Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Cancer ; 17(1): 622, 2017 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-28874135

RESUMEN

BACKGROUND: Replication-competent adenoviruses (Ad) produced cytotoxic effects on infected tumors and have been examined for the clinical applicability. A biomarkers to predict the cytotoxicity is valuable in a clinical setting. METHODS: We constructed type 5 Ad (Ad5) of which the expression of E1A gene was activated by a 5' regulatory sequences of survivin, midkine or cyclooxygenase-2, which were highly expressed in human tumors. We also produced the same replication-competent Ad of which the fiber-knob region was replaced by that of Ad35 (AdF35). The cytotoxicity was examined by a colorimetric assay with human tumor cell lines, 4 kinds of pancreatic, 9 esophageal carcinoma and 5 mesothelioma. Ad infectivity and Ad-mediated gene expression were examined with replication-incompetent Ad5 and AdF35 which expressed the green fluorescence protein gene. Expression of cellular receptors for Ad5 and AdF35 was also examined with flow cytometry. A transcriptional activity of the regulatory sequences was investigated with a luciferase assay in the tumor cells. We then investigated a possible correlation between Ad-mediated cytotoxicity and the infectivity/gene expression, the transcriptional activity or the p53 genotype. RESULTS: We found that the cytotoxicity was greater with AdF35 than with Ad5 vectors, but was not correlated with the Ad infectivity/gene expression irrespective of the fiber-knob region or the E1A-activating transcriptional activity. In contrast, replication-competent Ad produced greater cytotoxicity in p53 mutated than in wild-type esophageal carcinoma cells, suggesting a possible association between the cytotoxicity and the p53 genotype. CONCLUSIONS: Sensitivity to Ad-mediated cytotoxic activity was linked with the p53 genotype but was not lineally correlated with the infectivity/gene expression or the E1A expression.


Asunto(s)
Adenoviridae/genética , Adenoviridae/metabolismo , Proteínas E1A de Adenovirus/metabolismo , Expresión Génica , Vectores Genéticos/genética , Secuencias Reguladoras de Ácidos Nucleicos , Proteína p53 Supresora de Tumor/genética , Replicación Viral , Línea Celular Tumoral , Efecto Citopatogénico Viral , Genes Reporteros , Genotipo , Humanos , Unión Proteica , Receptores Virales/genética , Receptores Virales/metabolismo , Activación Transcripcional , Transducción Genética , Transgenes , Proteína p53 Supresora de Tumor/metabolismo
2.
BMC Cancer ; 16: 455, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27405588

RESUMEN

BACKGROUND: Approximately 80 % of mesothelioma specimens have the wild-type p53 gene, whereas they contain homozygous deletions in the INK4A/ARF locus that encodes p14 (ARF) and the 16 (INK4A) genes. Consequently, the majority of mesothelioma is defective of the p53 pathways. We examined whether zoledronic acid (ZOL), a third generation bisphosphonate, and adenoviruses with a deletion of the E1B-55kD gene (Ad-delE1B55), which augments p53 levels in the infected tumors, could produce combinatory anti-tumor effects on human mesothelioma cells bearing the wild-type p53 gene. METHODS: Cytotoxicity of ZOL and Ad-delE1B55 was assessed with a WST assay. Cell cycle changes were tested with flow cytometry. Expression levels of relevant molecules were examined with western blot analysis to investigate a possible mechanism of cytotoxicity. Furthermore, the expressions of Ad receptors on target cells and infectivity were estimated with flow cytometry. Viral replication was assayed with the tissue culture infection dose method. RESULTS: A combinatory use of ZOL and Ad-delE1B55 suppressed cell growth and increased sub-G1 or S-phase populations compared with a single agent, depending on cells tested. The combinatory treatment up-regulated p53 levels and subsequently enhanced the cleavage of caspase-3, 8, 9 and poly (ADP-ribose) polymerase, but expression of molecules involved in autophagy pathways were inconsistent. ZOL-treated cells also increased Ad infectivity with a dose-dependent manner and augmented Ad replication although the expression levels of integrin molecules, one of the Ad receptors, were down-regulated. CONCLUSIONS: These findings indicated that ZOL and Ad-delE1B55 achieved combinatory anti-tumor effects through augmented apoptotic pathways or increased viral replication.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Difosfonatos/farmacología , Imidazoles/farmacología , Mesotelioma/terapia , Proteína p53 Supresora de Tumor/metabolismo , Células A549 , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Células HEK293 , Humanos , Cavidad Pleural/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Receptores Virales/genética , Proteína p53 Supresora de Tumor/genética , Ácido Zoledrónico
3.
J Cancer Res Clin Oncol ; 142(1): 27-36, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26077666

RESUMEN

PURPOSE: Activation-induced cytidine deaminase (AID) is involved in somatic hypermutation and class switch recombination processes in the antibody formation. The AID activity induces gene mutations and could be associated with transformation processes of B cells. Nevertheless, the relation between AID expression and the prognosis of B cell lymphoma patients remains uncharacterized. METHODS: We examined expression levels of the AID gene in 89 lymph node specimens from lymphoma and non-lymphoma patients with Northern blot analysis and investigated an association with their survival. RESULTS: The AID gene was preferentially expressed in B cell lymphoma in particular in diffuse large B cell lymphoma and follicular lymphoma. We confirmed AID protein expression in the mRNA-positive but not in the negative specimens with Western blot analysis and immunohistochemical staining. Survival of the patients treated with cyclophosphamide-/doxorubicin-/vincristine-/prednisone-based chemotherapy demonstrated that the prognosis of diffuse large B cell patients was unfavorable in the mRNA-positive group compared with the negative group, and that AID expression levels were correlated with the poor prognosis. In contrast, AID expression was not linked with the prognosis of follicular lymphoma patients. CONCLUSIONS: AID expression is a predictive marker for an unfavorable outcome in DLBCL patients treated with the chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/metabolismo , Citidina Desaminasa/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Northern Blotting , Western Blotting , Ciclofosfamida/uso terapéutico , Citidina Desaminasa/genética , Doxorrubicina/uso terapéutico , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/mortalidad , Linfoma de Células B Grandes Difuso/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Prednisona/uso terapéutico , Pronóstico , Tasa de Supervivencia , Vincristina/uso terapéutico , Adulto Joven
4.
Apoptosis ; 20(12): 1587-98, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26373551

RESUMEN

Pancreatic carcinoma is relatively resistant to chemotherapy and cell death induced by replication of adenoviruses (Ad) can be one of the therapeutic options. Transduction efficacy of conventional type 5 Ad (Ad5) is however low and the cytotoxic mechanism by replication-competent Ad was not well understood. We constructed replication-competent Ad5 of which the E1A promoter region was replaced with a transcriptional regulatory region of the midkine, the survivin or the cyclooxygenase-2 gene, all of which were expressed at a high level in human tumors. We also prepared replication-competent Ad5 that were activated with the same region but had the type 35 Ad-derived fiber-knob region (AdF35) to convert the major cellular receptor for Ad infection from the coxsackie adenovirus receptor to CD46 molecules. Replication-competent AdF35 that were activated with the exogenous region produced cytotoxic effects on human pancreatic carcinoma cells greater than the corresponding Ad5 bearing with the same regulatory region. Cells infected with the AdF35 showed cytopathic effects and increased sub-G1 fractions. Caspase-9, less significantly caspase-8 and poly (ADP-ribose) polymerase, but not caspase-3 was cleaved and expression of molecules involved in autophagy and caspase-independent cell death pathways remained unchanged. Nevertheless, H2A histone family member X molecules were phosphorylated, and N-acetyl-L-cystein, an inhibitor for reactive oxygen species, suppressed the AdF35-mediated cytotoxicity. These data indicated a novel mechanism of Ad-mediated cell death and suggest a possible clinical application of the fiber-knob modified Ad.


Asunto(s)
Adenoviridae/genética , Neoplasias Pancreáticas/virología , Especies Reactivas de Oxígeno/metabolismo , Replicación Viral/genética , Acetilcisteína/metabolismo , Caspasas/metabolismo , Muerte Celular/fisiología , Línea Celular , Línea Celular Tumoral , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Ciclooxigenasa 2/metabolismo , Células HEK293 , Humanos , Proteína Cofactora de Membrana/metabolismo , Neoplasias Pancreáticas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Regiones Promotoras Genéticas/genética , Transducción Genética/métodos , Neoplasias Pancreáticas
5.
BMC Cancer ; 15: 464, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26059686

RESUMEN

BACKGROUND: Improvement of transduction and augmentation of cytotoxicity are crucial for adenoviruses (Ad)-mediated gene therapy for cancer. Down-regulated expression of type 5 Ad (Ad5) receptors on human tumors hampered Ad-mediated transduction. Furthermore, a role of the p53 pathways in cytotoxicity mediated by replication-competent Ad remained uncharacterized. METHODS: We constructed replication-competent Ad5 of which the E1 region genes were activated by a transcriptional regulatory region of the midkine or the survivin gene, which is expressed preferentially in human tumors. We also prepared replication-competent Ad5 which were regulated by the same region but had a fiber-knob region derived from serotype 35 (AdF35). We examined the cytotoxicity of these Ad and a possible combinatory use of the replication-competent AdF35 and Ad5 expressing the wild-type p53 gene (Ad5/p53) in esophageal carcinoma cells. Expression levels of molecules involved in cell death, anti-tumor effects in vivo and production of viral progenies were also investigated. RESULTS: Replication-competent AdF35 in general achieved greater cytotoxic effects to esophageal carcinoma cells than the corresponding replication-competent Ad5. Infection with the AdF35 induced cleavages of caspases and increased sub-G1 fractions, but did not activate the autophagy pathway. Transduction with Ad5/p53 in combination with the replication-competent AdF35 further enhanced the cytotoxicity in a synergistic manner. We also demonstrated the combinatory effects in an animal model. Transduction with Ad5/p53 however suppressed production of replication-competent AdF35 progenies, but the combination augmented Ad5/p53-mediated p53 expression levels and the downstream pathways. CONCLUSIONS: Combination of replication-competent AdF35 and Ad5/p53 achieved synergistic cytotoxicity due to enhanced p53-mediated apoptotic pathways.


Asunto(s)
Adenoviridae/genética , Carcinoma/genética , Neoplasias Esofágicas/genética , Terapia Genética , Proteína p53 Supresora de Tumor/genética , Apoptosis/genética , Carcinoma/patología , Carcinoma/terapia , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/terapia , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Humanos , ARN Mensajero/biosíntesis , Transducción Genética , Proteína p53 Supresora de Tumor/biosíntesis
6.
Tumour Biol ; 36(10): 8137-45, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25990458

RESUMEN

Type 5 adenoviruses expressing mda-7 gene (Ad-mda-7) induced cell death in various kinds of human tumors, but pancreatic carcinoma cells were relatively resistant to Ad-mda-7-mediated cytotoxicity. We then examined whether infection of Ad-mda-7 together with replication-competent Ad produced combinatory cytotoxic effects. We prepared replication-competent Ad, defective of the E1B55kDa gene or activated by a transcriptional regulatory region of the midkine or the survivin gene of which the expression was up-regulated in human tumors. Type 5 Ad bearing the exogenous regulatory region were further modified by replacing the fiber-knob region with that of type 35 Ad. Pancreatic carcinoma cells were infected with replication-incompetent Ad-mda-7 and the replication-competent Ad. Combinatory effects were examined with the CalcuSyn software and cell cycle analyses. Ad-mda-7 and the replication-competent Ad achieved cytotoxicity to pancreatic carcinoma. A combinatory use of Ad-mda-7 and either Ad defective of the E1B55kDa gene or Ad activated by the regulatory region produced synergistic cytotoxic effects. Cell cycle analyses demonstrated that the combination increased sub-G1 populations. These data collectively suggest that expression of MDA-7 augments cytotoxicity of replication-competent Ad and achieves adjuvant effects on Ad-mediated cell death.


Asunto(s)
Adenoviridae/fisiología , Apoptosis , Interleucinas/genética , Neoplasias Pancreáticas/terapia , Replicación Viral , Western Blotting , Ciclo Celular , Proliferación Celular , Vectores Genéticos/administración & dosificación , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Células Tumorales Cultivadas , Neoplasias Pancreáticas
7.
BMC Cancer ; 14: 713, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25255777

RESUMEN

BACKGROUND: Transduction of human mesenchymal stem cells (MSCs) with type 5 adenoviruses (Ad5) is limited in the efficacy because of the poor expression level of the coxsackie adenovirus receptor (CAR) molecules. We examined a possible improvement of Ad-mediated gene transfer in MSCs by substituting the fiber region of type 5 Ad with that of type 35 Ad. METHODS: Expression levels of CAR and CD46 molecules, which are the major receptors for type 5 and type 35 Ad, respectively, were assayed with flow cytometry. We constructed vectors expressing the green fluorescent protein gene with Ad5 or modified Ad5 bearing the type 35 fiber region (AdF35), and examined the infectivity to MSCs with flow cytometry. We investigated anti-tumor effects of MSCs transduced with interleukin (IL)-28A gene on human lung carcinoma cells with a colorimetric assay. Expression of IL-28A receptors was tested with the polymerase chain reaction. A promoter activity of transcriptional regulatory regions in MSCs was determined with a luciferase assay and a tumor growth-promoting ability of MSCs was tested with co-injection of human tumor cells in nude mice. RESULTS: MSCs expressed CD46 but scarcely CAR molecules, and subsequently were transduced with AdF35 but not with Ad5. Growth of MSCs transduced with the IL-28A gene remained the same as that of untransduced cells since MSCs were negative for the IL-28A receptors. The IL-28A-transduced MSCs however suppressed growth of lung carcinoma cells co-cultured, whereas MSCs transduced with AdF35 expressing the ß-galactosidase gene did not. A regulatory region of the cyclooygenase-2 gene possessed transcriptional activities greater than other tumor promoters but less than the cytomegalovirus promoter, and MSCs themselves did not support tumor growth in vivo. CONCLUSIONS: AdF35 is a suitable vector to transduce MSCs that are resistant to Ad5-mediated gene transfer. MSCs infected with AdF35 that activate an exogenous gene by the cytomegalovirus promoter can be a vehicle to deliver the gene product to targeted cells.


Asunto(s)
Adenovirus Humanos/genética , Citotoxicidad Inmunológica , Interleucinas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Transducción Genética , Animales , Línea Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Ratones , Regiones Promotoras Genéticas , Transcripción Genética
8.
Am J Clin Pathol ; 140(4): 544-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24045552

RESUMEN

OBJECTIVES: To pathologically distinguish mesothelioma from lung carcinoma, particularly adenocarcinoma. METHODS: We conducted immunohistochemical analyses on clinical specimens, including 26 cases of mesothelioma, 28 cases of lung adenocarcinoma, and 33 cases of lung squamous cell carcinoma. RESULTS: We found that CD90 expression was useful in making a differential diagnosis between epithelioid mesothelioma and lung adenocarcinoma, whereas sarcomatoid mesothelioma and lung carcinoma specimens, irrespective of the histologic types, were negative in general. The sensitivity and specificity of CD90 expression in epithelioid mesothelioma and lung adenocarcinoma were comparable to those of well-established markers used for the differential diagnosis. CONCLUSIONS: These data collectively indicate that CD90 is a novel diagnostic marker that contributes to a diagnosis of epithelioid mesothelioma.


Asunto(s)
Adenocarcinoma/diagnóstico , Carcinoma de Células Escamosas/diagnóstico , Neoplasias Pulmonares/diagnóstico , Mesotelioma/diagnóstico , Neoplasias Pleurales/diagnóstico , Adenocarcinoma/metabolismo , Adenocarcinoma/cirugía , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/cirugía , Línea Celular Tumoral , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/patología , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/cirugía , Mesotelioma/metabolismo , Mesotelioma/cirugía , Neoplasias Pleurales/metabolismo , Neoplasias Pleurales/cirugía , Sensibilidad y Especificidad , Antígenos Thy-1/metabolismo
9.
PLoS One ; 8(8): e72709, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977343

RESUMEN

Interferons (IFNs) have been tested for the therapeutic effects in various types of malignancy, but mechanisms of the anti-tumors effects and the differential biological activities among IFN members are dependent on respective cell types. In this study, we examined growth inhibitory activities of type I and III IFNs on 5 kinds of human mesothelioma cells bearing wild-type p53 gene, and showed that type I IFNs but not type III IFNs decreased the cell viabilities. Moreover, growth inhibitory activities and up-regulated expression levels of the major histocompatibility complexes class I antigens were greater with IFN-ß than with IFN-α treatments. Cell cycle analyses demonstrated that type I IFNs increased S- and G2/M-phase populations, and subsequently sub-G1-phase fractions. The cell cycle changes were also greater with IFN-ß than IFN-α treatments, and these data collectively showed that IFN-ß had stronger biological activities than IFN-α in mesothelioma. Type I IFNs-treated cells increased p53 expression and the phosphorylation levels, and activated apoptotic pathways. A combinatory use of IFN-ß and cisplatin or pemetrexed, both of which are the current first-line chemotherapeutic agents for mesothelioma, produced synergistic anti-tumor effects, which were also evidenced by increased sub-G1-phase fractions. These data demonstrated firstly to our knowledge that IFN-ß produced synergistic anti-tumor effects with cisplatin or pemetrexed on mesothelioma through up-regulated p53 expression.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cisplatino/uso terapéutico , Glutamatos/uso terapéutico , Guanina/análogos & derivados , Interferón beta/uso terapéutico , Mesotelioma/tratamiento farmacológico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Sinergismo Farmacológico , Glutamatos/farmacología , Guanina/farmacología , Guanina/uso terapéutico , Humanos , Interferón beta/farmacología , Mesotelioma/patología , Pemetrexed , Receptores de Interferón/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
10.
Front Biosci (Landmark Ed) ; 18(3): 909-18, 2013 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-23747856

RESUMEN

Type III interferons (IFNs), a new type IFN family consisting of 3 IFN-lambdas, have been identified through a homology search. They include IFN-lambda1, IFN-lambda2 and IFN-lambda3, which are also named as interleukin (IL)-29, IL-28A and IL-28B, respectively. The receptor complex of IFN-lambdas is composed of the IL-10 receptor beta (IL-10Rbeta) and a novel IL-28 receptor alpha (IL-28Ralpha). The signal transductions of type III IFNs seem to be similar to those of type I IFNs. Both type I and III IFNs activate Janus activated kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway and transcribe a number of IFN-associated genes. Various types of viruses induce expressions of type III IFNs as well as type I IFNs; however, the biological functions of type III IFNs could be distinct from those of type I IFNs partly because of the tissue-restricted expression of the type III receptor complexes. In this review, we encapsulate recent understandings about type III IFNs in particular the anti-tumor effects, and discuss possible mechanisms and a potential use for cancer therapy.


Asunto(s)
Interferones/fisiología , Neoplasias/patología , Receptores de Interferón/metabolismo , Animales , Apoptosis , Ciclo Celular , Humanos , Interferones/metabolismo , Neoplasias/metabolismo , Neoplasias/fisiopatología , Transducción de Señal
11.
PLoS One ; 8(3): e60297, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555949

RESUMEN

We examined anti-tumor effects of zoledronic acid (ZOL), one of the bisphosphonates agents clinically used for preventing loss of bone mass, on human mesothelioma cells bearing the wild-type p53 gene. ZOL-treated cells showed activation of caspase-3/7, -8 and -9, and increased sub-G1 phase fractions. A combinatory use of ZOL and cisplatin (CDDP), one of the first-line anti-cancer agents for mesothelioma, synergistically or additively produced the cytotoxicity on mesothelioma cells. Moreover, the combination achieved greater anti-tumor effects on mesothelioma developed in the pleural cavity than administration of either ZOL or CDDP alone. ZOL-treated cells as well as CDDP-treated cells induced p53 phosphorylation at Ser 15, a marker of p53 activation, and up-regulated p53 protein expression levels. Down-regulation of p53 levels with siRNA however did not influence the ZOL-mediated cytotoxicity but negated the combinatory effects by ZOL and CDDP. In addition, ZOL treatments augmented cytotoxicity of adenoviruses expressing the p53 gene on mesothelioma. These data demonstrated that ZOL-mediated augmentation of p53, which was not linked with ZOL-induced cytotoxicity, played a role in the combinatory effects with a p53 up-regulating agent, and suggests a possible clinical use of ZOL to mesothelioma with anti-cancer agents.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Cisplatino/farmacología , Cisplatino/uso terapéutico , Difosfonatos/farmacología , Difosfonatos/uso terapéutico , Imidazoles/farmacología , Imidazoles/uso terapéutico , Proteína p53 Supresora de Tumor/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Western Blotting , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ácido Zoledrónico
12.
J Thorac Oncol ; 7(12): 1850-1857, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23154556

RESUMEN

INTRODUCTION: Genetic characterization of malignant mesothelioma shows a homozygous deletion of the INK4A/ARF locus, which results in inactivation of the p53 pathways. METHODS: We examined possible antitumor effects of adenoviruses with a deletion of the E1B-55kD gene (Ad-delE1B55) on mesothelioma and investigated combinatory actions with the first-line chemotherapeutic agents. RESULTS: Ad-delE1B55 produced cytotoxicity on mesothelioma cells, which was associated with p53 phosphorylation, pRb dephosphorylation, and cleavage of caspases. Ad-delE1B55-infected cells displayed hyperploidy at the cell-cycle analysis and showed enlarged nuclear configurations. Combination of Ad-delE1B55 plus cisplatin or pemetrexed produced antitumor effects in vitro. Furthermore, Ad-delE1B55 and cisplatin showed combinatory effects in an orthotopic animal model. CONCLUSIONS: Cell death caused by Ad-delE1B55 is attributable to cell-cycle arrest at M-phase checkpoint followed by activated apoptotic pathways, and combination of the first-line chemotherapeutic agents and the oncolytic adenovirus is a potential therapeutic for mesothelioma.


Asunto(s)
Adenoviridae/fisiología , Proteínas E1B de Adenovirus/deficiencia , Apoptosis/efectos de los fármacos , Mesotelioma/patología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Citometría de Flujo , Humanos , Mesotelioma/tratamiento farmacológico , Mesotelioma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ploidias , Células Tumorales Cultivadas
13.
J Thorac Oncol ; 7(5): 873-82, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22481236

RESUMEN

INTRODUCTION: We examined whether zoledronic acid (ZOL), the third generation of bisphosphonates, produced cytotoxic effects on human mesothelioma cells in vitro and in vivo, and investigated a possible involvement of p53, Ras, and extracellular signal-regulated kinase1/2 (ERK1/2) pathways. METHODS: Cytotoxicity and cell cycles were assessed with a colorimetric assay and flow cytometry, respectively. Expression levels of apoptosis-linked proteins and prenylation of small guanine-nucleotide-binding regulatory proteins were tested with p53-small interfering RNA, an ERK kinase1/2-inhibitor, and prenyl alcohols. The antitumor activity was examined in an orthotopic animal model. RESULTS: ZOL treatments suppressed growth of mesothelioma cells bearing the wild-type p53 gene through apoptosis induction accompanied by activation of caspases, or S-phase arrest by up-regulated cyclin A and B1. ZOL induced p53 phosphorylation and subsequent activation of the downstream pathways. Down-regulated p53 expression with the small interfering RNA, however, showed that both apoptosis and S-phase arrest were irrelevant to the p53 activation. Geranylgeranyl but not farnesyl pyrophosphate inhibited ZOL-induced apoptosis and S-phase arrest, and the geranylgeraniol supplement decreased ZOL-mediated Rap1A but not Ras unprenylation. Inhibition of ERK1/2 pathways suppressed ZOL-induced apoptosis but not S-phase arrest. We further demonstrated that ZOL, administrated intrapleurally, inhibited the tumor growth in the pleural cavity. CONCLUSIONS: These data indicate that ZOL induces apoptosis or S-phase arrest, both of which are independent of p53 activation and Ras unprenylation, and suggest that ZOL is a possible therapeutic agent to mesothelioma partly through non-Ras- and ERK1/2-mediated pathways.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Difosfonatos/farmacología , Imidazoles/farmacología , Mesotelioma/tratamiento farmacológico , Mesotelioma/metabolismo , Prenilación/efectos de los fármacos , Fase S/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Proteínas ras/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Citometría de Flujo , Humanos , Mesotelioma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ácido Zoledrónico
14.
Cell Immunol ; 275(1-2): 90-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22475191

RESUMEN

Expression of human interleukin (IL)-24 in tumors achieved anti-tumor effects through apoptosis. IL-24 also induced secretion of proinflammatory cytokines, suggesting the role in immunity. We showed that murine IL-24 transcripts started from the second initiation codon and that expressed mIL-24 in tumors failed to induce apoptosis. Proliferation of murine cells expressing mIL-24 was the same as that of the parent cells and inoculation of the mIL-24-expressing tumors into syngeneic mice did not produce anti-tumor effects. Secretory mIL-24 did not induce the expression of the IL-6, TNF-α or IFN-γ gene in spleen cells. Expression of mIL-24 receptor subunits, IL-22R and IL-20R1, was undetectable in spleen cells even though they were stimulated by anti-CD3, anti-CD40 antibody or concanavalin A. Transduction of murine tumors with adenoviruses expressing the human IL-24 gene however suppressed the viability and decreased the tumor growth. These data suggest that mIL-24 is functionally irrelevant to the human counterpart.


Asunto(s)
Apoptosis , Citocinas/inmunología , Regulación de la Expresión Génica , Interleucinas/inmunología , Neoplasias/inmunología , Secuencia de Aminoácidos , Animales , Línea Celular , Citocinas/química , Citocinas/genética , Femenino , Humanos , Interleucinas/genética , Ratones , Datos de Secuencia Molecular , Neoplasias/genética , Neoplasias/patología , Alineación de Secuencia
15.
Clin Dev Immunol ; 2011: 479013, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22013482

RESUMEN

Recently identified interleukin-28 and -29 belong to a novel type III interferon (IFN) family, which could have distinct biological properties from type I and II IFNs. Type I IFNs, IFN-α/ß, have been clinically applied for treating a certain kind of malignancies for over 30 years, but a wide range of the adverse effects hampered the further clinical applications. Type III IFNs, IFN-λs, have similar signaling pathways as IFN-α/ß and inhibits proliferation of tumor cells through cell cycle arrest or apoptosis. Restricted patterns of type III IFN receptor expression in contrast to ubiquitously expressed IFN-α/ß receptors suggest that type III IFNs have limited cytotoxicity to normal cells and can be a possible anticancer agent. In this paper, we summarize the current knowledge on the IFN-λs-mediated tumor cell death and discuss the functional difference between type I and III IFNs.


Asunto(s)
Citocinas/inmunología , Inmunoterapia Activa , Neoplasias/inmunología , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/uso terapéutico , Citotoxicidad Inmunológica/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Inmunoterapia Activa/tendencias , Interferón Tipo I/inmunología , Interferón Tipo I/uso terapéutico , Interferón gamma/inmunología , Interferón gamma/uso terapéutico , Ratones , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Receptores de Interferón/inmunología , Transducción de Señal/inmunología
16.
Eur J Cancer ; 46(1): 180-90, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19879751

RESUMEN

Signal pathways of novel type III interferons (IFN-lambdas) are similar to those of type I IFNs (IFN-alpha/beta) but their distinct functions have not been well characterised. We examined the growth suppressive activity of IFN-lambda1 with nine human oesophageal carcinoma cell lines expressing the IFN-lambda receptor complexes. Among them, three lines but not others showed IFN-lambda1-mediated growth suppression by inducing G1 phase arrest or apoptosis. The G1 phase arrest was accompanied by the up-regulation of p21 and dephosphorylation of retinoblastoma (Rb), and the apoptosis was evidenced by cleavage of caspase-3 and poly (ADP-ribose) polymerase (PARP). Similar but not identical susceptibility was found in IFN-alpha-treated oesophageal carcinoma cells. Despite the differential suppressive responses among the cells, all the cells increased the expression of the myxovirus resistance A (MxA) and 2',5'-oligoadenylate synthetase (2',5'-OAS) genes and class I antigens of the major histocompatibility complexes (MHC) with IFN-lambda1 treatment. Fibroblasts and mesenchymal stem cells, positive for IFN-alpha receptor (IFNAR), lacked one of the IFN-lambda receptor complexes and Het-1A, immortalised oesophageal epithelium cells, were insensible to the IFN-lambda1-induced growth suppression. IFN-lambda1 produced combinatory anti-tumour effects with chemotherapeutic agents, cisplatin (CDDP) and 5-fluorouracil (5-FU), in IFN-lambda1-sensitive oesophageal carcinoma cells but not in normal or Het-1A cells, while IFN-alpha achieved the combinatory suppressive effects to normal cells. These data collectively show that IFN-lambda1 responsiveness is tissue-specific due to the restricted receptors expression and is diversified even among cells of the same lineage, and suggest that IFN-lambda1 is a potential therapeutic agent for oesophageal carcinoma without damaging surrounding tissues.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Esofágicas/patología , Fase G1/efectos de los fármacos , Interleucinas/farmacología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Neoplasias Esofágicas/metabolismo , Humanos , Interferones , Subunidad beta del Receptor de Interleucina-10/metabolismo , Receptores de Citocinas/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Células Tumorales Cultivadas
17.
Immunology ; 128(1 Suppl): e816-25, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19740343

RESUMEN

Interleukin-27 (IL-27) is a new IL-12-related heterodimeric cytokine comprising a novel p28 molecule and the Epstein-Barr-virus-induced gene 3 (EBI3) molecules. It augments initiation of T helper type 1-mediated immunity by enhancing the proliferation and cytokine production of T cells. In this study, we examined whether a secreted form of IL-27 subunits would inhibit IL-27-mediated immunological responses. COS-7 cells transduced with the mouse (m) p28 gene secreted a monomeric mp28 protein; however, those transduced with the mEBI3 gene did not detect a mEBI3 protein in the culture supernatants. The secreted mp28 prevented the IL-27-mediated signal transduction and activator of transcription 1 phosphorylation and subsequently inhibited the IL-27-mediated intercellular adhesion molecule-1 induction and interferon-gamma production in CD4(+) T cells. We generated mp28-expressing murine carcinoma Colon 26 cells and inoculated a mixture of the mp28- and mIL-27-expressing Colon 26 cells into syngeneic BALB/c mice. Simultaneous production of mp28 and mIL-27 from Colon 26 cells suppressed IL-27-mediated anti-tumour effects in the mice. We examined the p28-mediated immune suppression by inoculating mp28-expressing myoblasts into allogeneic mice. Forced production of mp28 suppressed the allogeneic cytotoxic T-lymphocyte induction and subsequently retarded the graft rejection. Furthermore, production of both mp28 and mp40, which inhibits the functions of IL-12 and IL-23, prolonged the graft survival longer than the grafts expressing either mp28 or mp40. We propose that p28 can be a regulatory subunit for IL-27-mediated cellular immune responses and a possible therapeutic agent to suppress unfavourable immune responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Supervivencia de Injerto/inmunología , Interleucina-17/inmunología , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Células COS , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Interferón gamma/biosíntesis , Interferón gamma/efectos de los fármacos , Interferón gamma/inmunología , Interleucina-17/genética , Interleucina-17/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Mioblastos/inmunología , Mioblastos/metabolismo , Fosforilación , Subunidades de Proteína/genética , Subunidades de Proteína/inmunología , Subunidades de Proteína/farmacología , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Receptores de Interleucina-12/agonistas , Receptores de Interleucina-12/inmunología , Receptores de Interleucina-12/metabolismo , Factor de Transcripción STAT1/agonistas , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Linfocitos T Citotóxicos/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Transducción Genética
18.
Immunity ; 29(1): 33-43, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18619870

RESUMEN

Modulation of surface T cell antigen receptor (TCR) expression is an important mechanism for the regulation of immune responses and the prevention of T cell hyperactivation and autoimmunity. The TCR is rapidly internalized after antigen stimulation and then degraded in lysosomes. However, few of the molecules involved in this process have been identified. We demonstrate that the lysosomal protein LAPTM5 negatively regulated surface TCR expression by specifically interacting with the invariant signal-transducing CD3zeta chain and promoting its degradation without affecting other CD3 proteins, CD3epsilon, CD3delta, or CD3gamma. TCR downmodulation required the polyproline-tyrosine motifs and the ubiquitin-interacting motif of LAPTM5. LAPTM5 deficiency resulted in elevated TCR expression on both CD4(+)CD8(+) thymocytes and spleen T cells after CD3 stimulation, as well as enhanced T cell responses in vitro and in vivo. These results identify a lysosomal protein important for CD3zeta degradation and illustrate a unique mechanism for the control of surface TCR expression and T cell activation.


Asunto(s)
Complejo CD3/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Activación de Linfocitos/inmunología , Proteínas de la Membrana/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Animales , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas Inmediatas-Precoces/inmunología , Immunoblotting , Inmunoprecipitación , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Proteínas/inmunología , Proteínas/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología
19.
DNA Repair (Amst) ; 7(8): 1392-8, 2008 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-18562254

RESUMEN

The immunoglobulin genes undergo a high frequency of point mutations at both C:G and A:T pairs in the germinal center (GC) B cells. This hypermutation process is initiated by the activation-induced cytidine deaminase (AID), which converts cytosine to uracil and generates a U:G lesion. Replication of this lesion, or its repair intermediate the abasic site, could introduce C:G mutations but the mechanisms leading to mutations at non-damaged A:T pairs remain elusive. Using a lacZ-transgenic system in which endogenous genome mutations can be detected with high sensitivity, we found that GC B cells exhibited a much higher ratio of A:T mutations as compared to naïve B, non-GC B, and cells of other tissues. This property does not require AID or active transcription of the target gene, and is dependent on DNA polymerase eta. These in vivo results demonstrate that GC B cells are unique in having an intrinsic propensity to generate A:T mutations during repair of endogenous DNA damage. These findings have important implications in understanding how AID, which can only target C:G base pairs, is able to induce the entire spectrum of mutations observed in immunoglobulin variable region genes in GC B cells.


Asunto(s)
Adenina/química , Linfocitos B/ultraestructura , Mutación Puntual , Timina/química , Animales , Secuencia de Bases , Cartilla de ADN , Ratones , Transcripción Genética
20.
Front Biosci ; 13: 2578-87, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17981735

RESUMEN

Direct destruction of targeted tumors and subsequent induction of systemic immunity is not pertinent to gene therapy but gene therapy is probably the most suitable therapeutic modality to achieve the local and systemic anti-tumor effects. Current strategies for cancer gene therapy in fact consist of direct inhibition of tumor growth and activation of systemic host defense mechanisms. We have been working on development of oncolytic adenoviruses and cytokine-mediated activation of host immune systems to produce better therapeutic effects. The adenoviruses in which the E1A expression is controlled by an exogenous regulatory region are preferentially cytotoxic to target tumor cells depending on the specificity of the regulatory region and cytokines that differentiate naive T cells into T helper type 1 cells can amplify immune responses generated. Combination of the two strategies has an advantage. Tumor destruction by oncolytic viruses does not impair immune systems in contract to chemotherapy and radiotherapy but enable to produce anti-tumor responses against putative tumor antigens that are subsequently released from the destroyed tumor. In this process, dendritic cells play a pivotal role since they act as professional antigen presenting cells and are involved in an initial phase of immune responses, either activation of immunity or induction of immune tolerance. Antigen loading with subsequent appropriate activation of dendritic cells is thereby crucial for activated anti-tumor responses, which possibly eliminate even distant metastatic foci. Combinatory gene therapy with oncolytic viruses and activation of host immune system thereby can evoke immune responses against all the tumor antigens expressed by the process of "antigen-spreading" mechanisms.


Asunto(s)
Adenoviridae/genética , Citocinas/metabolismo , Células Dendríticas/citología , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Neoplasias/metabolismo , Proteínas E1A de Adenovirus/metabolismo , Animales , Presentación de Antígeno , Antineoplásicos/química , Células Dendríticas/metabolismo , Humanos , Sistema Inmunológico , Interleucina-12/biosíntesis , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...