Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncol Rep ; 39(6): 2482-2498, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29620220

RESUMEN

Using a series of potential biomarkers relevant to mechanisms of protein synthesis, we observed that estrogen receptor (ER)-positive breast tumor cells exist in two distinct yet interconvertible phenotypic states (of roughly equal proportion) which differ in the degree of differentiation and use of IRES-mediated translation. Nascently translated IGF1R in the cytoplasm positively correlated with IRES activity and the undifferentiated phenotype, while epitope accessibility of RACK1, an integral component of the 40S ribosomal subunit, aligned with the more differentiated IRES-off state. When deprived of soluble growth factors, the entire tumor cell population shifted to the undifferentiated phenotype in which IRES-mediated translation was active, facilitating survival under these adverse microenvironmental conditions. However, if IRES-mediated translation was inhibited, the cells instead were forced to transition uniformly to the more differentiated state. Notably, cytoplasmic localization of estrogen receptor α (ERα/ESR1) precisely mirrored the pattern observed with nascent IGF1R, correlating with the undifferentiated IRES-active phenotype. Inhibition of IRES-mediated translation resulted in both a shift in ERα to the nucleus (consistent with differentiation) and a marked decrease in ERα abundance (consistent with the inhibition of ERα synthesis via its IRES). Although breast tumor cells tolerated forced differentiation without extensive loss of their viability, their reproductive capacity was severely compromised. In addition, CDK1 was decreased, connexin 43 eliminated and Myc translation altered as a consequence of IRES inhibition. Isolated or low-density ER-positive breast tumor cells were particularly vulnerable to IRES inhibition, losing the ability to generate viable cohesive colonies, or undergoing massive cell death. Collectively, these results provide further evidence for the integral relationship between IRES-mediated translation and the undifferentiated phenotype and demonstrate how therapeutic manipulation of this specialized mode of protein synthesis may be used to limit the phenotypic plasticity and incapacitate or eliminate these otherwise highly resilient breast tumor cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Citoplasma/metabolismo , Receptor alfa de Estrógeno/metabolismo , Sitios Internos de Entrada al Ribosoma/efectos de los fármacos , Receptores de Somatomedina/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Neoplasias de la Mama/genética , Diferenciación Celular , Línea Celular Tumoral , Femenino , Humanos , Proteínas de Neoplasias/metabolismo , Fenotipo , Biosíntesis de Proteínas , Transporte de Proteínas , Receptor IGF Tipo 1 , Receptores de Cinasa C Activada/metabolismo
2.
JCI Insight ; 1(15): e87536, 2016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27699245

RESUMEN

Chronic obstructive pulmonary disease (COPD) is the third leading cause of death in the US. The majority of COPD patients have symptoms of chronic bronchitis, which lacks specific therapies. A major impediment to therapeutic development has been the absence of animal models that recapitulate key clinical and pathologic features of human disease. Ferrets are well suited for the investigation of the significance of respiratory diseases, given prior data indicating similarities to human airway physiology and submucosal gland distribution. Here, we exposed ferrets to chronic cigarette smoke and found them to approximate complex clinical features of human COPD. Unlike mice, which develop solely emphysema, smoke-exposed ferrets exhibited markedly higher numbers of early-morning spontaneous coughs and sporadic infectious exacerbations as well as a higher level of airway obstruction accompanied by goblet cell metaplasia/hyperplasia and increased mucus expression in small airways, indicative of chronic bronchitis and bronchiolitis. Overall, we demonstrate the first COPD animal model exhibiting clinical and pathologic features of chronic bronchitis to our knowledge, providing a key advance that will greatly facilitate the preclinical development of novel treatments for this disease.


Asunto(s)
Bronquitis Crónica/fisiopatología , Modelos Animales de Enfermedad , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Animales , Bronquitis Crónica/inducido químicamente , Femenino , Hurones , Humanos , Pulmón/fisiopatología , Masculino , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Humo/efectos adversos
3.
Cancer Biol Ther ; 15(8): 1053-60, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25084100

RESUMEN

TRA-8, a monoclonal antibody targeting death receptor, has demonstrated high therapeutic effect for triple negative breast cancer (TNBC) in preclinical models. Tamoxifen, the standard of care for ERα-positive breast cancer, induces apoptosis via ERß, which commonly presents in TNBC cells. The current study investigates the combination effects of TRA-8 and tamoxifen for TNBC. In vitro assays were implemented with two ERß-positive TNBC cell lines, SUM159 and 2LMP, and in vivo therapy studies were followed using orthotopic breast tumor mouse models. IC50 of tamoxifen for SUM159 and 2LMP were 29 µM and 38 µM, respectively. Synergy between TRA-8 (0-1000 ng/mL) and tamoxifen (20 µM) was observed for both the cell lines. Tamoxifen (400 mg/kg diet) markedly suppressed the growth of SUM159 tumors for 6 weeks after therapy initiation, but it did not induce antitumor effect for 2LMP tumors. TRA-8 (0.1 mg, weekly, i.p.) successfully arrested the growth of both SUM159 and 2LMP tumors during therapy, but an antagonistic effect was observed when tamoxifen was combined. TRA-8 uptake into tumors was not changed by tamoxifen treatment. Histological analysis confirmed that caspase-3 activation induced by TRA-8 was significantly decreased when tamoxifen was used in combination. In conclusion, our findings suggest that the combined use of TRA-8 and tamoxifen may cause antagonistic effects for TNBC patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/administración & dosificación , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Concentración 50 Inhibidora , Ratones Desnudos , Multimerización de Proteína , Tamoxifeno/administración & dosificación
4.
Mol Imaging Biol ; 16(1): 85-94, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23836505

RESUMEN

PURPOSE: To examine the antagonistic effects of anti-extracellular matrix metalloprotease inducer (anti-EMMPRIN) antibody when combined with chemotherapy using a hypovascular pancreatic tumor model. PROCEDURES: Severely compromised immunodeficient mice bearing orthotopic MIA PaCa-2 tumors were used (five to six animals per group). Dynamic contrast-enhanced magnetic resonance imaging was used to examine the relationship between tumor vascularity and size. Therapy was initiated when tumors were hypovascular. Treatments included: (1) gemcitabine alone, (2) anti-EMMPRIN antibody alone, and (3) combination, each for 2 weeks. Additionally, another treatment arm included ß-lapachone, an NAD(P)H/quinone 1 (NQO1) bioactivated agent. (18)F-fluoro-D-glucose-positron emission tomography/computed tomography imaging was used weekly to monitor therapeutic effects. RESULTS: Gemcitabine or anti-EMMPRIN monotherapy significantly delayed tumor growth, but the combination therapy showed an antagonistic effect. Similarly, tumor growth was significantly suppressed by ß-lapachone alone, and additive effects were noted when combined with gemcitabine, but the therapeutic efficacy was reduced when anti-EMMPRIN antibody was added. CONCLUSIONS: Anti-EMMPRIN antibody with chemotherapy in hypovascular tumors results in antagonistic effects.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Basigina/inmunología , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Desoxiglucosa , Sistemas de Liberación de Medicamentos , Femenino , Ratones , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/inmunología , Tomografía de Emisión de Positrones , Carga Tumoral/efectos de los fármacos , Gemcitabina
5.
Mol Cancer Ther ; 11(2): 405-15, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22203731

RESUMEN

The goal of the study was to assess the efficacy of combined extracellular matrix metalloprotease inducer (EMMPRIN)- and death receptor 5 (DR5)-targeted therapy for pancreatic adenocarcinoma in orthotopic mouse models with multimodal imaging. Cytotoxicity of anti-EMMPRIN antibody and anti-DR5 antibody (TRA-8) in MIA PaCa-2 and PANC-1 cell lines was measured by ATPlite assay in vitro. The distributions of Cy5.5-labeled TRA-8 and Cy3-labeled anti-EMMPRIN antibody in the 2 cell lines were analyzed by fluorescence imaging in vitro. Groups 1 to 12 of severe combined immunodeficient mice bearing orthotopic MIA PaCa-2 (groups 1-8) or PANC-1 (groups 9-12) tumors were used for in vivo studies. Dynamic contrast-enhanced-MRI was applied in group 1 (untreated) or group 2 (anti-EMMPRIN antibody). The tumor uptake of Tc-99m-labeled TRA-8 was measured in group 3 (untreated) and group 4 (anti-EMMPRIN antibody). Positron emission tomography/computed tomography imaging with (18)F-FDG was applied in groups 5 to 12. Groups 5 to 8 (or groups 9 to 12) were untreated or treated with anti-EMMPRIN antibody, TRA-8, and combination, respectively. TRA-8 showed high killing efficacy for both MIA PaCa-2 and PANC-1 cells in vitro, but additional anti-EMMPRIN treatment did not improve the cytotoxicity. Cy5.5-TRA-8 formed cellular caps in both the cell lines, whereas the maximum signal intensity was correlated with TRA-8 cytotoxicity. Anti-EMMPRIN therapy significantly enhanced the tumor delivery of the MR contrast agent, but not Tc-99m-TRA-8. Tumor growth was significantly suppressed by the combination therapy, and the additive effect of the combination was shown in both MIA PaCa-2 and PANC-1 tumor models.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Basigina/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Basigina/metabolismo , Carbocianinas/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Fluorodesoxiglucosa F18/farmacocinética , Humanos , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Microscopía Fluorescente/métodos , Imagen Multimodal/métodos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Tomografía de Emisión de Positrones , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Tomografía Computarizada por Rayos X , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Virol ; 80(15): 7308-15, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16840311

RESUMEN

Previous studies have described in vitro serial passage of a Deltagamma(1)34.5 herpes simplex virus type 1 (HSV-1) strain in SK-N-SH neuroblastoma cells and selection of mutants that have acquired the ability to infect and replicate in this previously nonpermissive cell line. Here we describe the selection of a mutant HSV-1 strain by in vivo serial passage, which prolongs survival in two separate experimental murine brain tumor models. Two conditionally replication-competent Deltagamma(1)34.5 viruses, M002, which expresses murine interleukin-12, and its parent virus, R3659, were serially passaged within human malignant glioma D54-MG cell lines in vitro or flank tumor xenografts in vivo. The major findings are (i) viruses passaged in vivo demonstrate decreased neurovirulence, whereas those passaged in vitro demonstrate a partial recovery of the neurovirulence associated with HSV-1; and (ii) vvD54-M002, the virus selected after in vivo serial passage of M002 in D54-MG tumors, improves survival in two independent murine brain tumor models compared to the parent (unpassaged) M002. Additionally, in vitro-passaged, but not in vivo-passaged, M002 displayed changes in the protein synthesis profile in previously nonpermissive cell lines, as well as early U(S)11 transcription. Thus, a mutant HSV-1 strain expressing a foreign gene can be selected for enhanced antitumor efficacy via in vivo serial passage within flank D54-MG tumor xenografts. The enhanced antitumor efficacy of vvD54-M002 is not due to restoration of protein synthesis or early U(S)11 expression. This finding emphasizes the contribution of the in vivo tumor environment for selecting novel oncolytic HSV specifically adapted for tumor cell destruction in vivo.


Asunto(s)
Neoplasias Encefálicas/virología , Glioma/virología , Herpesvirus Humano 1/patogenicidad , Neuroblastoma/terapia , Virulencia/fisiología , Animales , Chlorocebus aethiops , Efecto Citopatogénico Viral , Herpesvirus Humano 1/fisiología , Humanos , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos A , Ratones Endogámicos CBA , Ratones SCID , Mutación/genética , Neoplasias Experimentales/virología , Neuroblastoma/metabolismo , Neuroblastoma/virología , Trasplante Heterólogo , Células Tumorales Cultivadas , Células Vero , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...