Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Br J Cancer ; 128(12): 2326-2337, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37076563

RESUMEN

BACKGROUND: Current strategies to inhibit androgen receptor (AR) are circumvented in castration-resistant prostate cancer (CRPC). Cyclin-dependent kinase 7 (CDK7) promotes AR signalling, in addition to established roles in cell cycle and global transcription, providing a rationale for its therapeutic targeting in CRPC. METHODS: The antitumour activity of CT7001, an orally bioavailable CDK7 inhibitor, was investigated across CRPC models in vitro and in xenograft models in vivo. Cell-based assays and transcriptomic analyses of treated xenografts were employed to investigate the mechanisms driving CT7001 activity, alone and in combination with the antiandrogen enzalutamide. RESULTS: CT7001 selectively engages with CDK7 in prostate cancer cells, causing inhibition of proliferation and cell cycle arrest. Activation of p53, induction of apoptosis, and suppression of transcription mediated by full-length and constitutively active AR splice variants contribute to antitumour efficacy in vitro. Oral administration of CT7001 represses growth of CRPC xenografts and significantly augments growth inhibition achieved by enzalutamide. Transcriptome analyses of treated xenografts indicate cell cycle and AR inhibition as the mode of action of CT7001 in vivo. CONCLUSIONS: This study supports CDK7 inhibition as a strategy to target deregulated cell proliferation and demonstrates CT7001 is a promising CRPC therapeutic, alone or in combination with AR-targeting compounds.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Nitrilos/uso terapéutico , Quinasas Ciclina-Dependientes/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Proliferación Celular
3.
Cell Rep ; 42(4): 112297, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36961816

RESUMEN

Anti-tumor efficacy of targeted therapies is variable across patients and cancer types. Even in patients with initial deep response, tumors are typically not eradicated and eventually relapse. To address these challenges, we present a systematic screen for targets that limit the anti-tumor efficacy of EGFR and ALK inhibitors in non-small cell lung cancer and BRAF/MEK inhibitors in colorectal cancer. Our approach includes genome-wide CRISPR screens with or without drugs targeting the oncogenic driver ("anchor therapy"), and large-scale pairwise combination screens of anchor therapies with 351 other drugs. Interestingly, targeting of a small number of genes, including MCL1, BCL2L1, and YAP1, sensitizes multiple cell lines to the respective anchor therapy. Data from drug combination screens with EGF816 and ceritinib indicate that dasatinib and agents disrupting microtubules act synergistically across many cell lines. Finally, we show that a higher-order-combination screen with 26 selected drugs in two resistant EGFR-mutant lung cancer cell lines identified active triplet combinations.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas B-raf/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Recurrencia Local de Neoplasia/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores ErbB/genética , Proteínas Tirosina Quinasas Receptoras/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Mutación , Línea Celular Tumoral
4.
Nat Med ; 24(11): 1732-1742, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30297908

RESUMEN

Available corrector drugs are unable to effectively rescue the folding defects of CFTR-ΔF508 (or CFTR-F508del), the most common disease-causing mutation of the cystic fibrosis transmembrane conductance regulator, a plasma membrane (PM) anion channel, and thus to substantially ameliorate clinical phenotypes of cystic fibrosis (CF). To overcome the corrector efficacy ceiling, here we show that compounds targeting distinct structural defects of CFTR can synergistically rescue mutant expression and function at the PM. High-throughput cell-based screens and mechanistic analysis identified three small-molecule series that target defects at nucleotide-binding domain (NBD1), NBD2 and their membrane-spanning domain (MSD) interfaces. Although individually these compounds marginally improve ΔF508-CFTR folding efficiency, function and stability, their combinations lead to ~50-100% of wild-type-level correction in immortalized and primary human airway epithelia and in mouse nasal epithelia. Likewise, corrector combinations were effective against rare missense mutations in various CFTR domains, probably acting via structural allostery, suggesting a mechanistic framework for their broad application.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Fibrosis Quística/tratamiento farmacológico , Pliegue de Proteína/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Regulación Alostérica/efectos de los fármacos , Bronquios/citología , Bronquios/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/genética , Fibrosis Quística/genética , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mutación , Mucosa Nasal/citología , Mucosa Nasal/efectos de los fármacos , Dominios Proteicos/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
5.
Clin Cancer Res ; 23(11): 2856-2868, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-27986745

RESUMEN

Purpose: Anaplastic lymphoma kinase (ALK) is the most frequently mutated oncogene in the pediatric cancer neuroblastoma. We performed an in vitro screen for synergistic drug combinations that target neuroblastomas with mutations in ALK to determine whether drug combinations could enhance antitumor efficacy.Experimental Design: We screened combinations of eight molecularly targeted agents against 17 comprehensively characterized human neuroblastoma-derived cell lines. We investigated the combination of ceritinib and ribociclib on in vitro proliferation, cell cycle, viability, caspase activation, and the cyclin D/CDK4/CDK6/RB and pALK signaling networks in cell lines with representative ALK status. We performed in vivo trials in CB17 SCID mice bearing conventional and patient-derived xenograft models comparing ceritinib alone, ribociclib alone, and the combination, with plasma pharmacokinetics to evaluate for drug-drug interactions.Results: The combination of ribociclib, a dual inhibitor of cyclin-dependent kinase (CDK) 4 and 6, and the ALK inhibitor ceritinib demonstrated higher cytotoxicity (P = 0.008) and synergy scores (P = 0.006) in cell lines with ALK mutations as compared with cell lines lacking mutations or alterations in ALK Compared with either drug alone, combination therapy enhanced growth inhibition, cell-cycle arrest, and caspase-independent cell death. Combination therapy achieved complete regressions in neuroblastoma xenografts with ALK-F1174L and F1245C de novo resistance mutations and prevented the emergence of resistance. Murine ribociclib and ceritinib plasma concentrations were unaltered by combination therapy.Conclusions: This preclinical combination drug screen with in vivo validation has provided the rationale for a first-in-children trial of combination ceritinib and ribociclib in a molecularly selected pediatric population. Clin Cancer Res; 23(11); 2856-68. ©2016 AACR.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Neuroblastoma/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Aminopiridinas/administración & dosificación , Quinasa de Linfoma Anaplásico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D/genética , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/genética , Sinergismo Farmacológico , Humanos , Ratones , Mutación , Neuroblastoma/genética , Neuroblastoma/patología , Purinas/administración & dosificación , Pirimidinas/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/genética , Proteína de Retinoblastoma/genética , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Sulfonas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cell Metab ; 24(4): 582-592, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27667666

RESUMEN

Using high-throughput screening we identified small molecules that suppress superoxide and/or H2O2 production during reverse electron transport through mitochondrial respiratory complex I (site IQ) without affecting oxidative phosphorylation (suppressors of site IQ electron leak, "S1QELs"). S1QELs diminished endogenous oxidative damage in primary astrocytes cultured at ambient or low oxygen tension, showing that site IQ is a normal contributor to mitochondrial superoxide-H2O2 production in cells. They diminished stem cell hyperplasia in Drosophila intestine in vivo and caspase activation in a cardiomyocyte cell model driven by endoplasmic reticulum stress, showing that superoxide-H2O2 production by site IQ is involved in cellular stress signaling. They protected against ischemia-reperfusion injury in perfused mouse heart, showing directly that superoxide-H2O2 production by site IQ is a major contributor to this pathology. S1QELs are tools for assessing the contribution of site IQ to cell physiology and pathology and have great potential as therapeutic leads.


Asunto(s)
Citoprotección , Complejo I de Transporte de Electrón/metabolismo , Peróxido de Hidrógeno/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Células Madre/patología , Superóxidos/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citoprotección/efectos de los fármacos , Drosophila/efectos de los fármacos , Drosophila/metabolismo , Corazón/efectos de los fármacos , Hiperplasia , Intestinos/citología , Ratones , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Perfusión , Ratas , Células Madre/efectos de los fármacos , Tunicamicina/farmacología
7.
J Biomol Screen ; 21(8): 832-41, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27313114

RESUMEN

The correction or removal of signal errors in high-throughput screening (HTS) data is critical to the identification of high-quality lead candidates. Although a number of strategies have been previously developed to correct systematic errors and to remove screening artifacts, they are not universally effective and still require fair amount of human intervention. We introduce a fully automated quality control (QC) pipeline that can correct generic interplate systematic errors and remove intraplate random artifacts. The new pipeline was first applied to ~100 large-scale historical HTS assays; in silico analysis showed auto-QC led to a noticeably stronger structure-activity relationship. The method was further tested in several independent HTS runs, where QC results were sampled for experimental validation. Significantly increased hit confirmation rates were obtained after the QC steps, confirming that the proposed method was effective in enriching true-positive hits. An implementation of the algorithm is available to the screening community.


Asunto(s)
Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento/normas , Relación Estructura-Actividad , Algoritmos , Artefactos , Simulación por Computador , Humanos , Control de Calidad
8.
Nat Chem Biol ; 11(11): 834-6, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26368590

RESUMEN

Mitochondrial electron transport drives ATP synthesis but also generates reactive oxygen species, which are both cellular signals and damaging oxidants. Superoxide production by respiratory complex III is implicated in diverse signaling events and pathologies, but its role remains controversial. Using high-throughput screening, we identified compounds that selectively eliminate superoxide production by complex III without altering oxidative phosphorylation; they modulate retrograde signaling including cellular responses to hypoxic and oxidative stress.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Depuradores de Radicales Libres/farmacología , Mitocondrias/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Superóxidos/antagonistas & inhibidores , Adenosina Trifosfato/biosíntesis , Animales , Antimicina A/análogos & derivados , Antimicina A/antagonistas & inhibidores , Antimicina A/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Mitocondrias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Transducción de Señal , Superóxidos/metabolismo
9.
J Biomol Screen ; 16(1): 36-43, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21088147

RESUMEN

Dipeptidyl peptidase 1 (DPP1) (EC 3.4.14.1; also known as cathepsin C, cathepsin J, dipeptidyl aminopeptidase, and dipeptidyl aminotransferase) is a lysosomal cysteinyl protease of the papain family involved in the intracellular degradation of proteins. Isolated enzyme assays for DPP1 activity using a variety of synthetic substrates such as dipeptide or peptide linked to amino-methyl-coumarin (AMC) or other fluorophores are well established. There is, however, no report of a simple whole-cell-based assay for measuring lysosomal DPP1 activity other than the use of flow cytometry (fluorescence-activated cell sorting) or the use of invasive activity-based probes or the production of physiological products such as neutrophil elastase. The authors investigated a number of DPP1 fluorogenic substrates that have the potential to access the lysosome and enable the measurement of DPP1 enzyme activity in situ. They describe the development and evaluation of a simple noninvasive fluorescence assay for measuring DPP1 activity in fresh or cryopreserved human THP-1 cells using the substrate H-Gly-Phe-AFC (amino-fluoro-coumarin). This cell-based fluorescence assay can be performed in a 96-well plate format and is ideally suited for determining the cell potency of potential DPP1 enzyme inhibitors.


Asunto(s)
Aminocumarinas/metabolismo , Bioensayo , Catepsina C/análisis , Dipéptidos/metabolismo , Colorantes Fluorescentes/metabolismo , Lisosomas/enzimología , Animales , Línea Celular Tumoral , Fluorescencia , Humanos , Espacio Intracelular/enzimología , Especificidad por Sustrato
10.
Mol Cell Biochem ; 341(1-2): 73-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20333445

RESUMEN

Within the liver, hormonal control of glycogen metabolism allows for rapid release and uptake of glucose from the circulation, providing a reserve of glucose that can be utilised by other organs. Traditionally, cellular glycogen storage has been detected using Periodic acid Schiff (PAS) staining of histopathology samples or a biochemical assay. Colorimetric measurement of glycogen content using PAS staining is hard to quantify whilst biochemical techniques give limited information about events such as cytotoxicity or allow analysis of hepatic heterogeneity. Here, we describe the development of an imaging based method to quantify glycogen storage in 96-well cultures of primary rat hepatocytes using the inherent fluorescence properties of the Schiff reagent. PAS-stained hepatocytes were imaged using an automated fluorescent microscope, with the amount of glycogen present in each cell being quantified. Using this technique, we found an increase in glycogen storage in response to insulin (EC50 = 0.31 nM) that was in agreement with that determined using biochemical quantification (EC50 = 0.32 nM). Furthermore, a dose dependent increase in glycogen storage was also seen in response to glycogen synthase kinase inhibitors and glycogen phosphorylase inhibitors. This technique allows rapid assessment of cellular glycogen storage in response to hormones and small molecule inhibitors.


Asunto(s)
Diagnóstico por Imagen/métodos , Glucógeno/metabolismo , Hepatocitos/metabolismo , Análisis por Micromatrices/métodos , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Glucógeno Fosforilasa/antagonistas & inhibidores , Glucógeno Sintasa/antagonistas & inhibidores , Hepatocitos/citología , Insulina/farmacología , Métodos , Microscopía Fluorescente , Ratas , Bases de Schiff
11.
Expert Opin Drug Saf ; 7(4): 351-65, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18613800

RESUMEN

BACKGROUND: Ximelagatran, the first oral agent in the new class of direct thrombin inhibitors, was withdrawn from the market due to increased rates of liver enzyme elevations in long-term treatments. Despite intensive pre clinical investigations the cellular mechanisms behind the observed hepatic effects remain unknown. OBJECTIVE: The aim of this study was to assess drug-induced cytotoxicity in primary human hepatocyte cultures by ximelagatran and other reference pharmaceutical agents with known in vivo hepatotoxic profiles. METHODS: Drugs cause liver injury by many distinct mechanisms that result in abnormal cellular functioning and different patterns of injury. To address many potential toxic mechanisms in a human-relevant model, freshly isolated human hepatocytes were used in automated imaging assays. Ximelagatran was used as a test compound to study biochemical and morphological changes in human hepatocytes. In addition, 11 control, reference and comparator compounds with known liver-toxic potential in humans were used. The response to these compounds was assessed across five different hepatocyte donor preparations. RESULTS: Cytotoxicity induced by a number of compounds was quantitatively monitored using an automated imaging technique. A variety of morphological changes in hepatocyte cytoskeleton and mitochondrial function could be identified at sublethal doses of test compounds. Doses of ximelagatran up to 500 microM did not cause a cytotoxic response in the majority of preparations and no subcytotoxic response was observed at doses below 125 microM. CONCLUSIONS: The experiments described here demonstrate that primary human hepatocytes may be used in a medium-throughput format for screening using imaging-based assays for the identification of cellular responses. Overall, it is concluded that ximelagatran did not cause a significant decrease in cell viability when incubated for 24 h at considerably higher concentrations than are found in plasma following therapeutic dosing.


Asunto(s)
Anticoagulantes/efectos adversos , Azetidinas/efectos adversos , Bencilaminas/efectos adversos , Hepatocitos/efectos de los fármacos , Anticoagulantes/administración & dosificación , Azetidinas/administración & dosificación , Bencilaminas/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Microscopía Fluorescente/métodos , Mitocondrias Hepáticas/efectos de los fármacos
12.
Am J Physiol Endocrinol Metab ; 285(2): E262-71, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12644449

RESUMEN

We studied acute changes of secretory vesicle pH in pancreatic beta-cells with a fluorescent pH indicator, lysosensor green DND-189. Fluorescence was decreased by 0.66 +/- 0.10% at 149 +/- 16 s with 22.2 mM glucose stimulation, indicating that vesicular pH was alkalinized by approximately 0.016 unit. Glucose-responsive pH increase was observed when cytosolic Ca2+ influx was blocked but disappeared when an inhibitor of glycolysis or mitochondrial ATP synthase was present. Glutamate dimethyl ester (GME), a plasma membrane-permeable analog of glutamate, potentiated glucose-stimulated insulin secretion at 5 mM without changing cellular ATP content or cytosolic Ca2+ concentration ([Ca2+]). Application of GME at basal glucose concentration decreased DND-189 fluorescence by 0.83 +/- 0.19% at 38 +/- 2 s. These results indicated that the acutely alkalinizing effect of glucose on beta-cell secretory vesicle pH was dependent on glucose metabolism but independent of modulations of cytosolic [Ca2+]. Moreover, glutamate derived from glucose may be one of the mediators of this alkalinizing effect of glucose, which may have potential relevance to the alteration of secretory function by glutamate.


Asunto(s)
Glucosa/metabolismo , Glucosa/farmacología , Glutamatos/farmacología , Insulina/metabolismo , Islotes Pancreáticos/ultraestructura , Vesículas Secretoras/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Colorantes Fluorescentes , Concentración de Iones de Hidrógeno , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Canales de Potasio/efectos de los fármacos , Canales de Potasio/fisiología , Vesículas Secretoras/metabolismo
13.
J Physiol ; 544(2): 429-45, 2002 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-12381816

RESUMEN

Glucose-responsive (GR) neurons from hypothalamic nuclei are implicated in the regulation of feeding and satiety. To determine the role of intracellular ATP in the closure of ATP-sensitive K(+) (K(ATP)) channels in these cells and associated glia, the cytosolic ATP concentration ([ATP](c)) was monitored in vivo using adenoviral-driven expression of recombinant targeted luciferases and bioluminescence imaging. Arguing against a role for ATP in the closure of K(ATP) channels in GR neurons, glucose (3 or 15 mM) caused no detectable increase in [ATP](c), monitored with cytosolic luciferase, and only a small decrease in the concentration of ATP immediately beneath the plasma membrane, monitored with a SNAP25-luciferase fusion protein. In contrast to hypothalamic neurons, hypothalamic glia responded to glucose (3 and 15 mM) with a significant increase in [ATP](c). Both neurons and glia from the cerebellum, a glucose-unresponsive region of the brain, responded robustly to 3 or 15 mM glucose with increases in [ATP](c). Further implicating an ATP-independent mechanism of K(ATP) channel closure in hypothalamic neurons, removal of extracellular glucose (10 mM) suppressed the electrical activity of GR neurons in the presence of a fixed, high concentration (3 mM) of intracellular ATP. Neurons from both brain regions responded to 5 mM lactate (but not pyruvate) with an oligomycin-sensitive increase in [ATP](c). High levels of the plasma membrane lactate-monocarboxylate transporter, MCT1, were found in both cell types, and exogenous lactate efficiently closed K(ATP) channels in GR neurons. These data suggest that (1) ATP-independent intracellular signalling mechanisms lead to the stimulation of hypothalamic neurons by glucose, and (2) these effects may be potentiated in vivo by the release of lactate from neighbouring glial cells.


Asunto(s)
Adenosina Trifosfato/metabolismo , Células Quimiorreceptoras/fisiología , Citosol/metabolismo , Glucosa/metabolismo , Hipotálamo/fisiología , Neuronas/fisiología , Adenosina Trifosfato/fisiología , Animales , Membrana Celular/metabolismo , Células Cultivadas , Cerebelo/citología , Cerebelo/metabolismo , Electrofisiología , Metabolismo Energético , Hipotálamo/citología , Lactatos/metabolismo , Masculino , Transportadores de Ácidos Monocarboxílicos/metabolismo , NADP/metabolismo , Neuroglía/metabolismo , Concentración Osmolar , Canales de Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar
14.
J Cell Sci ; 115(Pt 21): 4177-89, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12356920

RESUMEN

Recruitment of secretory vesicles to the cell surface is essential for the sustained secretion of insulin in response to glucose. At present, the molecular motors involved in this movement, and the mechanisms whereby they may be regulated, are undefined. To investigate the role of kinesin family members, we labelled densecore vesicles in clonal beta-cells using an adenovirally expressed, vesicle-targeted green fluorescent protein (phogrin.EGFP), and employed immunoadsorption to obtain highly purified insulin-containing vesicles. Whereas several kinesin family members were expressed in this cell type, only conventional kinesin heavy chain (KHC) was detected in vesicle preparations. Expression of a dominant-negative KHC motor domain (KHC(mut)) blocked all vesicular movements with velocity >0.4 micro m second(-1), which demonstrates that kinesin activity was essential for vesicle motility in live beta-cells. Moreover, expression of KHC(mut) strongly inhibited the sustained, but not acute, stimulation of secretion by glucose. Finally, vesicle movement was stimulated by ATP dose-dependently in permeabilized cells, which suggests that glucose-induced increases in cytosolic [ATP] mediate the effects of the sugar in vivo, by enhancing kinesin activity. These data therefore provide evidence for a novel mechanism whereby glucose may enhance insulin release.


Asunto(s)
Células Clonales/metabolismo , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Cinesinas/metabolismo , Transporte de Proteínas/genética , Vesículas Secretoras/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Células Cultivadas , Células Clonales/efectos de los fármacos , Citosol/metabolismo , Relación Dosis-Respuesta a Droga , Exocitosis/efectos de los fármacos , Exocitosis/genética , Glucosa/farmacología , Proteínas Fluorescentes Verdes , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Cinesinas/genética , Proteínas Luminiscentes , Proteínas de la Membrana , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación/genética , Transporte de Proteínas/efectos de los fármacos , Proteínas Tirosina Fosfatasas , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores , Proteínas Recombinantes de Fusión , Vesículas Secretoras/efectos de los fármacos
15.
J Biol Chem ; 277(40): 37702-10, 2002 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-12149258

RESUMEN

The mechanisms by which glucose may affect protein kinase C (PKC) activity in the pancreatic islet beta-cell are presently unclear. By developing adenovirally expressed chimeras encoding fusion proteins between green fluorescent protein and conventional (betaII), novel (delta), or atypical (zeta) PKCs, we show that glucose selectively alters the subcellular localization of these enzymes dynamically in primary islet and MIN6 beta-cells. Examined by laser scanning confocal or total internal reflection fluorescence microscopy, elevated glucose concentrations induced oscillatory translocations of PKCbetaII to spatially confined regions of the plasma membrane. Suggesting that increases in free cytosolic Ca(2+) concentrations ([Ca(2+)](c)) were primarily responsible, prevention of [Ca(2+)](c) increases with EGTA or diazoxide completely eliminated membrane recruitment, whereas elevation of cytosolic [Ca(2+)](c) with KCl or tolbutamide was highly effective in redistributing PKCbetaII both to the plasma membrane and to the surface of dense core secretory vesicles. By contrast, the distribution of PKCdelta.EGFP, which binds diacylglycerol but not Ca(2+), was unaffected by glucose. Measurement of [Ca(2+)](c) immediately beneath the plasma membrane with a ratiometric "pericam," fused to synaptic vesicle-associated protein-25, revealed that depolarization induced significantly larger increases in [Ca(2+)](c) in this domain. These data demonstrate that nutrient stimulation of beta-cells causes spatially and temporally complex changes in the subcellular localization of PKCbetaII, possibly resulting from the generation of Ca(2+) microdomains. Localized changes in PKCbetaII activity may thus have a role in the spatial control of insulin exocytosis.


Asunto(s)
Glucosa/farmacología , Islotes Pancreáticos/fisiología , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Animales , Línea Celular , Membrana Celular/enzimología , Membrana Celular/fisiología , Células Clonales , Citosol/enzimología , Diazóxido/farmacología , Ácido Egtácico/farmacología , Proteínas Fluorescentes Verdes , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/enzimología , Cinética , Proteínas Luminiscentes/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Potasio/farmacología , Proteína Quinasa C beta , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo
16.
Biochem Biophys Res Commun ; 291(4): 1081-8, 2002 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-11866475

RESUMEN

Glucose-stimulated increases in mitochondrial metabolism are generally thought to be important for the activation of insulin secretion. Pyruvate dehydrogenase (PDH) is a key regulatory enzyme, believed to govern the rate of pyruvate entry into the citrate cycle. We show here that elevated glucose concentrations (16 or 30 vs 3 mM) cause an increase in PDH activity in both isolated rat islets, and in a clonal beta-cell line (MIN6). However, increases in PDH activity elicited with either dichloroacetate, or by adenoviral expression of the catalytic subunit of pyruvate dehydrogenase phosphatase, were without effect on glucose-induced increases in mitochondrial pyridine nucleotide levels, or cytosolic ATP concentration, in MIN6 cells, and insulin secretion from isolated rat islets. Similarly, the above parameters were unaffected by blockade of the glucose-induced increase in PDH activity by adenovirus-mediated over-expression of PDH kinase (PDK). Thus, activation of the PDH complex plays an unexpectedly minor role in stimulating glucose metabolism and in triggering insulin release.


Asunto(s)
Adenoviridae/genética , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Piruvato Deshidrogenasa (Lipoamida)/metabolismo , Animales , Células Cultivadas , Células Clonales , Ácido Dicloroacético/farmacología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Vectores Genéticos , Glucosa/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/enzimología , Masculino , Microscopía Fluorescente , Mitocondrias/metabolismo , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/genética , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Ratas , Ratas Wistar , Transfección
17.
Diabetes ; 51 Suppl 1: S162-70, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11815476

RESUMEN

Normal glucose-stimulated insulin secretion is pulsatile, but the molecular mechanisms underlying this pulsatility are poorly understood. Oscillations in the intracellular free [ATP]/[ADP] ratio represent one possible mechanism because they would be expected to cause fluctuations in ATP-sensitive K(+) channel activity and hence oscillatory Ca(2+) influx. After imaging recombinant firefly luciferase, expressed via an adenoviral vector in single human or mouse islet beta-cells, we report here that cytosolic free ATP concentrations oscillate and that these oscillations are affected by glucose. In human beta-cells, oscillations were observed at both 3 and 15 mmol/l glucose, but the oscillations were of a longer wavelength at the higher glucose concentration (167 vs. 66 s). Mouse beta-cells displayed oscillations in both cytosolic free [Ca(2+)] and [ATP] only at elevated glucose concentrations, both with a period of 120 s. To explore the causal relationship between [Ca(2+)] and [ATP] oscillations, the regulation of each was further investigated in populations of MIN6 beta-cells. Incubation in Ca(2+)-free medium lowered cytosolic [Ca(2+)] but increased [ATP] in MIN6 cells at both 3 and 30 mmol/l glucose. Removal of external Ca(2+) increased [ATP], possibly by decreasing ATP consumption by endoplasmic reticulum Ca(2+)-ATPases. These results allow a model to be constructed of the beta-cell metabolic oscillator that drives nutrient-induced insulin secretion.


Asunto(s)
Adenosina Trifosfato/metabolismo , Señalización del Calcio/fisiología , Glucosa/farmacología , Islotes Pancreáticos/metabolismo , Periodicidad , Animales , Calcio/metabolismo , ATPasas Transportadoras de Calcio/metabolismo , Citosol/metabolismo , Genes Reporteros , Humanos , Insulinoma , Islotes Pancreáticos/citología , Luciferasas/genética , Masculino , Ratones , Ratones Endogámicos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...