Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Oncol ; 45(3): 919-28, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24969394

RESUMEN

Drug development for castration resistant prostate cancer (CRPC) is challenging, since this cancer is still associated with high mortality and limited therapeutic options. In 2004, docetaxel became the first-line chemotherapy for CRPC improving survival by a few months and remains the standard of care in CRPC patients. However, existing or developing resistance to docetaxel in patients is the main limitation of its efficacy. The present review presents the molecular mechanisms involved in docetaxel toxicity and in docetaxel resistance in prostate cancer cells. We outlined the endogenous mechanisms of resistance and the role of tumor microenvironment in the resistance of CRPC to docetaxel. This has led us to focus on molecules associated with resistance, such as the molecular chaperones heat shock proteins (HSPs) and clusterin (CLU), and the cytokines interleukin-6 (IL-6) and the divergent member of the tumor growth factor family MIC-1 (macrophage inhibitory cytokine-1 also named GDF-15). We discuss their interest as blood-based markers to monitor docetaxel resistance. Finally, new therapies intended to overcome docetaxel resistance of CRPC targeted on these molecular resistance pathways are present.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Terapia Molecular Dirigida , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/sangre , Línea Celular Tumoral , Clusterina/antagonistas & inhibidores , Clusterina/metabolismo , Docetaxel , Factor 15 de Diferenciación de Crecimiento/antagonistas & inhibidores , Factor 15 de Diferenciación de Crecimiento/metabolismo , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/metabolismo , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Taxoides/uso terapéutico , Microambiente Tumoral/efectos de los fármacos
2.
Oncogene ; 25(58): 7635-45, 2006 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-16767158

RESUMEN

Anticancer drugs can induce tumor cell death by caspase-dependent apoptosis. The observation that procaspase-10 expression decreased in leukemic cells from acute myeloblastic leukemia patients at first relapse led us to explore the role of caspase-10 in cytotoxic drug-induced apoptosis. We show that caspase-10 is activated in etoposide-treated cells in a dose- and time-dependent manner. A caspase-10 peptide inhibitor, a caspase-10 dominant-negative mutant or a small interfering RNA (siRNA)-mediated downregulation of the enzyme negatively interfere with drug-induced cell death and caspase-2, -3, -8 and -9 activation. The extrinsic pathway to apoptosis is not involved in drug-induced caspase-10 activation that occurs downstream of Bax redistribution to mitochondria and cytochrome c release from this organelle. siRNA-mediated downregulation of Apaf-1 prevents etoposide-mediated activation of caspase-10. In a cell-free assay, cytochrome c and dATP treatment of cell extracts after immunodepletion of either caspase-3 or caspase-9 indicates that caspase-10 is activated downstream of caspase-9. Then, caspase-10 is involved in a feedback amplification loop that amplifies caspase-9 and -3 activities. Altogether, these data indicate an active role for caspase-10 in cytotoxic drug-induced tumor cell death, downstream of the mitochondria.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis , Caspasa 10/fisiología , Etopósido/farmacología , Transducción de Señal , Apoptosomas/efectos de los fármacos , Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Activación Enzimática , Humanos , Leucemia Mieloide Aguda/diagnóstico , Mitocondrias/metabolismo , Recurrencia
3.
Br J Cancer ; 94(2): 259-67, 2006 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-16404427

RESUMEN

This study evaluated by immunohistochemistry (IHC) immune cell response during neoadjuvant primary systemic therapy (PST) with trastuzumab in patients with HER2-positive primary breast cancer. In all, 23 patients with IHC 3+ primary breast cancer were treated with trastuzumab plus docetaxel. Pathological complete and partial responses were documented for nine (39%) and 14 (61%) patients, respectively. Case-matched controls comprised patients treated with docetaxel-based PST without trastuzumab (D; n=23) or PST without docetaxel or trastuzumab (non-taxane, non-trastuzumab, NT-NT; n=23). All surgical specimens were blind-analysed by two independent pathologists, with immunohistochemical evaluation of B and T lymphocytes, macrophages, dendritic cells and natural killer (NK) cells. Potential cytolytic cells were stained for Granzyme B and TiA1. HER2 expression was also evaluated in residual tumour cells. Trastuzumab treatment was associated with significantly increased numbers of tumour-associated NK cells and increased lymphocyte expression of Granzyme B and TiA1 compared with controls. This study supports an in vivo role for immune (particularly NK cell) responses in the mechanism of trastuzumab action in breast cancer. These results suggest that trastuzumab plus taxanes lead to enhanced NK cell activity, which may partially account for the synergistic activity of trastuzumab and docetaxel in breast cancer.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Receptor ErbB-2/metabolismo , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Antineoplásicos Fitogénicos/administración & dosificación , Linfocitos B/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Células Dendríticas/efectos de los fármacos , Docetaxel , Femenino , Humanos , Inmunohistoquímica , Células Asesinas Naturales/efectos de los fármacos , Macrófagos/efectos de los fármacos , Persona de Mediana Edad , Terapia Neoadyuvante , Linfocitos T/efectos de los fármacos , Taxoides/administración & dosificación , Trastuzumab
4.
Am J Pathol ; 157(6): 1863-74, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11106559

RESUMEN

Tumor neovascularization is considered to be a critical step in the development of a malignant tumor. Endothelin (ET)-1 is a powerful vasoconstrictor and mitogenic peptide that is produced by many cancer cell lines. The cellular distribution of the ET components was evaluated in human colon tumors and compared to normal colon. There was more of the ET components (preproET-1, endothelin-converting enzyme-1, and ETA and ETB receptors) in adenomas and adenocarcinomas than in the normal colon. There was overproduction of preproET-1 and endothelin-converting enzyme-1 in carcinoma cells and stromal vessels, suggesting that they are a local source of ET-1. ETA receptors were present in stromal myofibroblasts of neoplastic tissue, and there were large amounts of ETB receptors in the endothelium and myofibroblasts. There was also a redistribution of alpha-smooth muscle actin-positive cells in the vascular structures of tumors. An experimental rat model of induced colon cancer treated for 30 days with bosentan, a mixed antagonist of both ET receptors, confirmed the morphological changes observed during the tumor vascularization. Our data suggest that ET-1 and its receptor play a role in colon cancer progression, with ET-1 functioning as a negative modulator of the stromal response.


Asunto(s)
Adenocarcinoma/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Neoplasias del Colon/metabolismo , Endotelinas/metabolismo , Precursores de Proteínas/metabolismo , Receptores de Endotelina/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Ácido Aspártico Endopeptidasas/genética , Bosentán , Colon/metabolismo , Neoplasias del Colon/patología , Antagonistas de los Receptores de Endotelina , Endotelina-1 , Enzimas Convertidoras de Endotelina , Endotelinas/genética , Femenino , Humanos , Masculino , Metaloendopeptidasas , Persona de Mediana Edad , Precursores de Proteínas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas , Receptor de Endotelina A , Receptor de Endotelina B , Receptores de Endotelina/genética , Valores de Referencia , Sulfonamidas/farmacología , Distribución Tisular
5.
FASEB J ; 14(15): 2393-5, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11024011

RESUMEN

There is considerable interest in determining the conditions leading to enhanced inducible nitric oxide synthase (iNOS) gene expression and nitric oxide (NO) biosynthesis. Using in vivo footprinting, we demonstrate that heat shock of murine macrophages concurrent with lipopolysaccharide (LPS) treatment stimulated changes in guanine methylation sensitivity at ?898/9, at a putative partial heat shock element (HSE) and at -893/4, a site bordering an E-box, within the iNOS gene enhancer, suggesting inducible occupation by transcription factors at these regions. LPS treatment accompanied by heat shock provoked increased iNOS gene transcription, increased levels of iNOS protein, and increased production of NO compared with LPS treatment alone. Electrophoretic mobility shift analysis revealed low constitutive levels of specific binding to an E-box and a partial HSE within the iNOS enhancer. Binding to the E-box was increased by LPS treatment or by heat shock, achieving a greater increase by a combination of both treatments. The proteins occupying this site were identified as belonging to the USF family of transcription factors. Heat shock or LPS increased binding to the HSE, and the factor responsible for this interaction was identified as heeat shock factor-1 (HSF-1). Mutations at the HSE revealed the importance of HSF-1 in the induction of iNOS by LPS. Thus, our data reveal two novel regulatory sites in the murine iNOS gene, one of which is implicated in enhancing iNOS expression via LPS stimulation, and provide the first evidence that heat shock enhances transcription of the iNOS gene. These results could have implications in the host response mechanism to fever-associated gram-negative infection.


Asunto(s)
Proteínas de Unión al ADN , Respuesta al Choque Térmico/genética , Óxido Nítrico Sintasa/genética , Animales , Sitios de Unión , Huella de ADN , Lipopolisacáridos/farmacología , Ratones , Modelos Genéticos , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo II , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/metabolismo , Activación Transcripcional , Factores Estimuladores hacia 5'
6.
Int J Cancer ; 86(2): 182-7, 2000 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-10738244

RESUMEN

Imbalanced proliferation and apoptosis is important in tumor progression. Endothelin (ET)-1, a 21-amino-acid peptide with vasoconstricting and mitogenic activities, has been shown to be involved in the regulation of apoptosis. Progressive and regressive rat colon (PROb and REGb cells) carcinoma cell lines express the components of the ET-1 system (preproET-1, ET-converting enzyme and ET-receptors) and secrete ET-1. These cells also express the Fas(APO-1, CD95)/FasL system, but are resistant to FasL-induced apoptosis. We thus addressed the role of ET-1 in FasL-dependent cell death. Bosentan, a mixed ET(A)/ET(B) receptor antagonist, potentiated FasL-induced apoptosis in these cells. At low concentrations (10(-13) to 10(-10) M), ET-1 dose-dependently reversed bosentan-induced apoptosis. Bosentan sensitization to FasL-induced apoptosis was not mediated by increased expression of Fas receptor and was blocked by the caspase inhibitor zVAD-fmk. The specific inhibition of enzymes involved in ceramide production did not restore survival of cells exposed to FasL and bosentan. Our results suggest that ET-1 is a survival factor able to protect in vitro colon carcinoma cells against FasL-induced apoptosis.


Asunto(s)
Apoptosis , Neoplasias del Colon/patología , Antagonistas de los Receptores de Endotelina , Glicoproteínas de Membrana/farmacología , Adenocarcinoma/química , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Ácido Aspártico Endopeptidasas/genética , Bosentán , Inhibidores de Caspasas , Neoplasias del Colon/química , Neoplasias del Colon/metabolismo , Sinergismo Farmacológico , Endotelina-1/metabolismo , Endotelina-1/fisiología , Enzimas Convertidoras de Endotelina , Endotelinas/genética , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Humanos , Glicoproteínas de Membrana/fisiología , Metaloendopeptidasas , Precursores de Proteínas/genética , ARN Mensajero/análisis , Ratas , Receptores de Endotelina/genética , Receptores de Endotelina/fisiología , Sulfonamidas/farmacología , Células Tumorales Cultivadas , Receptor fas/biosíntesis
7.
Clin Exp Metastasis ; 17(4): 299-306, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10545016

RESUMEN

The antitumoral effect of the new lipid A OM 174 was investigated in a model of colon cancer in rats. Peritoneal carcinomatosis were induced in BDIX rats by intraperitoneal injection of syngeneic PROb cancer cells. The treatment started 2 weeks later, when rats had macroscopic peritoneal nodules. An antitumoral effect was first obtained with OM 174 intraperitoneally injected, then an intravenous treatment was developed. When injected 15 times intravenously, at the dose of 1 mg/kg, 2 days apart, OM 174 induced the complete regression of tumors and hemorrhagic ascitis in 90% of the tumor-bearing rats, whereas all the untreated rats died of their tumors. To our knowledge, this treatment is the most effective ever applied to macroscopic tumors. Furthermore, the treatment induced the immunization of rats since the reinjection of PROb tumor cells in OM 174-cured rats did not cause the formation of new tumors while injection of another syngenic colon tumor cells did. Only in treated rats tumors were infiltrated with lymphocytes, macrophages and fibroblasts. The treatment did not increase necrosis but generated apoptotic areas. OM 174 was not directly toxic for tumor cells, and thus the observed effect involved the host-mediated antitumor reaction. Therefore we hypothesize that OM 174 therapy induces tumor cell apoptosis, stimulates the phagocytosis of apoptotic bodies and then activates immune system by antigen presentation.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Vacunas contra el Cáncer/uso terapéutico , Neoplasias del Colon/patología , Lipopolisacáridos/uso terapéutico , Metástasis de la Neoplasia/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Metástasis de la Neoplasia/terapia , Ratas , Reproducibilidad de los Resultados
8.
Lab Invest ; 79(10): 1215-25, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10532585

RESUMEN

Nitric oxide (NO) is generated by a family of isoenzymes named nitric oxide synthases (NOS) which includes a cytokine-inducible form, NOSII. NO is a free radical known to inhibit cell proliferation, to induce apoptosis, and to be a mediator of macrophage cytostatic and cytotoxic effects. We investigated NOS in 40 human breast carcinomas and 8 benign breast lesions. NOSII was localized in tumor cells by immunohistochemistry. NOS activity, measured with the citrulline assay, was detected in 27 of 40 tumors. Neither immunohistologic labeling nor NOS activity was detected in benign samples. NOS labeling and activity were significantly related (p < 0.02). For the first time, a significant negative relationship between NOS activity and tumor cell proliferation (p < 0.002) was found. We also showed that tumors with high NOS activity expressed progesterone receptors (p < 0.04). These results are consistent with the observation of high NOS activity in tumors with low grade (p < 0.05). These in vivo observations were related to in vitro data: cytokines (IL-1beta, IFN-gamma, and TNF-alpha) induced NOSII expression in human MCF-7 breast cancer cells, and NO inhibited their proliferation. Thus, we show herein that tumors with high NOS activity have low proliferation rate and low grade, which correlates with the in vitro observation of the inhibition of proliferation of human breast cancer cells by NO. These results may have future therapeutic implications.


Asunto(s)
Neoplasias de la Mama/enzimología , Óxido Nítrico Sintasa/análisis , Receptores de Progesterona/análisis , Neoplasias de la Mama/patología , Diferenciación Celular/fisiología , División Celular/fisiología , Femenino , Humanos , Inmunohistoquímica , Metástasis Linfática , Invasividad Neoplásica , Óxido Nítrico Sintasa de Tipo II , Pronóstico , Células Tumorales Cultivadas
9.
Am J Pathol ; 154(6): 1867-76, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10362813

RESUMEN

Transforming growth factor-beta1 (TGF-beta1) has been shown to down-regulate NO synthesis in a variety of normal cells. In the present study, we investigated the influence of TGF-beta1 upon NO production in tumor cells and its consequences for tumor development. During the growth of PROb colon carcinoma cells intraperitoneally injected in syngeneic BDIX rats, intratumoral concentration of TGF-beta1 increases while NO concentration stays very low. Tumor regression induced by intraperitoneal injections of a lipid A is associated with a decrease in TGF-beta1 and an increase in NO intratumoral concentration. In these tumors, PROb tumor cells are the NO- and TGF-beta1-secreting cells. Using PROb cells transfected with an expression vector coding for TGF-beta1 antisense mRNA, we demonstrate in vitro that there is an inverse correlation between the amount of TGF-beta1 secreted and the ability of PROb cells to secrete NO. As the same results were obtained in the presence of an anti-TGF-beta type II receptor neutralizing antibody, and as exogenous TGF-beta1 is without any effect on NO secretion by PROb cells, TGF-beta1 apparently down-regulates NO synthesis in PROb cells by an intracellular mechanism. These results suggest that endogenous TGF-beta1 constitutes a potential target in a search for new antitumoral agents.


Asunto(s)
Receptores de Activinas Tipo I , Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Líquido Intracelular/metabolismo , Óxido Nítrico/biosíntesis , Factor de Crecimiento Transformador beta/fisiología , Animales , Carcinoma/tratamiento farmacológico , Carcinoma/enzimología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/enzimología , Regulación hacia Abajo , Femenino , Inmunoterapia , Lípido A/uso terapéutico , Masculino , Trasplante de Neoplasias , Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo II , Proteínas Serina-Treonina Quinasas/biosíntesis , ARN sin Sentido/genética , ARN sin Sentido/farmacología , Ratas , Ratas Endogámicas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Sistemas de Mensajero Secundario/fisiología , Transfección , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Células Tumorales Cultivadas
10.
Int J Cancer ; 81(5): 755-60, 1999 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-10328229

RESUMEN

It is well documented that nitric oxide (NO) is an effector molecule of macrophage-mediated tumor cell toxicity in vitro; however, little is known about the role of NO in the antitumor immune response in vivo. We have developed a treatment protocol using lipid A. We have investigated the effects of lipid A on inducible NO synthase (NOS II) expression and evolution inside tumors during the course of treatment. Lipid A (OM-174) treatment induced tumor regression in rats bearing established colon tumors. Furthermore, NO was synthesized and secreted inside the tumors of lipid A-treated rats, as demonstrated by the increase of NOS II mRNA and NOS II content in the tumors, as well as of NOS II activity and NO production. During treatment, NOS II was localized in tumor cells only. Lipid A had no direct effect on tumor cells in vitro, while the combination of interferon gamma (IFN-gamma) plus interleukin-1 beta (IL-1beta) induced production of NO by tumor cells which was cytostatic. The content of IFN-gamma and IL-1beta in tumors was enhanced during lipid A treatment; this is in agreement with an indirect effect of lipid A in vivo via the IFN-gamma and IL-1beta pathways.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/enzimología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/enzimología , Lípido A/uso terapéutico , Óxido Nítrico Sintasa/biosíntesis , Adenocarcinoma/metabolismo , Animales , División Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Femenino , Interferón gamma/metabolismo , Interferón gamma/farmacología , Interleucina-1/metabolismo , Interleucina-1/farmacología , Masculino , Trasplante de Neoplasias , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas , Células Tumorales Cultivadas
11.
Eur J Immunol ; 28(9): 2960-70, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9754583

RESUMEN

Repeated exposure to bacterial endotoxin causes a diminished response by the host to further exposure. One important feature of this hyporesponsiveness is a reduced macrophage production of nitric oxide (NO) via the inducible nitric oxide synthase (iNOS) pathway. Using a murine macrophage model, we observed that hyporesponsiveness was accompanied by a decrease in the levels of NO release (measured as nitrite), iNOS protein and iNOS gene transcription. The expression of the putative lipopolysaccharide (LPS) receptor, CD14, was not altered. In vivo genomic footprinting showed that the same binding sites are occupied in the iNOS promoter and enhancer of desensitized macrophages and of LPS-responsive macrophages, yet the composition of NF-kappaB in the nuclei of these cells was found to be altered. The transcriptionally inactive homodimer p50-p50 represented the predominant binding activity in nuclei from LPS-pretreated cells before and after stimulation. Nuclei from cells which had not been pretreated but were stimulated contained more of the transcriptionally active p50-p65 heterodimer than their pretreated counterparts. Consistent with this, the cytosolic steady-state level of an inhibitor of NF-kappaB activity, I-kappaBalpha, was decreased in normal cells but not in pretreated cells. We propose that the presence of an overwhelming excess of transcriptionally inactive p50 homodimers on their kappaB sites in the iNOS control region in pretreated cells may block kappaB site binding by p50-p65, thereby reducing the activity of the protein complex governing iNOS transcription.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/inmunología , Lipopolisacáridos/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , FN-kappa B/inmunología , Óxido Nítrico Sintasa/inmunología , Animales , Línea Celular , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/genética , Ratones , FN-kappa B/genética , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II
12.
Cancer Immunol Immunother ; 46(3): 160-6, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9625539

RESUMEN

Nitric oxide (NO) has been shown to inhibit the proliferation of lymphocytes. However, in tumour-bearing rats treated with the immunomodulator OM 163, the regressing nodules were heavily infiltrated by T lymphocytes, although they contained high levels of NO. We show here that NO, while inhibiting the proliferation of lymphocytes, increased their life-span, pointing to the ambivalence of this molecule in the course of tumour growth and regression.


Asunto(s)
Óxido Nítrico/farmacología , Linfocitos T/citología , Linfocitos T/inmunología , Adyuvantes Inmunológicos/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Carcinoma/inmunología , Carcinoma/terapia , Supervivencia Celular/efectos de los fármacos , Escherichia coli , Inmunoterapia , Activación de Linfocitos/efectos de los fármacos , Óxido Nítrico/biosíntesis , Neoplasias Peritoneales/inmunología , Neoplasias Peritoneales/terapia , Ratas
13.
J Pharmacol Exp Ther ; 278(2): 926-33, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8768749

RESUMEN

Although PROb colonic tumor cells are immunosuppressive, the immunomodulator OM 163 induced the disappearance of macroscopic peritoneal nodules in 50% of rats bearing a peritoneal carcinomatosis (p.c.) induced by PROb cells. When the p.c. developed, the number of T lymphocytes was low and the expression of interferon (IFN)-gamma mRNA in tumor nodules decreased very rapidly. The TGF-beta 1 secreted by PROb cells could be responsible for the immunosuppression, because the PROb cell supernatant inhibited IFN-gamma production and T lymphocyte proliferation. When the rats were treated with OM 163, an infiltration of T lymphocytes was observed in tumor nodules, as well as a high expression of the IFN-gamma mRNA. The antitumor efficiency of the immunomodulator OM 163 could be explained in part by a direct effect of OM 163 on T lymphocytes, because in vitro it stimulated the proliferation and the secretion of IFN-gamma of T lymphocytes. OM 163 could also act at the TGF-beta 1 level. Although OM 163 alone or in combination with IFN-gamma did not modify the TGF-beta 1 secretion by PROb cells in vitro, the expression of the TGF-beta 1 mRNA and the TGF-beta 1 protein content was decreased in vivo in treated tumor nodules.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antineoplásicos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Linfocitos/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Animales , Relación Dosis-Respuesta a Droga , Escherichia coli , Expresión Génica/efectos de los fármacos , Terapia de Inmunosupresión , ARN Mensajero/metabolismo , Ratas
14.
J Leukoc Biol ; 59(5): 728-32, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-8656059

RESUMEN

In a model of colon cancer, spleen cells from tumor-bearing rats are neither cytotoxic nor proliferative in vitro in the presence of tumor cells. Interleukin-13 (IL-13) induced an in vitro cytolytic activity of spleen cells from tumor-bearing rats in response to the tumor they bore, but had no effect on spleen cells from normal rats. This cytotoxic response was dependent on both adherent and non- adherent cells, involving both an antigen-presenting activity that was enhanced by IL-13 and a cytolytic activity of lymphocytes. So, IL-13 reversed the tumor-induced immunosuppression.


Asunto(s)
Citotoxicidad Inmunológica/efectos de los fármacos , Interleucina-13/farmacología , Neoplasias Experimentales/inmunología , Bazo/efectos de los fármacos , Animales , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Ratas , Ratas Endogámicas , Bazo/inmunología , Células Tumorales Cultivadas
15.
Nucleic Acids Res ; 24(9): 1682-7, 1996 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-8649986

RESUMEN

A wide variety of cells usefully but sometimes destructively produce nitric oxide via inducible nitric oxide synthase (iNOS). Data obtained by gel shift analysis and reporter assays have linked murine iNOS gene induction by cytokines and bacterial products with the binding of a number of proteins to a proximal promoter, as well as to a distal enhancer of the iNOS gene. Nevertheless, these techniques do not necessarily reflect protein occupation of sites in vivo. To address this, we have used dimethyl sulphate in vivo footprinting to determine binding events in the two murine iNOS transcription control regions, using a classical lipopolysaccharide induction of RAW 264.7 macrophages. Protein-DNA interactions are absent before activation. Exposure to lipopolysaccharide induces protection at a NF-kappaB site and hypersensitivity at a shared gamma-activated site/interferon-stimulated response element within the enhancer. Protections are seen at a NF-IL6, and an Oct site within the promoter. We also observe modulations in guanine methylation at two regions which do not correspond to any known putative binding elements. Furthermore, we confirm the probable involvement of interferon regulatory factor-1 (binding to its -901 to -913 site) and the binding of NF-kappaB to its proximal site. Our data demonstrate an abundance of hitherto-unrecognised protein-DNA binding events upon simple lipopolysaccharide activation of the iNOS gene and suggests a role for protein-protein interactions in its transcriptional induction.


Asunto(s)
Huella de ADN/métodos , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/fisiología , Óxido Nítrico Sintasa/genética , Regiones Promotoras Genéticas/fisiología , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT , Línea Celular , Citocinas/farmacología , ADN/química , ADN/metabolismo , Inducción Enzimática , Guanina/química , Factor 1 Regulador del Interferón , Lipopolisacáridos/farmacología , Macrófagos , Metilación , Ratones , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Óxido Nítrico Sintasa/biosíntesis , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Transcripción Genética
16.
Biochem Biophys Res Commun ; 209(1): 73-9, 1995 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-7537042

RESUMEN

The activity of the inducible nitric oxide synthase enzyme (iNOS) is tightly controlled, partly at the transcriptional level. We find NF-kappa B/Rel activation (p50-p50 and p50-p65) in RAW 264.7 macrophages after lipopolysaccharide treatment and binding to both NF-kappa B sites in the mouse iNOS promoter. To delineate the importance of NF-kappa B/Rel in iNOS gene transcription, we used an unusually direct approach to try to improve on the antioxidant-treatment or reporter techniques, namely the depletion of NF-kappa B/Rel activity through the use of a phosphorothioate-modified oligonucleotide containing three copies of the NF-kappa B consensus sequence. The reduction in NF-kappa B/Rel activity (particularly that binding to the downstream of the two sites) was associated with a 50% reduction in NO output and a reduction in the quantity of the iNOS protein expressed. These results point to the probability that physiologically relevant NF-kappa B/Rel activators or repressors other than lipopolysaccharide might crucially affect the macrophage NO response.


Asunto(s)
Aminoácido Oxidorreductasas/genética , FN-kappa B/metabolismo , Transcripción Genética , Aminoácido Oxidorreductasas/metabolismo , Animales , Secuencia de Bases , Unión Competitiva , Regulación Enzimológica de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Subunidad p50 de NF-kappa B , Óxido Nítrico Sintasa , Oligodesoxirribonucleótidos , Factor de Transcripción ReIA , Factores de Transcripción/metabolismo
17.
Anticancer Res ; 14(3A): 977-9, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-8074501

RESUMEN

In a model of colon cancer in rats (peritoneal carcinomatosis), IL-8 was found to have a highly reproducible antitumoural effect. During IL-8-induced tumour regression the infiltration of nodules by CD4+ T lymphocytes was enhanced. However, splenic lymphocytes did not proliferate in response to tumour cells in vitro. IL-8 antitumour effect was associated with a local but not with a systemic activation of T lymphocytes.


Asunto(s)
Interleucina-8/uso terapéutico , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Neoplasias Experimentales/terapia , Linfocitos T/efectos de los fármacos , Animales , Interleucina-8/farmacología , Neoplasias Experimentales/inmunología , Ratas , Ratas Endogámicas
18.
J Immunol ; 152(10): 5077-83, 1994 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-7513729

RESUMEN

The mechanisms of immunosuppression induced by colon cancer in rats were investigated at the systemic and tumor levels. During tumor growth (after i.p. injection of rat colon adenocarcinoma cells in syngeneic BD IX rats), Con A-induced proliferation of splenic mononuclear cells decreased and nitric oxide (NO) production by splenic macrophages increased concomitantly. Incubating splenic mononuclear cells with an inhibitor of NO synthase, NG-monomethyl-L-arginine, restored lymphocyte proliferation. A low level of inducible NO synthase mRNA was detectable in tumors by Northern blotting, with a weak increase during tumor growth. The NO concentration measured in the tumor nodules increased weakly parallel to the tumor growth. Five and six weeks after tumor cell injection, tumor-infiltrating lymphocytes from disaggregated tumors did not proliferate in the presence of Con A. Addition of NG-monomethyl-L-arginine inhibited the production of NO in tumor dissociations and enhanced tumor-infiltrating lymphocyte proliferation. Glyceryl trinitrate (a NO-releasing compound) totally inhibited the lymphocyte proliferation in vitro while it slightly reduced the tumor cell proliferation. T lymphocytes were therefore more sensitive to NO than were tumor cells. Culture medium from tumor cells induced NO production by splenic macrophages, although the factor involved has not yet been identified. Furthermore, tumor cells could also play a part in NO production by tumors because the tumor cells were induced to produce NO by IFN-gamma plus IL-1. These results strongly suggest the participation of NO in the tumor-induced immunosuppression in rats.


Asunto(s)
Tolerancia Inmunológica , Neoplasias Experimentales/inmunología , Óxido Nítrico/fisiología , Aminoácido Oxidorreductasas/biosíntesis , Animales , Células Cultivadas , Femenino , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Óxido Nítrico Sintasa , Ratas
19.
Immunobiology ; 190(3): 243-54, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8088853

RESUMEN

The correlation between the activation of macrophages by lipopolysaccharides (LPS) from four different bacterial species and their antitumor effect in a rat model of colon cancer was investigated. The efficacy of LPS from Neisseria meningitidis (Nm), Salmonella minnesota (Sm), Escherichia coli (Ec) and Bordetella pertussis (Bp) was evaluated as the smallest concentration inducing rat peritoneal macrophages (pm psi) to produce tumor necrosis factor (TNF), interleukin-1 (IL-1), IL-6 and nitric oxide (NO). The cytokine production was measured in bioassays and NO production quantitatively with Griess reactant. Nm was the most effective LPS with concentrations of 1 ng/10(6) pm psi for the induction of TNF, IL-1 and IL-6 activities and 0.01 ng/10(6) pm psi for the induction of NO production. The range between efficacy of different LPS was broad from 1 to 10(4)-10(5) for TNF activity, 1 to 10(2)-10(3) for NO production and IL-6 activity and 1 to 10-10(2) for IL-1 activity. In vivo antitumor effect was evaluated on the growth of peritoneal carcinomatosis. Complete tumor regressions were observed, the LPS rating with respect to decreasing efficacy was Nm, Sm, Ec then Bp; Nm, Sm and Ec were very closed while Bp was not effective. These results show the correlation between the antitumor effect in vivo of LPS and their capacity to induce in vitro IL-1 activity, but not between their ability to induce NO production, TNF and IL-6 activities.


Asunto(s)
Bacterias/química , Citocinas/biosíntesis , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Neoplasias Experimentales/terapia , Óxido Nítrico/biosíntesis , Animales , Secuencia de Carbohidratos , Células Cultivadas , Femenino , Interleucina-1/biosíntesis , Interleucina-6/biosíntesis , Masculino , Ratones , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Ratas , Ratas Endogámicas , Factor de Necrosis Tumoral alfa/biosíntesis
20.
Cancer Immunol Immunother ; 38(3): 167-70, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8124684

RESUMEN

The antitumor effect of lipopolysaccharides (LPS) has been observed in several experimental models and is likely to be mediated by macrophages. Stimulation of macrophages with LPS results in the release of several cytokines, including tumor necrosis factor, interleukin-1 and neutrophil-activating peptide-1/interleukin-8 (IL-8), which activates polymorphonuclear leukocytes (PMN) in vitro. Since PMN have an antitumor activity, we tested the in vivo effect of IL-8 on the growth of peritoneal carcinomatoses induced by PROb colon cancer cells in syngeneic rats. IL-8 induced a significant regression of tumors measuring 1-5 mm, and a complete regression was observed in 8 out of 40 rats in four independent experiments. IL-8 was not directly cytotoxic in vitro for tumor cells and was effective in vivo in a narrow range of doses. IL-8 had a significant chemotactic effect for peritoneal PMN in both normal and tumor-bearing rats. PMN taken from the peritoneum of tumor-bearing rats during IL-8 treatment had the same cytotoxic activity against PROb tumor cells as PMN from untreated control rats. Microscopic examinations of tumors during the treatment showed poor infiltrating by PMN. We conclude that the antitumor activity of IL-8 in this model is not mediated by PMN cytotoxicity.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Interleucina-8/uso terapéutico , Neutrófilos/inmunología , Animales , Antineoplásicos , Citotoxicidad Inmunológica , Inmunidad Celular , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...