Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Exp Mol Med ; 53(4): 643-653, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33859351

RESUMEN

DNA damage-induced apoptosis suppressor (DDIAS) promotes the progression of lung cancer and hepatocellular carcinoma through the regulation of multiple pathways. We screened a chemical library for anticancer agent(s) capable of inhibiting DDIAS transcription. DGG-100629 was found to suppress lung cancer cell growth through the inhibition of DDIAS expression. DGG-100629 induced c-Jun NH(2)-terminal kinase (JNK) activation and inhibited NFATc1 nuclear translocation. Treatment with SP600125 (a JNK inhibitor) or knockdown of JNK1 restored DDIAS expression and reversed DGG-100629-induced cell death. In addition, DGG-100629 suppressed the signal transducer and activator of transcription (STAT3) signaling pathway. DDIAS or STAT3 overexpression restored lung cancer cell growth in the presence of DGG-100629. In a xenograft assay, DGG-100629 inhibited tumor growth by reducing the level of phosphorylated STAT3 and the expression of STAT3 target genes. Moreover, DGG-100629 inhibited the growth of lung cancer patient-derived gefitinib-resistant cells expressing NFATc1 and DDIAS. Our findings emphasize the potential of DDIAS blockade as a therapeutic approach and suggest a novel strategy for the treatment of gefitinib-resistant lung cancer.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Ciclo Celular/metabolismo , Factores de Transcripción NFATC/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Neoplasias Pulmonares , MAP Quinasa Quinasa 4/metabolismo , Modelos Biológicos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Med Chem ; 63(7): 3763-3783, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32189500

RESUMEN

The immunoproteasome (iP), an inducible proteasome variant harboring three immunosubunits, low molecular mass polypeptide-2 (LMP2), multicatalytic endopeptidase complex subunit-1, and low molecular mass polypeptide-7 (LMP7), is involved in multiple facets of inflammatory responses. We recently reported that YU102, a dual inhibitor of the iP subunit LMP2 and the constitutive proteasome catalytic subunit ß1, ameliorates cognitive impairments in mouse models of Alzheimer's disease (AD) independently of amyloid deposits. To investigate whether inhibition of LMP2 is sufficient to improve the cognitive functions of AD mice, here we prepared 37 YU102 analogues and identified a potent LMP2 inhibitor DB-310 (28) (IC50: 80.6 nM) with improved selectivity and permeability in cells overexpressing ABCB1 transporters. We show that DB-310 induces suppression of IL-1α production in microglia cells and improves cognitive functions in the Tg2576 transgenic mouse model of AD. This study supports that inhibition of LMP2 is a promising therapeutic strategy for treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/uso terapéutico , Nootrópicos/uso terapéutico , Oligopéptidos/uso terapéutico , Animales , Línea Celular Transformada , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/toxicidad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Interleucina-1alfa/metabolismo , Ratones Transgénicos , Microglía/efectos de los fármacos , Estructura Molecular , Nootrópicos/síntesis química , Nootrópicos/toxicidad , Oligopéptidos/síntesis química , Oligopéptidos/toxicidad , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Bibliotecas de Moléculas Pequeñas/toxicidad , Relación Estructura-Actividad
3.
Sci Rep ; 9(1): 18393, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804556

RESUMEN

The immunoproteasome (iP) is a variant of the constitutive proteasome (cP) that is abundantly expressed in immune cells which can also be induced in somatic cells by cytokines such as TNF-α or IFN-γ. Accumulating evidence support that the iP is closely linked to multiple facets of inflammatory response, eventually leading to the development of several iP inhibitors as potential therapeutic agents for autoimmune diseases. Recent studies also found that the iP is upregulated in reactive glial cells surrounding amyloid ß (Aß) deposits in brains of Alzheimer's disease (AD) patients, but the role it plays in the pathogenesis of AD remains unclear. In this study, we investigated the effects of several proteasome inhibitors on cognitive function in AD mouse models and found that YU102, a dual inhibitor of the iP catalytic subunit LMP2 and the cP catalytic subunit Y, ameliorates cognitive impairments in AD mouse models without affecting Aß deposition. The data obtained from our investigation revealed that YU102 suppresses the secretion of inflammatory cytokines from microglial cells. Overall, this study indicates that there may exist a potential link between LMP2/Y and microglia-mediated neuroinflammation and that inhibition of these subunits may offer a new therapeutic strategy for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Cisteína Endopeptidasas/genética , Neuroglía/efectos de los fármacos , Inhibidores de Proteasoma/farmacología , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/patología , Línea Celular , Disfunción Cognitiva/enzimología , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Cisteína Endopeptidasas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Proteínas Quimioatrayentes de Monocitos/genética , Proteínas Quimioatrayentes de Monocitos/metabolismo , Neuroglía/enzimología , Neuroglía/patología , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Bazo/efectos de los fármacos , Bazo/enzimología , Bazo/patología
4.
Int J Mol Sci ; 20(2)2019 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-30646562

RESUMEN

Cyclodextrins (CDs) have beneficial characteristics for drug delivery, including hydrophobic interior surfaces. Nanocarriers with ß-CD ligands have been prepared with simple surface modifications as drug delivery vehicles. In this study, we synthesized ß-CD derivatives on an Ag-embedded silica nanoparticle (NP) (SiO2@Ag NP) structure to load and release doxorubicin (DOX). Cysteinyl-ß-CD and ethylenediamine-ß-CD (EDA-ß-CD) were immobilized on the surface of SiO2@Ag NPs, as confirmed by transmission electron microscopy (TEM), ultraviolet-visible (UV-Vis) spectrophotometry, and Fourier transform infrared (FTIR) spectroscopy. DOX was introduced into the ß-CD on the SiO2@Ag NPs and then successfully released. Neither cysteinyl-ß-CD and EDA-ß-CD showed cytotoxicity, while DOX-loaded cysteinyl-ß-CD and EDA-ß-CD showed a significant decrease in cell viability in cancer cells. The SiO2@Ag NPs with ß-CD provide a strategy for designing a nanocarrier that can deliver a drug with controlled release from modified chemical types.


Asunto(s)
Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Nanopartículas/química , beta-Ciclodextrinas/química , Neoplasias de la Mama/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/química , Portadores de Fármacos/administración & dosificación , Femenino , Humanos , Células MCF-7 , Microscopía Electrónica de Transmisión , Nanopartículas/administración & dosificación , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/química , Plata/química , Espectroscopía Infrarroja por Transformada de Fourier , beta-Ciclodextrinas/administración & dosificación
5.
Bioconjug Chem ; 29(2): 528-537, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29376329

RESUMEN

Graphene oxide (GO) is known to strongly bind single-stranded nucleic acids with fluorescence quenching near the GO surface. However, GO exhibits weak biocompatibility characteristics, such as low dispersibility in cell culture media and significant cytotoxicity. To improve dispersibility in cell culture media and cell viability of GO, we prepared nanosized GO (nGO) constructs and modified the nGO surface using polyethylene glycol (PEG-nGO). Single-stranded peptide nucleic acid (PNA) was adsorbed onto the PEG-nGO and was readily desorbed by adding complementary RNA or under low pH conditions. PNA adsorbed on the PEG-nGO was efficiently delivered into lung cancer cells via endocytosis without affecting cell viability. Furthermore, antisense PNA delivered using PEG-nGO effectively downregulated the expression of the target gene in cancer cells. Our results suggest that PEG-nGO is a biocompatible carrier useful for PNA delivery into cells and serves as a promising gene delivery tool.


Asunto(s)
Materiales Biocompatibles/química , Portadores de Fármacos/química , Grafito/química , Nanoestructuras/química , Ácidos Nucleicos de Péptidos/administración & dosificación , Polietilenglicoles/química , Línea Celular Tumoral , Endocitosis , Humanos , Nanoestructuras/ultraestructura , Óxidos/química , Ácidos Nucleicos de Péptidos/farmacocinética , Propiedades de Superficie
6.
BMB Rep ; 51(1): 27-32, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28893372

RESUMEN

Non-small-cell lung cancer (NSCLC) is commonly caused by a mutation in the epidermal growth factor receptor (EGFR) and subsequent aberrant EGFR signaling with uncontrolled kinase activity. A deletion mutation in EGFR exon 19 is frequently observed in EGFR gene mutations. We designed a DNAzyme to suppress the expression of mutant EGFR by cleaving the mutant EGFR mRNA. The DNAzyme (named Ex19del Dz) specifically cleaved target RNA and decreased cancer cell viability when transfected into gefitinib-resistant lung cancer cells harboring EGFR exon 19 deletions. The DNAzyme decreased EGFR expression and inhibited its downstream signaling pathway. In addition to EGFR downregulation, Ex19del Dz containing CpG sites activated Toll-like receptor 9 (TLR9) and its downstream signaling pathway via p38 kinase, causing an immunostimulatory effect on EGFR-mutated NSCLC cells. Thus, dual effects of this DNAzyme harboring the CpG site, such as TLR9 activation and EGFR downregulation, leads to apoptosis of EGFR-mutated NSCLC cells. [BMB Reports 2018; 51(1): 27-32].


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Islas de CpG , ADN Catalítico/farmacología , Receptores ErbB/genética , Neoplasias Pulmonares/tratamiento farmacológico , Receptor Toll-Like 9/metabolismo , Secuencia de Bases , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/genética , ADN Catalítico/genética , ADN Catalítico/metabolismo , Regulación hacia Abajo , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Exones , Gefitinib , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mutación , Quinazolinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Toll-Like 9/biosíntesis , Receptor Toll-Like 9/genética
7.
Autophagy ; 13(2): 248-263, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28045574

RESUMEN

Contribution of autophagy and regulation of related proteins to the degeneration of retinal pigment epithelium (RPE) in age-related macular degeneration (AMD) remain unknown. We report that upregulation of KRT8 (keratin 8) as well as its phosphorylation are accompanied with autophagy and attenuated with the inhibition of autophagy in RPE cells under oxidative stress. KRT8 appears to have a dual role in RPE pathophysiology. While increased expression of KRT8 following autophagy provides a cytoprotective role in RPE, phosphorylation of KRT8 induces pathologic epithelial-mesenchymal transition (EMT) of RPE cells under oxidative stress, which is mediated by MAPK1/ERK2 (mitogen-activated protein kinase 1) and MAPK3/ERK1. Inhibition of autophagy further promotes EMT, which can be reversed by inhibition of MAPK. Thus, regulated enhancement of autophagy with concurrent increased expression of KRT8 and the inhibition of KRT8 phosphorylation serve to inhibit oxidative stress-induced EMT of RPE cells as well as to prevent cell death, suggesting that pharmacological manipulation of KRT8 upregulation through autophagy with combined inhibition of the MAPK1/3 pathway may be attractive therapeutic strategies for the treatment of AMD.


Asunto(s)
Autofagia , Queratina-8/metabolismo , Estrés Oxidativo , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Apoptosis , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Línea Celular , Movimiento Celular , Núcleo Celular/metabolismo , Citoprotección , Transición Epitelial-Mesenquimal , Humanos , Lisosomas/metabolismo , Lisosomas/ultraestructura , Fusión de Membrana , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Modelos Biológicos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Epitelio Pigmentado de la Retina/ultraestructura , Transducción de Señal , Superóxidos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Regulación hacia Arriba
8.
Proteomics ; 8(22): 4748-67, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18991268

RESUMEN

The endogenous ROS levels were increased during HepG2 apoptosis, whereas they were decreased during SK-N-SH apoptosis in response to capsaicin treatments. We used 2-DE-based proteomics to analyze the altered protein levels in both cells, with special attention on oxidative stress proteins before and after capsaicin treatments. The 2-DE analysis demonstrated that 23 proteins were increased and 26 proteins were decreased significantly (fold change>1.4) in capsaicin-treated apoptotic HepG2 and SK-N-SH cells, respectively. The distinct effect of capsaicin-induced apoptosis on the expression pattern of HepG2 proteins includes the downregulation of some antioxidant enzymes including aldose reductase (AR), catalase, enolase 1, peroxiredoxin 1, but upregulation of peroxiredoxin 6, cytochrome c oxidase, and SOD2. In contrast, most antioxidant enzymes were increased in SK-N-SH cells in response to capsaicin, where catalase might play a pivotal role in maintenance of low ROS levels in the course of apoptosis. The global gene expression for oxidative stress and antioxidant defense genes revealed that 84 gene expressions were not significantly different in HepG2 cells between control and capsaicin-treated cells. In contrast, a number of oxidative genes were downregulated in SK-N-SH cells, supporting the evidence of low ROS environment in apoptotic SK-N-SH cells after capsaicin treatment. It was concluded that the different relationship between endogenous ROS levels and apoptosis of two cancer cells presumably resulted from complicated expression patterns of many oxidative stress and antioxidant genes, rather than the individual role of some classical antioxidant enzymes such as SOD and catalase.


Asunto(s)
Apoptosis , Capsaicina/farmacología , Carcinoma Hepatocelular/metabolismo , Proteínas de Neoplasias/análisis , Neuroblastoma/metabolismo , Proteómica/métodos , Análisis de Varianza , Antioxidantes/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Electroforesis en Gel Bidimensional , Regulación de la Expresión Génica , Glutatión/análisis , Humanos , Proteínas de Neoplasias/genética , Neuroblastoma/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo , Oxidorreductasas/análisis , Oxidorreductasas/genética , Especies Reactivas de Oxígeno/análisis , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
9.
J Microbiol Biotechnol ; 18(3): 512-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18388470

RESUMEN

Three different polysaccharide-peptide complexes (PPC, named as Fr-I, Fr-II, and Fr-III) were produced by submerged mycelial culture of an entomopathogenic fungus Cordyceps sphecocephala, and their anticancer activities were investigated in human hepatocarcinoma (HepG2) and neuroblastoma (SK-N-SH) cells. The highest inhibitory effects of PPC on both HepG2 and SK-N-SH cells were achieved with Fr-I, whereas Fr-III with low molecular mass showed lower inhibition effects. Interestingly, the inhibitory effects of the three fractions were increased after protease digestion, suggesting that the inhibitory effects resulted mainly from the carbohydrate moiety, at least in the case of Fr-II and Fr-III, of PPC. The results of DNA fragmentation in PPC-induced apoptotic cells were confirmed by both DNA ladder assay and comet assay. Our investigation also showed that PPC-induced apoptosis of both cancer cells was associated with intracellular events including DNA fragmentation, activation of caspase-3, and modulation of Bcl-2 and Bax. We conclude that PPC has potential as a novel therapeutic agent for the treatment of both HepG2 and SK-N-SH cancer cells without any cytotoxicity against normal cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Abejas/microbiología , Cordyceps/metabolismo , Micelio/metabolismo , Proteoglicanos/farmacología , Animales , Anticarcinógenos/química , Anticarcinógenos/aislamiento & purificación , Anticarcinógenos/farmacología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cordyceps/crecimiento & desarrollo , Fragmentación del ADN/efectos de los fármacos , Humanos , Corea (Geográfico) , Micelio/crecimiento & desarrollo , Proteoglicanos/química , Proteoglicanos/aislamiento & purificación
10.
Appl Microbiol Biotechnol ; 78(3): 419-29, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18188554

RESUMEN

In the present study, optimum culture conditions for the production of extracellular polysaccharides (EPS) in submerged culture of an edible mushroom, Laetiporus sulphureus var. miniatus and their stimulatory effects on insulinoma cell (RINm5F) proliferation and insulin secretion were investigated. The maximum mycelial growth (4.1 g l(-1)) and EPS production (0.6 g l(-1)) in submerged flask culture were achieved in a medium containing 30 g l(-1) maltose, 2 g l(-1) soy peptone, and 2 mM MnSO(4).5H2O at an initial pH 2.0 and temperature 25 degrees C. In the stirred-tank fermenter under optimized medium, the concentrations of mycelial biomass and EPS reached a maximum level of 8.1 and 3.9 g l(-1), respectively. Interestingly, supplementation of deep sea water (DSW) into the culture medium significantly increased both mycelial biomass and EPS production by 4- and 6.7-fold at 70% (v/v) DSW medium, respectively. The EPS were proved to be glucose-rich polysaccharides and were able to increase proliferation and insulin secretary function of rat insulinoma RINm5F cells, in a dose-dependent manner. In addition, EPS also strikingly reduced the streptozotocin-induced apoptosis in RINm5F cells indicating the mode of the cytoprotective role of EPS on RINm5F cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Insulina/metabolismo , Micelio/metabolismo , Polyporales/metabolismo , Polisacáridos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Reactores Biológicos , Carbono/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Concentración de Iones de Hidrógeno , Minerales/metabolismo , Micelio/crecimiento & desarrollo , Nitrógeno/metabolismo , Polyporales/crecimiento & desarrollo , Polisacáridos/química , Polisacáridos/farmacología , Ratas , Estreptozocina/farmacología , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA