Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Neuromolecular Med ; 22(2): 218-226, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31664682

RESUMEN

Mutations in LRRK2 are currently recognized as the most common monogenetic cause of Parkinsonism. The elevation of kinase activity of LRRK2 that frequently accompanies its mutations is widely thought to contribute to its toxicity. Accordingly, many groups have developed LRRK2-specific kinase inhibitors as a potential therapeutic strategy. Given that protein phosphorylation is a reversible event, we sought to elucidate the phosphatase(s) that can reverse LRRK2-mediated phosphorylation, with the view that targeting this phosphatase(s) may similarly be beneficial. Using an unbiased RNAi phosphatase screen conducted in a Drosophila LRRK2 model, we identified PP2A as a genetic modulator of LRRK2-induced neurotoxicity. Further, we also identified ribosomal S6 kinase (S6K), a target of PP2A, as a novel regulator of LRRK2 function. Finally, we showed that modulation of PP2A or S6K activities ameliorates LRRK2-associated disease phenotype in Drosophila.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/enzimología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína Fosfatasa 2/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Animales , Animales Modificados Genéticamente , Línea Celular , Ceramidas/farmacología , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Mutación con Ganancia de Función , Técnicas de Silenciamiento del Gen , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Mutación Missense , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/fisiología , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Proteínas Quinasas S6 Ribosómicas/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
2.
Cancer Sci ; 110(12): 3761-3772, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31553107

RESUMEN

Differentiation-inducing factor-1 (DIF-1) has been reported to inhibit the proliferation of various mammalian cells by unknown means, although some possible mechanisms of its action have been proposed, including the activation of glycogen synthase kinase-3 (GSK-3). Here, we report an alternative mechanism underlying the action of DIF-1 in human breast cancer cell line MCF-7, on which the effects of DIF-1 have not been examined previously. Intragastric administration of DIF-1 reduced the tumor growth from MCF-7 cells injected into a mammary fat pad of nude mice, without causing adverse effects. In cultured MCF-7, DIF-1 arrested the cell cycle in G0 /G1 phase and suppressed cyclin D1 expression, consistent with our previous results obtained in other cell species. However, DIF-1 did not inhibit the phosphorylation of GSK-3. Investigating an alternative mechanism for the reduction of cyclin D1, we found that DIF-1 reduced the protein levels of signal transducer and activator of transcription 3 (STAT3). The STAT3 inhibitor S3I-201 suppressed cyclin D1 expression and cell proliferation and the overexpression of STAT3 enhanced cyclin D1 expression and accelerated proliferation. Differentiation-inducing factor-1 did not reduce STAT3 mRNA or reduce STAT3 protein in the presence of cycloheximide, suggesting that DIF-1 inhibited STAT3 protein synthesis. Seeking its mechanism, we revealed that DIF-1 inhibited the activation of 70 kDa and/or 85 kDa ribosomal protein S6 kinase (p70S6K /p85S6K ). Inhibition of p70S6K /p85S6K by rapamycin also reduced the expressions of STAT3 and cyclin D1. Therefore, DIF-1 suppresses MCF-7 proliferation by inhibiting p70S6K /p85S6K activity and STAT3 protein synthesis followed by reduction of cyclin D1 expression.


Asunto(s)
Ciclina D1/antagonistas & inhibidores , Hexanonas/farmacología , Hidrocarburos Clorados/farmacología , Proteínas Quinasas S6 Ribosómicas/fisiología , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Proliferación Celular/efectos de los fármacos , Ciclina D1/análisis , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Fosforilación , Proteínas Quinasas S6 Ribosómicas 70-kDa , Factor de Transcripción STAT3/biosíntesis
3.
Artículo en Inglés, Español | MEDLINE | ID: mdl-29573781

RESUMEN

BACKGROUND AND OBJECTIVES: We have previously indicated that EGFR has a role in carcinogenesis in a subgroup of sinonasal squamous cell carcinomas (SNSCC). In addition, EGFR activates 2 of the most important intracellular signalling pathways: PI3K/pAKT/mTOR/pS6 and MAP pathway kinases. The objective of this study was to evaluate the involvement of the EGFR/PI3K/pAKT/mTOR/pS6 pathway and its relationship with clinical-pathological parameters and follow-up of sinonasal squamous cell carcinoma. MATERIAL AND METHODS: The immunohistochemical expression of different components of the PI3K/AKT/mTOR/pS6 pathway and its relationship with various clinical-pathological parameters was studied in a series of 54 patients with SNSCC. RESULTS: Loss of PTEN expression was observed in 33/54 cases (61%) and pAKT, mTOR and pS6 pre-expression was observed in 19/54 cases (35%), 8/54 cases (15%), and 47/54 cases (87%), respectively. Loss of PTEN expression was related to intracranial invasion and development of regional metastases (p=0.005). Overexpression of pS6 was associated with a decrease in survival (p=0.008), presence of local recurrences (p=0.055), and worsening of overall prognosis (p=0.007). No significant relationships were observed between pAKT and mTOR expression and the clinicopathological parameters studied. CONCLUSIONS: Alterations in the expression of EGFR/PI3K/pAKT/mTOR/pS6 pathway components are common in a subgroup of SNSCC. This study reveals that the absence of pS6 overexpression is associated with better clinical outcomes. Therefore, pS6 expression could be considered as an unfavourable prognostic marker.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/metabolismo , Proteínas de Neoplasias/fisiología , Neoplasias Nasales/metabolismo , Neoplasias de los Senos Paranasales/metabolismo , Transducción de Señal , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/mortalidad , Receptores ErbB/fisiología , Femenino , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/genética , Recurrencia Local de Neoplasia , Estadificación de Neoplasias , Neoplasias Nasales/química , Neoplasias Nasales/mortalidad , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Neoplasias de los Senos Paranasales/química , Neoplasias de los Senos Paranasales/mortalidad , Fosfatidilinositol 3-Quinasas/fisiología , Pronóstico , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Eliminación de Secuencia , Serina-Treonina Quinasas TOR/fisiología
4.
Semin Cancer Biol ; 48: 53-61, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28476656

RESUMEN

The 90kDa ribosomal S6 kinase (RSK) family is a group of Ser/Thr protein kinases (RSK1-4) that function downstream of the Ras/mitogen-activated protein kinase (MAPK) signalling pathway. RSK regulates many substrates involved in cell survival, growth, and proliferation, and as such, deregulated RSK activity has been associated with multiple cancer types. RSK expression and activity are dysregulated in several malignancies, including breast, prostate, and lung cancer, and available evidence suggests that RSK may be a promising cancer therapeutic target. Current limitations include the lack of RSK inhibitors with suitable pharmacokinetics and selectivity toward particular isoforms. This review briefly describes the current knowledge on RSK activation and function, with a particular emphasis on RSK-dependent mechanisms associated with tumorigenesis and pharmacological inhibition.


Asunto(s)
Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas S6 Ribosómicas/fisiología , Animales , Adhesión Celular , Ciclo Celular/fisiología , Proliferación Celular , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Proteínas Quinasas S6 Ribosómicas/antagonistas & inhibidores
5.
Biol Pharm Bull ; 40(5): 576-582, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28163294

RESUMEN

The homeostasis of muscle properties depends on both physical and metabolic stresses. Whereas physical stress entails metabolic response for muscle homeostasis, the latter does not necessarily involve the former and may thus solely affect the homeostasis. We here report that metabolic suppression by the hypometabolic agent 3-iodothyronamine (T1AM) induced muscle cell atrophy without physical stress. We observed that the oxygen consumption rate of C2C12 myotubes decreased 40% upon treatment with 75 µM T1AM for 6 h versus 10% in the vehicle (dimethyl sulfoxide) control. The T1AM treatment reduced cell diameter of myotubes by 15% compared to the control (p<0.05). The cell diameter was reversed completely by 9 h after T1AM was removed. The T1AM treatment also significantly suppressed the expression levels of heat shock protein 72 and αB-crystallin as well as the phosphorylation levels of Akt1, mammalian target of rapamycin (mTOR), S6K, forkhead box O1 (FoxO1) and FoxO3. In contrast, the levels of ubiquitin E3 ligase MuRF1 and chymotrypsin-like activity of proteasome were significantly elevated by T1AM treatment. These results suggest that T1AM-mediated metabolic suppression induced muscle cell atrophy via activation of catabolic signaling and inhibition of anabolic signaling.


Asunto(s)
Proteína Forkhead Box O1/fisiología , Atrofia Muscular/inducido químicamente , Complejo de la Endopetidasa Proteasomal/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Transducción de Señal/fisiología , Tironinas/farmacología , Animales , Células Cultivadas , Regulación hacia Abajo , Ratones , Fibras Musculares Esqueléticas , Serina-Treonina Quinasas TOR/fisiología
6.
Proc Natl Acad Sci U S A ; 113(32): E4620-9, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27457958

RESUMEN

Dietary restriction (DR) is a metabolic intervention that extends the lifespan of multiple species, including yeast, flies, nematodes, rodents, and, arguably, rhesus monkeys and humans. Hallmarks of lifelong DR are reductions in body size, fecundity, and fat accumulation, as well as slower development. We have identified atx-2, the Caenorhabditis elegans homolog of the human ATXN2L and ATXN2 genes, as the regulator of these multiple DR phenotypes. Down-regulation of atx-2 increases the body size, cell size, and fat content of dietary-restricted animals and speeds animal development, whereas overexpression of atx-2 is sufficient to reduce the body size and brood size of wild-type animals. atx-2 regulates the mechanistic target of rapamycin (mTOR) pathway, downstream of AMP-activated protein kinase (AMPK) and upstream of ribosomal protein S6 kinase and mTOR complex 1 (TORC1), by its direct association with Rab GDP dissociation inhibitor ß, which likely regulates RHEB shuttling between GDP-bound and GTP-bound forms. Taken together, this work identifies a previously unknown mechanism regulating multiple aspects of DR, as well as unknown regulators of the mTOR pathway. They also extend our understanding of diet-dependent growth retardation, and offers a potential mechanism to treat obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Ataxina-2/fisiología , Caenorhabditis elegans/crecimiento & desarrollo , Tamaño de la Célula , Serina-Treonina Quinasas TOR/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Animales , Caenorhabditis elegans/citología , Dieta , Proteínas Quinasas S6 Ribosómicas/fisiología , Transducción de Señal/fisiología
7.
Cell Signal ; 27(10): 2045-53, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26215099

RESUMEN

Increased rates of ribosome biogenesis and biomass accumulation are fundamental properties of rapidly growing and dividing malignant cells. The MYC oncoprotein drives growth predominantly via its ability to upregulate the ribosome biogenesis program, in particular stimulating the activity of the RNA Polymerase I (Pol I) machinery to increase ribosomal RNA (rRNA) transcription. Although MYC function is known to be highly dependent on the cellular signalling context, the pathways interacting with MYC to regulate transcription of ribosomal genes (rDNA) in vivo in response to growth factor status, nutrient availability and cellular stress are only beginning to be understood. To determine factors critical to MYC-dependent stimulation of rDNA transcription in vivo, we performed a transient expression screen for known oncogenic signalling pathways in Drosophila. Strikingly, from the broad range of pathways tested, we found that ribosomal protein S6 Kinase (S6K) activity, downstream of the TOR pathway, was the only factor rate-limiting for the rapid induction of rDNA transcription due to transiently increased MYC. Further, we demonstrated that one of the mechanism(s) by which MYC and S6K cooperate is through coordinate activation of the essential Pol I transcription initiation factor TIF-1A (RRN 3). As Pol I targeted therapy is now in phase 1 clinical trials in patients with haematological malignancies, including those driven by MYC, these data suggest that therapies dually targeting Pol I transcription and S6K activity may be effective in treating MYC-driven tumours.


Asunto(s)
ADN Ribosómico/genética , Drosophila melanogaster/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Transcripción Genética , Animales , Nucléolo Celular/enzimología , Nucléolo Celular/ultraestructura , Ojo Compuesto de los Artrópodos/enzimología , Ojo Compuesto de los Artrópodos/ultraestructura , ADN Ribosómico/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Nucleares/metabolismo , Glándulas Salivales/enzimología , Glándulas Salivales/ultraestructura , Factores de Transcripción/metabolismo
8.
Eur J Cell Biol ; 94(10): 470-81, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26054233

RESUMEN

It is well established that mTORC1 suppresses autophagy by phosphorylation and inactivation of proteins involved in autophagosome formation. However, the role of its substrate, p70S6 kinase1 (S6K1), in autophagy is quite controversial. In some models S6K1 activity correlates with autophagy suppression, however, some other studies show that S6K1 promotes rather than inhibits this process. Here, we investigated the role of S6K1 in prostate cancer cells (PC-3) and non-cancerous, mouse embryonic fibroblasts (MEF), either treated with autophagy inducer sulforaphane, an isothiocyanate derived from cruciferous plants, or deprived of serum. Our results indicate that constitutively active S6K1 decreases the level of LC3 processing and foci formation by autophagosomal vacuoles in cells treated with sulforaphane. On the other hand, presence of S6K1 is necessary for autophagosome maturation under conditions of autophagy induced by either sulforaphane or serum deprivation. Diminished level of S6K1 or lack of S6 kinases results in both, accumulation of autophagosomes and drop in the autophagolysosome number, and thus disturbs autophagy flux under stress conditions. Moreover, lack of S6 kinases reduces cell survival under stress conditions.


Asunto(s)
Anticarcinógenos/farmacología , Autofagia , Fibroblastos , Isotiocianatos/farmacología , Neoplasias de la Próstata , Proteínas Quinasas S6 Ribosómicas 70-kDa/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Animales , Autofagia/efectos de los fármacos , Autofagia/fisiología , Línea Celular Tumoral , Medio de Cultivo Libre de Suero , Fibroblastos/enzimología , Fibroblastos/ultraestructura , Humanos , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Fagosomas , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Sulfóxidos
9.
J Neurochem ; 134(5): 845-56, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26016889

RESUMEN

We examined the role of the mechanistic target of rapamycin (mTOR) pathway in delayed diazoxide (DZ)-induced preconditioning of cultured rat primary cortical neurons. Neurons were treated for 3 days with 500 µM DZ or feeding medium and then exposed to 3 h of continuous normoxia in Dulbecco's modified eagle medium with glucose or with 3 h of oxygen-glucose deprivation (OGD) followed by normoxia and feeding medium. The OGD decreased viability by 50%, depolarized mitochondria, and reduced mitochondrial respiration, whereas DZ treatment improved viability and mitochondrial respiration, and suppressed reactive oxygen species production, but did not restore mitochondrial membrane potential after OGD. Neuroprotection by DZ was associated with increased phosphorylation of protein kinase B (Akt), mTOR, and the major mTOR downstream substrate, S6 Kinase (S6K). The mTOR inhibitors rapamycin and Torin-1, as well as S6K-targeted siRNA abolished the protective effects of DZ. The effects of DZ on mitochondrial membrane potential and reactive oxygen species production were not affected by rapamycin. Preconditioning with DZ also changed mitochondrial and non-mitochondrial oxygen consumption rates. We conclude that in addition to reducing reactive oxygen species (ROS) production and mitochondrial membrane depolarization, DZ protects against OGD by activation of the Akt-mTOR-S6K pathway and by changes in mitochondrial respiration. Ischemic strokes have limited therapeutic options. Diazoxide (DZ) preconditioning can reduce neuronal damage. Using oxygen-glucose deprivation (OGD), we studied Akt/mTOR/S6K signaling and mitochondrial respiration in neuronal preconditioning. We found DZ protects neurons against OGD via the Akt/mTOR/S6K pathway and alters the mitochondrial and non-mitochondrial oxygen consumption rate. This suggests that the Akt/mTOR/S6k pathway and mitochondria are novel stroke targets.


Asunto(s)
Diazóxido/farmacología , Precondicionamiento Isquémico , Proteínas del Tejido Nervioso/fisiología , Neuronas/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas/fisiología , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Medios de Cultivo/farmacología , Activación Enzimática/efectos de los fármacos , Técnicas In Vitro , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Neuronas/metabolismo , Oxígeno/farmacología , Consumo de Oxígeno , Fosforilación , Cultivo Primario de Células , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Proteínas Quinasas S6 Ribosómicas/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas/genética , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
10.
Proc Natl Acad Sci U S A ; 112(19): 6188-93, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25918363

RESUMEN

Current treatments for major depressive disorder (MDD) have a time lag and are ineffective for a large number of patients. Development of novel pharmacological therapies requires a comprehensive understanding of the molecular events that contribute to MDD pathophysiology. Recent evidence points toward aberrant activity of synaptic proteins as a critical contributing factor. In the present studies, we used viral-mediated gene transfer to target a key mediator of activity-dependent synaptic protein synthesis downstream of mechanistic target of rapamycin complex 1 (mTORC1) known as p70 S6 kinase 1 (S6K1). Targeted delivery of two mutants of S6K1, constitutively active or dominant-negative, to the medial prefrontal cortex (mPFC) of rats allowed control of the mTORC1/S6K1 translational pathway. Our results demonstrate that increased expression of S6K1 in the mPFC produces antidepressant effects in the forced swim test without altering locomotor activity. Moreover, expression of active S6K1 in the mPFC blocked the anhedonia caused by chronic stress, resulting in a state of stress resilience. This antidepressant response was associated with increased neuronal complexity caused by enhanced S6K1 activity. Conversely, expression of dominant-negative S6K1 in the mPFC resulted in prodepressive behavior in the forced swim test and was sufficient to cause anhedonia in the absence of chronic stress exposure. Together, these data demonstrate a critical role for S6K1 activity in depressive behaviors, and suggest that pathways downstream of mTORC1 may underlie the pathophysiology and treatment of MDD.


Asunto(s)
Trastorno Depresivo Mayor/metabolismo , Regulación Enzimológica de la Expresión Génica , Corteza Prefrontal/metabolismo , Proteínas Quinasas S6 Ribosómicas/fisiología , Animales , Antidepresivos/uso terapéutico , Conducta Animal , Modelos Animales de Enfermedad , Ketamina/química , Masculino , Aprendizaje por Laberinto , Neuronas/metabolismo , Fenotipo , Fosforilación , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas S6 Ribosómicas/genética , Transducción de Señal , Sirolimus/química , Natación
11.
Oncogene ; 34(4): 413-23, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-24469061

RESUMEN

The metastatic cascade is a complex and multistep process with many potential barriers. Recent evidence has shown that microRNAs (miRNAs) are involved in carcinogenesis and tumor progression in non-small-cell lung cancer (NSCLC). In this study, by comparing the miRNA expression profiles of SPC-A-1sci (high metastatic) and SPC-A-1 (weakly metastatic) cells, we demonstrated that the downregulation and function of miR-193a-3p and miR-193a-5p in NSCLC metastasis and the expression of these miRNAs was suppressed in NSCLC compared with corresponding non-tumorous tissues. Decreased miR-193a-3p/5p expression was significantly associated with tumor node metastasis (TNM) and lymph node metastasis. Furthermore, functional assays showed that the overexpression of miR-193a-3p/5p inhibited NSCLC cell migration, invasion and epithelial-mesenchymal transition (EMT) in vitro and lung metastasis formation in vivo. In addition, we discovered that ERBB4 and S6K2 were the direct targets of miR-193a-3p and that PIK3R3 and mTOR were the direct targets of miR-193a-5p in NSCLC. We also observed that miR-193a-3p/5p could inactivate the AKT/mTOR signaling pathway. Thus, miR-193a-3p/5p functions as a tumor suppressor and has an important role in NSCLC metastasis through ERBB signaling pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , MicroARNs/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Receptor ErbB-4/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/secundario , Línea Celular Tumoral , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Metástasis de la Neoplasia/prevención & control , Transducción de Señal/fisiología
12.
FEBS J ; 281(14): 3177-92, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24848561

RESUMEN

The Ras-ERK and PI3K-mTOR pathways are hyperactivated in various malignant tumors. Feedforward (FF) and feedback (FB) regulations between the Ras-ERK and the PI3K-mTOR pathways have been suggested to attenuate sensitivity to drugs targeting these pathways and confer tumor resistance to therapies. However, because analyses of such regulations require measurements and perturbations with high temporal resolution, the quantitative roles played by FF and FB regulations in the intrinsic resistance to molecular targeting drugs still remain unclear. To address this issue, we quantified FF and FB regulations of the epidermal growth factor receptor (EGFR) signaling pathway by Förster/fluorescence resonance energy transfer (FRET) imaging. EGF-induced activation of EGFR, Ras, extracellular-signal-regulated kinase and S6K with or without inhibitors was measured by FRET imaging, and analyzed by semi-automatic image processing. Based on the imaging data set and kinetic parameters determined by our previous studies, we identified the roles played by a coherent FF regulation and two negative FB regulations, one of which was not recognized previously. The systems analyses revealed how these FF and FB regulations shape the temporal dynamics of extracellular-signal-regulated kinase activity upon EGF stimulation. Furthermore, the simulation model predicts the response of molecular targeting drugs applied solely or in combination with each other to BRaf- or KRas-mutated cancer cell lines, indicating the validity of a quantitative model integrating FF and FB regulations.


Asunto(s)
Receptores ErbB/fisiología , Retroalimentación Fisiológica/fisiología , Homeostasis/fisiología , Transducción de Señal/fisiología , Sistemas de Liberación de Medicamentos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Células HeLa , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de Unión al GTP Monoméricas/fisiología , Neuropéptidos/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro , Proteínas Quinasas S6 Ribosómicas/fisiología , Biología de Sistemas , Serina-Treonina Quinasas TOR/fisiología
14.
J Am Coll Cardiol ; 61(9): 971-80, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23449430

RESUMEN

OBJECTIVES: This study sought to examine the effect of oral metformin (Mf) therapy on endothelialization in the setting of drug-eluting stents (DES). BACKGROUND: Mf is a commonly used therapy in diabetic patients receiving DES. Mf and locally eluted mammalian target of rapamycin (mTOR) inhibitors used in DES have convergent molecular signaling; however, the impact of this drug interaction on stent endothelialization is unknown. METHODS: We examined human endothelial aortic cells (HAECs) and a rabbit model of stenting to determine points on molecular convergence between these 2 agents and their impact on stent endothelialization. RESULTS: Western blotting of HAECs treated with Mf and the mTOR inhibitor sirolimus and 14-day rabbit iliacs treated with the combination of zotarolimus-eluting stents (ZES) and oral Mf demonstrated greater inhibition of S6 kinase (S6K), a downstream effector of mTOR complex 1, than either treatment alone. HAEC proliferation was significantly inhibited by Mf or sirolimus treatments alone and further reduced when they were combined. Knockdown of S6K via short interfering RNA in HAECs impaired cell proliferation via a cyclin D1-dependent mechanism, whereas its overexpression rescued the antiproliferative effects of both agents. Last, endothelialization and endothelial cell proliferation at 14 days were assessed in rabbits receiving ZES or bare-metal stents and Mf or placebo by scanning electron microscopy and bromodeoxyuridine/CD31 labeling, respectively. Both endpoints were inhibited by ZES treatment alone and were further reduced by the combination of Mf and ZES. CONCLUSIONS: Significant convergence of signaling occurs between Mf and locally delivered mTOR inhibitors at S6K. This further impairs endothelial recovery/proliferation via an S6K-dependent mechanism. Patients receiving Mf in combination with stents that elute mTOR inhibitors are potentially at increased risk of delayed endothelial healing and stent thrombosis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Stents Liberadores de Fármacos , Células Endoteliales/efectos de los fármacos , Metformina/efectos adversos , Proteínas Quinasas S6 Ribosómicas/fisiología , Sirolimus/antagonistas & inhibidores , Administración Oral , Animales , Aorta/citología , Apoptosis , Células Cultivadas , Interacciones Farmacológicas , Células Endoteliales/citología , Endotelio Vascular/citología , Humanos , Arteria Ilíaca , Metformina/administración & dosificación , Microscopía Electrónica de Rastreo , Conejos , Proteínas Quinasas S6 Ribosómicas/antagonistas & inhibidores , Sirolimus/análogos & derivados , Sirolimus/farmacología
15.
Br J Pharmacol ; 167(5): 1148-60, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22708658

RESUMEN

BACKGROUND AND PURPOSE: Neuropeptide Y (NPY) is a 36-amino acid polypeptide found abundantly in the central and peripheral nervous systems. NPY exerts a potent depressor effect via the activation of both Y(1) and Y(2) receptors in the nucleus tractus solitarii (NTS) of rats. However, the precise mechanisms involved in this NPY-mediated action remained unclear. EXPERIMENTAL APPROACH: Effects of a selective antagonist of Y(1) receptors, a PKC inhibitor, a PI3 kinase inhibitor, a NOS inhibitor, an endothelial NOS (eNOS)-selective inhibitor, a neuronal NOS (nNOS)-specific inhibitor or a MAPK inhibitor, on responses to microinjection of NPY into the NTS of Wistar-Kyoto rats were studied to determine the underlying mechanisms. Blood pressure and heart rate were measured and, in NTS, protein phosphorylation assessed by immunohistochemical techniques. KEY RESULTS: Unilateral microinjection of exogenous NPY (4.65pmol/60nL) into the NTS of urethane-anesthetized Wistar-Kyoto rats markedly decreased blood pressure and heart rate. Microinjection of the Y(1) receptor antagonist BIBP3226 or the G(i) /G(o) -protein inhibitor, Pertussis toxin, into the NTS attenuated these NPY-induced hypotensive effects. A selective Y(1) receptor agonist increased expression of ERK1/2, ribosomal protein S6 kinase (RSK) and the phosphorylation of eNOS. RSK also bound directly to eNOS and induced its phosphorylation at Ser(1177) . Pretreatment of the NTS with an eNOS inhibitor, but not a nNOS inhibitor, attenuated the NPY-induced hypotensive effects. CONCLUSIONS AND IMPLICATIONS: Together, these results suggested that NPY-induced depressor effects were mediated by activating NPY Y(1) receptor-PKC-ERK-RSK-eNOS and Ca(2+) -eNOS signalling pathways, which are involved in regulation of blood pressure in the NTS.


Asunto(s)
Presión Sanguínea/fisiología , Hipotensión/fisiopatología , Neuropéptido Y/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Núcleo Solitario/fisiología , Animales , Calcio/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Frecuencia Cardíaca/fisiología , Masculino , Óxido Nítrico/fisiología , Proteína Quinasa C/fisiología , Ratas , Ratas Endogámicas WKY , Receptores de Neuropéptido Y/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología
16.
An R Acad Nac Med (Madr) ; 129(2): 541-63; discussion 563, 2012.
Artículo en Español | MEDLINE | ID: mdl-24298863

RESUMEN

Metabolic sensors play an important role in the control of food intake, utilization of nutrients and demonstration of feeding behaviour. In this work we describe the study done in our laboratory on glucokinase (GK) as brain glucose sensor, the AMP kinase (AMPK) as detector of the fall of intracellular energy charge and as the S6K in the signaling pathway of mTOR with opposite effects to AMPK. Glucose sensors are molecular designs that detect with accuracy glucose concentrations, facilitating therefore the homeostasis of this hexose. We consider GK as a component of a glucose sensor system that might modulates the feeding behaviour and indirectly the control of body weight. Our findings indicate that GK and GLUT-2 mRNAs and proteins are coexpressed mainly in areas of the hypothalamus implied in the control of food intake. We have also found a high glucose phosphorylating activity with kinetic properties similar to that reported in the liver, with a high apparent Km for glucose that displays no product inhibition by glucose-6-phosphate. GK may be also regulated by the presence of glucokinase regulatory protein (GKRP), which has been identified in the same brain areas than GK. The coexpression of these molecules might play a role as glucose sensors in which GLUT-2 has a permissive role and the interactions of GK with GKRP made possible a real sensor activity. Furthermore, the effects of anorexigenic peptides in this system should facilitate the transduction of signals required to produce a state of satiety. Thus, GLP-1 reduced significantly the glucose metabolism in areas of the hypothalamus and brainstem related with food intake, which open new ways to the study of pathophysiologicals aspects of feeding behaviour. Besides we have studied the functions of AMPK and mTOR pathway in the hypothalamic areas ventromedial (VMH) and lateral (LH) under situations with alterations of the nutritional status and energy balance. Our results revealed that the activation of AMPK and S6K in VMH y LH occur in response to the changes of glucose concentrations or in the changes in the nutritional state, as well as GLP-1/exendin-4 act by counteracting the activation/inactivation of these kinases, which support a modulating role of these peptides on the kinases. On the other hand, GLP-1/exendin-4 might contribute to the normalization of the altered values of these kinases in pathophysiological states such as obesity.


Asunto(s)
Peso Corporal/fisiología , Encéfalo/metabolismo , Metabolismo Energético , Conducta Alimentaria/fisiología , Adenilato Quinasa/fisiología , Exenatida , Péptido 1 Similar al Glucagón/fisiología , Glucoquinasa/fisiología , Homeostasis , Humanos , Hipotálamo/fisiología , Péptidos/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Transducción de Señal , Serina-Treonina Quinasas TOR/fisiología , Ponzoñas
17.
Dev Growth Differ ; 53(8): 924-33, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21958163

RESUMEN

In the early embryogenesis of the frog, Xenopus laevis, cells proliferate by rapid and synchronous divisions, followed by cell cycle elongation and prolongation of the S phases, and then the appearance of the G2 and G1 phases after the midblastula transition (MBT). The beginning of cell cycle elongation was thought to depend on an increase in the nucleo-cytoplasmic (N/C) ratio in blastomeres and a decrease in cortical cytoplasmic factors necessary for cell cycle progression, although these factors are unknown. In the present study, we demonstrated that a regulatory subunit of PI3K (p85α) was localized in the cortical cytoplasm of the blastomere during the MBT. When the embryos were treated with a PI3K inhibitor, LY294002, or a TOR inhibitor, rapamycin, cell cycle elongation was initiated before the MBT. In addition, the inhibition of S6K expression by antisense morpholino oligo enhanced the initiation of cell cycle elongation. In contrast, the activation of PI3K-TOR by Rheb-S16H expression delayed the initiation of cell cycle elongation. These results indicate that a decrease in translational activity dependent on the PI3K-TOR-S6K pathway causes the initiation of cell cycle elongation at the onset of the MBT.


Asunto(s)
Ciclo Celular/genética , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Serina-Treonina Quinasas TOR/fisiología , Xenopus/embriología , Animales , Animales Modificados Genéticamente , Ciclo Celular/fisiología , Ciclo Celular/efectos de la radiación , Células Cultivadas , Fase de Segmentación del Huevo/metabolismo , Fase de Segmentación del Huevo/fisiología , Embrión no Mamífero , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo , Xenopus/genética , Xenopus/metabolismo
18.
Cancer Res ; 71(15): 5204-13, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21670080

RESUMEN

Most cancer cells utilize aerobic glycolysis, and activation of the phosphoinositide 3-kinase/Akt/mTOR pathway can promote this metabolic program to render cells glucose dependent. Although manipulation of glucose metabolism may provide a means to specifically eliminate cancer cells, mechanistic links between cell metabolism and apoptosis remain poorly understood. Here, we examined the role and metabolic regulation of the antiapoptotic Bcl-2 family protein Mcl-1 in cell death upon inhibition of Akt-induced aerobic glycolysis. In the presence of adequate glucose, activated Akt prevented the loss of Mcl-1 expression and protected cells from growth factor deprivation-induced apoptosis. Mcl-1 associated with and inhibited the proapoptotic Bcl-2 family protein Bim, contributing to cell survival. However, suppression of glucose metabolism led to induction of Bim, decreased expression of Mcl-1, and apoptosis. The proapoptotic Bcl-2/Bcl-xL/Bcl-w inhibitor, ABT-737, shows clinical promise, but Mcl-1 upregulation can promote resistance. Importantly, inhibition of glucose metabolism or mTORC1 overcame Mcl-1-mediated resistance in diffuse large B cell leukemic cells. Together these data show that Mcl-1 protein synthesis is tightly controlled by metabolism and that manipulation of glucose metabolism may provide a mechanism to suppress Mcl-1 expression and sensitize cancer cells to apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Glucosa/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/fisiología , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Compuestos de Bifenilo/farmacología , Proteínas de Ciclo Celular , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Humanos , Células Jurkat/efectos de los fármacos , Linfoma de Células B Grandes Difuso/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas de la Membrana/metabolismo , Ratones , Complejos Multiproteicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Nitrofenoles/farmacología , Fosfoproteínas/fisiología , Piperazinas/farmacología , Proteínas/antagonistas & inhibidores , Proteínas/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Sulfonamidas/farmacología , Linfocitos T/efectos de los fármacos , Serina-Treonina Quinasas TOR
20.
Aging (Albany NY) ; 2(8): 487-503, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20739737

RESUMEN

Deregulated nutrient signaling plays pivotal roles in body ageing and in diabetic complications; biochemical cascades linking energy dysmetabolism to cell damage and loss are still incompletely clarified, and novel molecular paradigms and pharmacological targets critically needed. We provide evidence that in the retrovirus-packaging cell line HEK293-T Phoenix, massive cell death in serum-free medium is remarkably prevented or attenuated by either glucose or aminoacid withdrawal, and by the glycolysis inhibitor 2-deoxy-glucose. A similar protection was also elicited by interference with mitochondrial function, clearly suggesting involvement of energy metabolism in increased cell survival. Oxidative stress did not account for nutrient toxicity on serum-starved cells. Instead, nutrient restriction was associated with reduced activity of the mTOR/S6 Kinase cascade. Moreover, pharmacological and genetic manipulation of the mTOR pathway modulated in an opposite fashion signaling to S6K/S6 and cell viability in nutrient-repleted medium. Additionally, stimulation of the AMP-activated Protein Kinase concomitantly inhibited mTOR signaling and cell death, while neither event was affected by overexpression of the NAD+ dependent deacetylase Sirt-1, another cellular sensor of nutrient scarcity. Finally, blockade of the mTOR cascade reduced hyperglycemic damage also in a more pathophysiologically relevant model, i.e. in human umbilical vein endothelial cells (HUVEC) exposed to hyperglycemia. Taken together these findings point to a key role of the mTOR/S6K cascade in cell damage by excess nutrients and scarcity of growth-factors, a condition shared by diabetes and other ageing-related pathologies.


Asunto(s)
Supervivencia Celular/fisiología , Privación de Alimentos/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/fisiología , Antimetabolitos/administración & dosificación , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero , Desoxiglucosa/administración & dosificación , Células HEK293 , Humanos , Mitocondrias/fisiología , Estrés Oxidativo/fisiología , Proteína S6 Ribosómica/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Transducción de Señal/fisiología , Sirtuina 1/fisiología , Serina-Treonina Quinasas TOR/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA