Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Cancer Res ; 76(18): 5467-78, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27503926

RESUMEN

SIRT2 is a protein deacetylase with tumor suppressor activity in breast and liver tumors where it is mutated; however, the critical substrates mediating its antitumor activity are not fully defined. Here we demonstrate that SIRT2 binds, deacetylates, and inhibits the peroxidase activity of the antioxidant protein peroxiredoxin (Prdx-1) in breast cancer cells. Ectopic overexpression of SIRT2, but not its catalytically dead mutant, increased intracellular levels of reactive oxygen species (ROS) induced by hydrogen peroxide, which led to increased levels of an overoxidized and multimeric form of Prdx-1 with activity as a molecular chaperone. Elevated levels of SIRT2 sensitized breast cancer cells to intracellular DNA damage and cell death induced by oxidative stress, as associated with increased levels of nuclear FOXO3A and the proapoptotic BIM protein. In addition, elevated levels of SIRT2 sensitized breast cancer cells to arsenic trioxide, an approved therapeutic agent, along with other intracellular ROS-inducing agents. Conversely, antisense RNA-mediated attenuation of SIRT2 reversed ROS-induced toxicity as demonstrated in a zebrafish embryo model system. Collectively, our findings suggest that the tumor suppressor activity of SIRT2 requires its ability to restrict the antioxidant activity of Prdx-1, thereby sensitizing breast cancer cells to ROS-induced DNA damage and cell cytotoxicity. Cancer Res; 76(18); 5467-78. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Peroxirredoxinas/metabolismo , Sirtuina 2/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Ensayo Cometa , Electroforesis en Gel Bidimensional , Femenino , Humanos , Immunoblotting , Inmunoprecipitación , Microscopía Confocal , Oxidantes/farmacología , Estrés Oxidativo/fisiología , Peroxidasa/metabolismo , Pez Cebra
3.
Mar Drugs ; 13(3): 1552-68, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25803180

RESUMEN

The biologically active lipopeptide kalkitoxin was previously isolated from the marine cyanobacterium Moorea producens (Lyngbya majuscula). Kalkitoxin exhibited N-methyl-D-aspartate (NMDA)-mediated neurotoxicity and acted as an inhibitory ligand for voltage-sensitive sodium channels in cultured rat cerebellar granule neurons. Subsequent studies revealed that kalkitoxin generated a delayed form of colon tumor cell cytotoxicity in 7-day clonogenic cell survival assays. Cell line- and exposure time-dependent cytostatic/cytotoxic effects were previously observed with mitochondria-targeted inhibitors of hypoxia-inducible factor-1 (HIF-1). The transcription factor HIF-1 functions as a key regulator of oxygen homeostasis. Therefore, we investigated the ability of kalkitoxin to inhibit hypoxic signaling in human tumor cell lines. Kalkitoxin potently and selectively inhibited hypoxia-induced activation of HIF-1 in T47D breast tumor cells (IC50 5.6 nM). Mechanistic studies revealed that kalkitoxin inhibits HIF-1 activation by suppressing mitochondrial oxygen consumption at electron transport chain (ETC) complex I (NADH-ubiquinone oxidoreductase). Further studies indicate that kalkitoxin targets tumor angiogenesis by blocking the induction of angiogenic factors (i.e., VEGF) in tumor cells.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Lípidos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Tiazoles/farmacología , Inhibidores de la Angiogénesis/administración & dosificación , Neoplasias de la Mama/patología , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Complejo I de Transporte de Electrón/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Femenino , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Concentración 50 Inhibidora , Lípidos/administración & dosificación , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Transducción de Señal/efectos de los fármacos , Tiazoles/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Invest Ophthalmol Vis Sci ; 54(1): 63-71, 2013 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-23169885

RESUMEN

PURPOSE: Hemochromatosis is a disorder of iron overload arising mostly from mutations in HFE. HFE is expressed in retinal pigment epithelium (RPE), and Hfe(-/-) mice develop age-related iron accumulation and retinal degeneration associated with RPE hyperproliferation. Here, the mechanism underlying the hyperproliferative phenotype in RPE was investigated. METHODS: Cellular senescence was monitored by ß-galactosidase activity. Gene expression was monitored by real-time PCR. Survivin was analyzed by Western blot and immunofluorescence. Migration and invasion were monitored using appropriate kits. Glucose transporters (GLUTs) were monitored by 3-O-methyl-D-glucose uptake. Histone deacetylases (HDACs) were studied by monitoring catalytic activity and acetylation status of histones H3/H4. RESULTS: Hfe(-/-) RPE cells exhibited slower senescence rate and higher survivin expression than wild type cells. Hfe(-/-) cells migrated faster and showed greater glucose uptake and increased expression of GLUTs. The expression of HDACs and DNA methyltransferase (DNMTs) also was increased. Similarly, RPE cells from hemojuvelin (Hjv)-knockout mice, another model of hemochromatosis, also had increased expression of GLUTs, HDACs, and DNMTs. The expression of Slc5a8 was decreased in Hfe(-/-) RPE cells, but treatment with a DNA methylation inhibitor restored the transporter expression, indicating involvement of DNA methylation in the silencing of Slc5a8 in Hfe(-/-) cells. CONCLUSIONS: RPE cells from iron-overloaded mice exhibit several features of tumor cells: decreased senescence, enhanced migration, increased glucose uptake, and elevated levels of HDACs and DNMTs. These features are seen in Hfe(-/-) RPE cells as well as in Hjv(-/-) RPE cells, providing a molecular basis for the hyperproliferative phenotype of Hfe(-/-) and Hjv(-/-) RPE cells.


Asunto(s)
Neoplasias del Ojo , Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Proteínas de la Membrana/genética , Epitelio Pigmentado de la Retina/patología , Epitelio Pigmentado de la Retina/fisiopatología , Sistemas de Transporte de Aminoácidos/genética , Animales , Proteínas de Transporte de Catión/genética , Movimiento Celular/fisiología , Transformación Celular Neoplásica/patología , Senescencia Celular/fisiología , Metilación de ADN/fisiología , Progresión de la Enfermedad , Neoplasias del Ojo/genética , Neoplasias del Ojo/patología , Neoplasias del Ojo/fisiopatología , Femenino , Glucosa/farmacocinética , Hemocromatosis/genética , Hemocromatosis/patología , Hemocromatosis/fisiopatología , Proteína de la Hemocromatosis , Proteínas Inhibidoras de la Apoptosis/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Transportadores de Ácidos Monocarboxílicos , Fenotipo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética , Cultivo Primario de Células , Proteínas Represoras/metabolismo , Survivin , Trasplante Heterólogo
5.
Biochem J ; 450(1): 169-78, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23167260

RESUMEN

SLC5A8 (solute carrier gene family 5A, member 8) is a sodium-coupled transporter for monocarboxylates. Among its substrates are the HDAC (histone deacetylase) inhibitors butyrate, propionate and pyruvate. Expression of SLC5A8 is silenced in cancers via DNA methylation, and ectopic expression of SLC5A8 in cancer cells induces apoptosis in the presence of its substrates that are HDAC inhibitors. In the present study we show that ectopic expression of SLC5A8 in cancer cells translocates the anti-apoptotic protein survivin to the plasma membrane through protein-protein interaction resulting in depletion of nuclear survivin and also decreases cellular levels of survivin through inhibition of transcription. These SLC5A8-induced changes in the location and levels of survivin result in cell-cycle arrest, disruption of the chromosome passenger complex involved in mitosis, induction of apoptosis and enhancement in chemosensitivity. These effects are seen independently of the transport function of SLC5A8 and histone acetylation status of the cell; in the presence of pyruvate, a SLC5A8 substrate and also an HDAC inhibitor, these effects are amplified. Ectopic expression of SLC5A8 in the breast cancer cell line MB231 inhibits the ability of cells to form colonies in vitro and to form tumours in mouse xenografts in vivo. The suppression of survivin transcription occurs independently of HDAC inhibition, and the underlying mechanism is associated with decreased phosphorylation of STAT3 (signal transducer and activator of transcription 3). The observed effects are specific for survivin with no apparent changes in expression of other inhibitor-of-apoptosis proteins. The present study unravels a novel, hitherto unrecognized, mechanism for the tumour-suppressive role of a plasma membrane transporter independent of its transport function.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas de Transporte de Catión/metabolismo , Membrana Celular/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Neoplasias Pancreáticas/patología , Animales , Apoptosis , Transporte Biológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Ratones , Ratones Desnudos , Transportadores de Ácidos Monocarboxílicos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Survivin , Trasplante Heterólogo
6.
PLoS One ; 7(1): e29916, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22238673

RESUMEN

Reporter-based assays underlie many high-throughput screening (HTS) platforms, but most are limited to in vitro applications. Here, we report a simple whole-organism HTS method for quantifying changes in reporter intensity in individual zebrafish over time termed, Automated Reporter Quantification in vivo (ARQiv). ARQiv differs from current "high-content" (e.g., confocal imaging-based) whole-organism screening technologies by providing a purely quantitative data acquisition approach that affords marked improvements in throughput. ARQiv uses a fluorescence microplate reader with specific detection functionalities necessary for robust quantification of reporter signals in vivo. This approach is: 1) Rapid; achieving true HTS capacities (i.e., >50,000 units per day), 2) Reproducible; attaining HTS-compatible assay quality (i.e., Z'-factors of ≥0.5), and 3) Flexible; amenable to nearly any reporter-based assay in zebrafish embryos, larvae, or juveniles. ARQiv is used here to quantify changes in: 1) Cell number; loss and regeneration of two different fluorescently tagged cell types (pancreatic beta cells and rod photoreceptors), 2) Cell signaling; relative activity of a transgenic Notch-signaling reporter, and 3) Cell metabolism; accumulation of reactive oxygen species. In summary, ARQiv is a versatile and readily accessible approach facilitating evaluation of genetic and/or chemical manipulations in living zebrafish that complements current "high-content" whole-organism screening methods by providing a first-tier in vivo HTS drug discovery platform.


Asunto(s)
Dosificación de Gen , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Pez Cebra , Animales , Animales Modificados Genéticamente , Automatización/métodos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Embrión no Mamífero , Dosificación de Gen/fisiología , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Concentración Osmolar , Reproducibilidad de los Resultados , Estudios de Validación como Asunto , Pez Cebra/embriología , Pez Cebra/genética
7.
J Biol Chem ; 286(36): 31830-8, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21771784

RESUMEN

SLC6A14, also known as ATB(0,+), is an amino acid transporter with unique characteristics. It transports 18 of the 20 proteinogenic amino acids. However, this transporter is expressed only at low levels in normal tissues. Here, we show that the transporter is up-regulated specifically in estrogen receptor (ER)-positive breast cancer, demonstrable with primary human breast cancer tissues and human breast cancer cell lines. SLC6A14 is an estrogen/ER target. The transport features of SLC6A14 include concentrative transport of leucine (an activator of mTOR), glutamine (an essential amino acid for nucleotide biosynthesis and substrate for glutaminolysis), and arginine (an essential amino acid for tumor cells), suggesting that ER-positive breast cancer cells up-regulate SLC6A14 to meet their increased demand for these amino acids. Consequently, treatment of ER-positive breast cancer cells in vitro with α-methyl-DL-tryptophan (α-MT), a selective blocker of SLC6A14, induces amino acid deprivation, inhibits mTOR, and activates autophagy. Prolongation of the treatment with α-MT causes apoptosis. Addition of an autophagy inhibitor (3-methyladenine) during α-MT treatment also induces apoptosis. These effects of α-MT are specific to ER-positive breast cancer cells, which express the transporter. The ability of α-MT to cause amino acid deprivation is significantly attenuated in MCF-7 cells, an ER-positive breast cancer cell line, when SLC6A14 is silenced with shRNA. In mouse xenograft studies, α-MT by itself is able to reduce the growth of the ER-positive ZR-75-1 breast cancer cells. These studies identify SLC6A14 as a novel and effective drug target for the treatment of ER-positive breast cancer.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Transporte de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Autofagia/efectos de los fármacos , Neoplasias de la Mama/patología , Femenino , Humanos , Ratones , Terapia Molecular Dirigida/métodos , Receptores de Estrógenos , Trasplante Heterólogo , Triptófano/análogos & derivados , Triptófano/farmacología , Células Tumorales Cultivadas
8.
Invest Ophthalmol Vis Sci ; 52(8): 5749-57, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21508099

RESUMEN

PURPOSE: To evaluate the role of SLC5A8 in the transport of 2-oxothiazolidine-4-carboxylate (OTC) and to determine whether OTC augments glutathione production in RPE cells, thereby providing protection against oxidative stress. METHODS: SLC5A8-mediated transport of OTC was monitored in Xenopus laevis oocytes by electrophysiological means. Saturation kinetics, Na(+)-activation kinetics, and inhibition by ibuprofen were analyzed by monitoring OTC-induced currents as a measure of transport activity. Oxidative stress was induced in ARPE-19 cells and primary RPE cells isolated from wild type and Slc5a8(-/-) mouse retinas using H(2)O(2), and the effects of OTC on cell death and intracellular glutathione concentration were examined. RESULTS: Heterologous expression of human SLC5A8 in X. laevis oocytes induced Na(+)-dependent inward currents in the presence of OTC under voltage-clamp conditions. The transport of OTC via SLC5A8 was saturable, with a K(t) of 104 ± 3 µM. The Na(+)-activation kinetics was sigmoidal with a Hill coefficient of 1.9 ± 0.1, suggesting involvement of two Na(+) in the activation process. Ibuprofen, a blocker of SLC5A8, inhibited SLC5A8-mediated OTC transport; the concentration necessary for half-maximal inhibition was 17 ± 1 µM. OTC increased glutathione levels and protected ARPE-19 and primary RPE cells isolated from wild type mouse retinas from H(2)O(2)-induced cell death. These effects were abolished in primary RPE isolated from Slc5a8(-/-) mouse retinas. CONCLUSIONS: OTC is a transportable substrate for SLC5A8. OTC augments glutathione production in RPE cells, thereby protecting them from oxidative damage. Transport via SLC5A8 is obligatory for this process.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Glutatión/metabolismo , Ácido Pirrolidona Carboxílico/farmacocinética , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Tiazolidinas/farmacocinética , Animales , Apoptosis , Transporte Biológico/fisiología , Proteínas de Transporte de Catión/genética , Supervivencia Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Interacciones Farmacológicas , Humanos , Peróxido de Hidrógeno/toxicidad , Ibuprofeno/farmacología , Cinética , Ratones , Ratones Mutantes , Transportadores de Ácidos Monocarboxílicos , Oocitos/fisiología , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ácido Pirrolidona Carboxílico/química , Epitelio Pigmentado de la Retina/citología , Tiazolidinas/química , Xenopus laevis
9.
Bioorg Med Chem ; 18(16): 5988-94, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20637638

RESUMEN

A natural product chemistry-based approach was applied to discover small-molecule inhibitors of hypoxia-inducible factor-1 (HIF-1). A Petrosaspongia mycofijiensis marine sponge extract yielded mycothiazole (1), a solid tumor selective compound with no known mechanism for its cell line-dependent cytotoxic activity. Compound 1 inhibited hypoxic HIF-1 signaling in tumor cells (IC(50) 1nM) that correlated with the suppression of hypoxia-stimulated tumor angiogenesis in vitro. However, 1 exhibited pronounced neurotoxicity in vitro. Mechanistic studies revealed that 1 selectively suppresses mitochondrial respiration at complex I (NADH-ubiquinone oxidoreductase). Unlike rotenone, MPP(+), annonaceous acetogenins, piericidin A, and other complex I inhibitors, mycothiazole is a mixed polyketide/peptide-derived compound with a central thiazole moiety. The exquisite potency and structural novelty of 1 suggest that it may serve as a valuable molecular probe for mitochondrial biology and HIF-mediated hypoxic signaling.


Asunto(s)
Complejo I de Transporte de Electrón/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Poríferos/química , Tiazoles/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Complejo I de Transporte de Electrón/metabolismo , Inhibidores Enzimáticos/aislamiento & purificación , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neuronas/efectos de los fármacos , Ratas , Tiazoles/aislamiento & purificación
10.
Clin Cancer Res ; 16(19): 4742-54, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20647473

RESUMEN

PURPOSE: Bortezomib induces unfolded protein response (UPR) and endoplasmic reticulum stress, as well as exhibits clinical activity in patients with relapsed and refractory mantle cell lymphoma (MCL). Here, we determined the molecular basis of the improved in vitro and in vivo activity of the combination of the pan-histone deacetylase inhibitor panobinostat and bortezomib against human, cultured, and primary MCL cells. EXPERIMENTAL DESIGN: Immunoblot analyses, reverse transcription-PCR, and immunofluorescent and electron microscopy were used to determine the effects of panobinostat on bortezomib-induced aggresome formation and endoplasmic reticulum stress in MCL cells. RESULTS: Treatment with panobinostat induced heat shock protein 90 acetylation; depleted the levels of heat shock protein 90 client proteins, cyclin-dependent kinase 4, c-RAF, and AKT; and abrogated bortezomib-induced aggresome formation in MCL cells. Panobinostat also induced lethal UPR, associated with induction of CAAT/enhancer binding protein homologous protein (CHOP). Conversely, knockdown of CHOP attenuated panobinostat-induced cell death of MCL cells. Compared with each agent alone, cotreatment with panobinostat increased bortezomib-induced expression of CHOP and NOXA, as well as increased bortezomib-induced UPR and apoptosis of cultured and primary MCL cells. Cotreatment with panobinostat also increased bortezomib-mediated in vivo tumor growth inhibition and improved survival of mice bearing human Z138C MCL cell xenograft. CONCLUSION: These findings suggest that increased UPR and induction of CHOP are involved in enhanced anti-MCL activity of the combination of panobinostat and bortezomib.


Asunto(s)
Ácidos Borónicos/farmacología , Retículo Endoplásmico/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Linfoma de Células del Manto/tratamiento farmacológico , Linfoma de Células del Manto/patología , Pirazinas/farmacología , Estrés Fisiológico/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Bortezomib , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Técnica del Anticuerpo Fluorescente , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Indoles , Linfoma de Células del Manto/metabolismo , Ratones , Microscopía Confocal , Panobinostat , Pliegue de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Factor de Transcripción CHOP/genética , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Nat Prod ; 73(5): 956-61, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20423107

RESUMEN

Products that contain twig extracts of pawpaw (Asimina triloba) are widely consumed anticancer alternative medicines. Pawpaw crude extract (CE) and purified acetogenins inhibited hypoxia-inducible factor-1 (HIF-1)-mediated hypoxic signaling pathways in tumor cells. In T47D cells, pawpaw CE and the acetogenins 10-hydroxyglaucanetin (1), annonacin (2), and annonacin A (3) inhibited hypoxia-induced HIF-1 activation with IC(50) values of 0.02 microg/mL, 12 nM, 13 nM, and 31 nM, respectively. This inhibition correlates with the suppression of the hypoxic induction of HIF-1 target genes VEGF and GLUT-1. The induction of secreted VEGF protein represents a key event in hypoxia-induced tumor angiogenesis. Both the extract and the purified acetogenins blocked the angiogenesis-stimulating activity of hypoxic T47D cells in vitro. Pawpaw extract and acetogenins inhibited HIF-1 activation by blocking the hypoxic induction of nuclear HIF-1alpha protein. The inhibition of HIF-1 activation was associated with the suppression of mitochondrial respiration at complex I. Thus, the inhibition of HIF-1 activation and hypoxic tumor angiogenesis constitutes a novel mechanism of action for these anticancer alternative medicines.


Asunto(s)
Acetogeninas/aislamiento & purificación , Acetogeninas/farmacología , Antineoplásicos Fitogénicos/aislamiento & purificación , Antineoplásicos Fitogénicos/farmacología , Asimina/química , Transportador de Glucosa de Tipo 1/efectos de los fármacos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Plantas Medicinales/química , Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Acetogeninas/química , Antineoplásicos Fitogénicos/química , Terapias Complementarias , Ensayos de Selección de Medicamentos Antitumorales , Transportador de Glucosa de Tipo 1/análisis , Transportador de Glucosa de Tipo 1/genética , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Factores de Crecimiento Endotelial Vascular/análisis , Factores de Crecimiento Endotelial Vascular/genética
12.
Mol Cancer Ther ; 9(4): 942-52, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20371724

RESUMEN

Increased levels of misfolded polypeptides in the endoplasmic reticulum (ER) triggers the dissociation of glucose-regulated protein 78 (GRP78) from the three transmembrane ER-stress mediators, i.e., protein kinase RNA-like ER kinase (PERK), activating transcription factor-6 (ATF6), and inositol-requiring enzyme 1alpha, which results in the adaptive unfolded protein response (UPR). In the present studies, we determined that histone deacetylase-6 (HDAC6) binds and deacetylates GRP78. Following treatment with the pan-histone deacetylase inhibitor panobinostat (Novartis Pharmaceuticals), or knockdown of HDAC6 by short hairpin RNA, GRP78 is acetylated in 11 lysine residues, which dissociates GRP78 from PERK. This is associated with the activation of a lethal UPR in human breast cancer cells. Coimmunoprecipitation studies showed that binding of HDAC6 to GRP78 requires the second catalytic and COOH-terminal BUZ domains of HDAC6. Treatment with panobinostat increased the levels of phosphorylated-eukaryotic translation initiation factor (p-eIF2alpha), ATF4, and CAAT/enhancer binding protein homologous protein (CHOP). Panobinostat treatment also increased the proapoptotic BIK, BIM, BAX, and BAK levels, as well as increased the activity of caspase-7. Knockdown of GRP78 sensitized MCF-7 cells to bortezomib and panobinostat-induced UPR and cell death. These findings indicate that enforced acetylation and decreased binding of GRP78 to PERK is mechanistically linked to panobinostat-induced UPR and cell death of breast cancer cells. Mol Cancer Ther; 9(4); 942-52. (c)2010 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Retículo Endoplásmico/patología , Proteínas de Choque Térmico/metabolismo , Ácidos Hidroxámicos/farmacología , Estrés Fisiológico/efectos de los fármacos , Acetilación/efectos de los fármacos , Secuencia de Aminoácidos , Dominio Catalítico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Retículo Endoplásmico/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Femenino , Técnicas de Silenciamiento del Gen , Proteínas de Choque Térmico/química , Histona Desacetilasa 6 , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Humanos , Indoles , Modelos Biológicos , Datos de Secuencia Molecular , Panobinostat , Unión Proteica/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , eIF-2 Quinasa/metabolismo
13.
J Nat Prod ; 72(12): 2104-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19921787

RESUMEN

The transcription factor hypoxia-inducible factor-1 (HIF-1) represents an important molecular target for anticancer drug discovery. In a T47D cell-based reporter assay, the Caulerpa spp. algal pigment caulerpin (1) inhibited hypoxia-induced as well as 1,10-phenanthroline-induced HIF-1 activation. The angiogenic factor vascular endothelial growth factor (VEGF) is regulated by HIF-1. Caulerpin (10 microM) suppressed hypoxic induction of secreted VEGF protein and the ability of hypoxic T47D cell-conditioned media to promote tumor angiogenesis in vitro. Under hypoxic conditions, 1 (10 microM) blocked the induction of HIF-1alpha protein, the oxygen-regulated subunit that controls HIF-1 activity. Reactive oxygen species produced by mitochondrial complex III are believed to act as a signal of cellular hypoxia that leads to HIF-1alpha protein induction and activation. Further mechanistic studies revealed that 1 inhibits mitochondrial respiration at electron transport chain (ETC) complex I (NADH-ubiquinone oxidoreductase). Under hypoxic conditions, it is proposed that 1 may disrupt mitochondrial ROS-regulated HIF-1 activation and HIF-1 downstream target gene expression by inhibiting the transport or delivery of electrons to complex III.


Asunto(s)
Caulerpa/química , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Indoles/farmacología , Colorantes/química , Colorantes/farmacología , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Femenino , Humanos , Indoles/química , Indoles/aislamiento & purificación , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estructura Molecular , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA