuencia Molecular , Receptor de Insulina/genética , Transfección , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/fisiología
17.
J Clin Endocrinol Metab ; 81(4): 1552-6, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8636366

RESUMEN

There are two insulin receptor (IR) isoforms (designated type A and type B), derived from alternative splicing of exon 11 of the IR gene. Recently, we reported (Huang Z., Bodkin N.L., Ortmeyer H.K., Hansen B.C., Shuldiner A. R., 1994, J Clin Invest, 94:1289-1296) that an increase in the exon 11- (i.e. lacking exon 11) (type A) IR messenger RNA (mRNA) variant in muscle is associated with hyperinsulinemia, an early risk factor for noninsulin-dependent diabetes mellitus (NIDDM), in the spontaneously obese, diabetic rhesus monkey. To explore further the role of IR mRNA splicing in insulin resistance of NIDDM, we studied liver, another target organ that is resistant to insulin action in NIDDM. The relative amounts of the two IR mRNA-splicing variants in liver were quantitated by RT-PCR in normal, prediabetic, and diabetic (NIDDM) monkeys. The percentage of the exon 11- mRNA variant in liver (n = 24) was significantly correlated with fasting plasma glucose (r = 0.55, P < 0.01) and intravenous glucose disappearance rate (r = -0.45, P < 0.05). The exon 11- mRNA variant was increased significantly from 29.8 +/- 1.6% in monkeys with normal fasting glucose to 39.2 +/- 2.9% in monkeys with elevated fasting glucose (P < 0.01). These studies provide the first direct evidence in vivo that the relative expression of the two IR mRNA-splicing variants is altered in liver and suggest that increased expression of the exon 11- IR isoform may contribute to hepatic insulin resistance and NIDDM or may compensate for some yet unidentified defect.


Asunto(s)
Empalme Alternativo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus/genética , Intolerancia a la Glucosa/genética , Hígado/metabolismo , Obesidad , Estado Prediabético/genética , ARN Mensajero/metabolismo , Receptor de Insulina/genética , Animales , Glucemia/metabolismo , Diabetes Mellitus/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Exones , Variación Genética , Intolerancia a la Glucosa/metabolismo , Humanos , Resistencia a la Insulina , Macaca mulatta , Estado Prediabético/metabolismo , Receptor de Insulina/biosíntesis , Receptor de Insulina/metabolismo , Especificidad de la Especie
18.
J Nutr ; 125(10): 2457-62, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7562079

RESUMEN

Starvation and refeeding affect glycogen metabolism. The effects of starvation and refeeding on the level of glycogen synthase (GS) gene expression were examined in rat liver. Depletion of hepatic glycogen stores by 72 h of starvation (7% of control) was supercompensated by 24 h of refeeding a standard laboratory diet (247% of control). Upon further refeeding, glycogen concentration gradually returned to control levels after 120 h. After 72 h of starvation, GS activity and immunoreactive protein in the liver were 60-64% lower than in control rats with free access to food. After 72 h of refeeding, GS activity and immunoreactive protein returned to control values. No significant differences in GS mRNA levels were found between fed, starved and refed rats, as determined by Northern blot analysis and PCR quantification, indicating that the long-term regulation of GS gene expression in starvation and refeeding occurs via a posttranscriptional mechanism. The amount of GS mRNA associated with polyribosomes was 90% lower in starved than in fed rats. These data indicate that the efficiency of GS mRNA translation, rather than its abundance, decreases during starvation.


Asunto(s)
Ingestión de Alimentos/fisiología , Glucógeno Sintasa/genética , Hígado/enzimología , Procesamiento Postranscripcional del ARN , Inanición/fisiopatología , Animales , Secuencia de Bases , Western Blotting , Regulación Enzimológica de la Expresión Génica , Glucógeno Sintasa/análisis , Masculino , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Ribosomas/química
19.
J Biol Chem ; 270(35): 20816-23, 1995 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-7657666

RESUMEN

We have demonstrated previously that dexamethasone treatment of HepG2 cells caused an enhancement of insulin's metabolic effects (Kosaki, A., and Webster, N. J. (1993) J. Biol. Chem. 268, 21990-21996). This correlated with increased expression of the mRNA encoding the B isoform of the insulin receptor (IR). In the present study, we have demonstrated that dexamethasone treatment caused in addition an enhancement in insulin-stimulated immediate-early gene expression (c-fos and egr-1). Dexamethasone treatment caused an increase in in vivo IR autophosphorylation and insulin receptor substrate-1 (IRS-1) phosphorylation both early events in the insulin signaling pathway. Furthermore, the IRS-1 phosphorylation was distinctly left shifted, although the level of IRS-1 protein was unchanged. Total cellular tyrosine phosphatase activity was unaltered when assayed with 32P-labeled IR and IRS-1. Studies in vitro on wheat-germ agglutinin-purified receptors showed that the B isoform of the IR had increased kinase activity both toward itself and the exogenous substrates poly.glu4:tyr1 and recombinant IRS-1 protein. In addition, two-dimensional tryptic phosphopeptide maps indicated that the B isoform has an additional phosphopeptide that is not seen for the A isoform. In conclusion, it appears that the B isoform of the IR signals more efficiently than the A isoform in HepG2 cells.


Asunto(s)
Dexametasona/farmacología , Expresión Génica/efectos de los fármacos , Genes Inmediatos-Precoces/efectos de los fármacos , Proteínas Inmediatas-Precoces , Insulina/farmacología , Receptor de Insulina/fisiología , Transducción de Señal , Secuencia de Bases , Cartilla de ADN , Proteínas de Unión al ADN/biosíntesis , Proteína 1 de la Respuesta de Crecimiento Precoz , Genes fos , Hepatoblastoma , Humanos , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina , Cinética , Neoplasias Hepáticas , Datos de Secuencia Molecular , Fosfoproteínas/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Receptor de Insulina/biosíntesis , Receptor de Insulina/metabolismo , Factores de Transcripción/biosíntesis , Células Tumorales Cultivadas , Dedos de Zinc
20.
J Clin Invest ; 96(1): 568-78, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7615829

RESUMEN

Chromogranin A, a soluble acidic protein, is a ubiquitous component of secretory vesicles throughout the neuroendocrine system. We reported previously the cloning and initial characterization of the mouse chromogranin A gene promoter, which showed that the promoter contains both positive and negative domains and that a proximal promoter spanning nucleotides -147 to +42 bp relative to the transcriptional start site is sufficient for neuroendocrine cell type-specific expression. The current study was undertaken to identify the particular elements within this proximal promoter that control tissue-specific expression. We found that deletion or point mutations in the potential cAMP response element (CRE) site at -68 bp virtually abolished promoter activity specifically in neuroendocrine (PC12 chromaffin or AtT20 corticotrope) cells, with little effect on activity in control (NIH3T3 fibroblast) cells; thus, the CRE box is necessary for neuroendocrine cell type-specific activity of the chromogranin A promoter. Furthermore, the effect of the CRE site is enhanced in the context of intact (wild-type) promoter sequences between -147 and -100 bp. DNase I footprint analysis showed that these regions (including the CRE box) bind nuclear proteins present in both neuroendocrine (AtT20) and control (NIH3T3) cells. In AtT20 cells, electrophoretic mobility shift assays and factor-specific antibody supershifts showed that an oligonucleotide containing the chromogranin A CRE site formed a single, homogeneous protein-DNA complex containing the CRE-binding protein CREB. However, in control NIH3T3 cells we found evidence for an additional immunologically unrelated protein in this complex. A single copy of this oligonucleotide was able to confer neuroendocrine-specific expression to a heterologous (thymidine kinase) promoter, albeit with less fold selectivity than the full proximal chromogranin A promoter. Hence, the CRE site was partially sufficient to explain the neuroendocrine cell type specificity of the promoter. The functional activity of the CRE site was confirmed through studies of the endogenous chromogranin A gene. Northern mRNA analysis showed that expression of the endogenous chromogranin A gene was stimulated seven- to eightfold by cAMP in PC12 cells, whereas no induction occurred in the NIH3T3 cells. Similar cAMP induction was obtained with the transfected chromogranin A promoter in PC12 cells, and abolition of the CRE site (by deletion or point mutation) eliminated the induction. Thus, the CRE site in the chromogranin A proximal promoter is functional and plays a crucial, indeed indispensable, role in neuroendocrine-specific expression of the gene. These results also provide insight into transcriptional mechanisms governing acquisition of the neuroendocrine secretory phenotype.


Asunto(s)
Cromograninas/genética , AMP Cíclico/farmacología , Gránulos Citoplasmáticos/metabolismo , Regiones Promotoras Genéticas , Células 3T3 , Animales , Secuencia de Bases , Cromogranina A , Cromograninas/biosíntesis , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/metabolismo , Células PC12 , ARN Mensajero/análisis , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...