Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Cell Biol ; 3(10): 867-73, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11584267

RESUMEN

Stretching of cardiac muscle modulates contraction through the enhancement of the Ca2+ transient, but how this occurs is still not known. We found that stretching of myocytes modulates the elementary Ca2+ release process from ryanodine-receptor Ca2+-release channels (RyRCs), Ca2+ sparks and the electrically stimulated Ca2+ transient. Stretching induces PtdIns-3-OH kinase (PI(3)K)-dependent phosphorylation of both Akt and the endothelial isoform of nitric oxide synthase (NOS), nitric oxide (NO) production, and a proportionate increase in Ca2+-spark frequency that is abolished by inhibiting NOS and PI(3)K. Exogenously generated NO reversibly increases Ca2+-spark frequency without cell stretching. We propose that myocyte NO produced by activation of the PI(3)K-Akt-endothelial NOS axis acts as a second messenger of stretch by enhancing RyRC activity, contributing to myocardial contractile activation.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Óxido Nítrico/metabolismo , Proteínas Serina-Treonina Quinasas , Animales , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes/metabolismo , Masculino , Contracción Miocárdica/efectos de los fármacos , Miocardio/citología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Sprague-Dawley , S-Nitroso-N-Acetilpenicilamina/farmacología , Espectrometría de Fluorescencia
2.
Ann Oncol ; 20(5): 874-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19179552

RESUMEN

BACKGROUND: Although most BRCA sequence variants are clearly deleterious and unequivocally pathogenetic, several are still classified as variants of unknown significance. PATIENTS AND METHODS: We followed families undergoing oncogenetic counseling from risk identification to risk definition by genetic testing and risk management. RESULTS: We identified two germline mutations in the BRCA2 gene in a woman with breast and ovarian cancer. One sequence alteration was 859/G>A in exon 7 (V211I). The other second sequence alteration (IVS13-2A>T) affected the splicing site in intron 13. The latter alteration is not yet listed in the Breast Cancer Information Core database. RT-PCR resulted in transcription of a sequence lacking exon 7 and a subsequent anomalous stop codon in exon 9 thereby confirming altered messenger RNA (mRNA) maturation. Amplification of the mutation in intron 13 resulted in transcription of a sequence lacking exon 14 and an anomalous stop codon in exon 15 thereby confirming altered mRNA maturation. Both mutations led to a truncated BRCA2 protein in its carboxy-terminal region. CONCLUSION: The two BRCA2 mutations identified affect mRNA splicing fidelity and play a pathogenetic role in breast and ovarian cancer.


Asunto(s)
Proteína BRCA2/genética , Neoplasias de la Mama/genética , Exones , Asesoramiento Genético , Pruebas Genéticas , Mutación de Línea Germinal , Neoplasias Ováricas/genética , Sitios de Empalme de ARN , Proteínas Reguladoras de la Apoptosis , Proteína BRCA1/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Análisis Mutacional de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Linaje , Fenotipo , Pronóstico , Medición de Riesgo
3.
Ann Oncol ; 20(5): 906-12, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19155242

RESUMEN

BACKGROUND: Preoperative chemoradiation is now standard treatment for stages II-III rectal cancer. Capecitabine (CAP) and oxaliplatin (OX) are synergistic with radiotherapy (RT) and active in colorectal neoplasms. PATIENTS AND METHODS: Two cycles of CAP 825 mg/m(2) b.i.d. (days 1-14) and OX 50 mg/m(2) (days 1 and 8) every 3 weeks were given concomitantly with pelvic conformal RT (45 Gy). Patients with a > or =T3 and/or node-positive rectal tumour were eligible. The pathologic tumour response was defined according to the tumour regression grade (TRG) scale. RESULTS: Forty-six patients were enrolled. Gastrointestinal adverse events were mostly G1-G2; only two patients experienced G3 vomiting and diarrhoea and six patients had G1 peripheral neuropathy. Haematological toxicity was rare. G2 proctitis and anal pain occurred in two patients. Pathological complete response (TRG1) was observed in nine patients (20.9%; 95% CI 8.7%-33.1%); TRG2 in 19 patients (44.2%); TRG3 in 12 patients (27.9%); and TRG4 in three patients (7%). Overall, nine patients recurred: five with distant metastases, one with local recurrence, and three with both local recurrence and distant metastases. CONCLUSIONS: CAP-OX-RT as preoperative treatment for rectal cancer induces a remarkable rate of complete or near-complete pathologically documented response and is well tolerated.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Procedimientos Quirúrgicos del Sistema Digestivo , Radioterapia Conformacional , Neoplasias del Recto/terapia , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Capecitabina , Quimioterapia Adyuvante , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Supervivencia sin Enfermedad , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/análogos & derivados , Humanos , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Radioterapia Conformacional/efectos adversos , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/mortalidad , Neoplasias del Recto/patología , Neoplasias del Recto/radioterapia , Neoplasias del Recto/cirugía , Factores de Tiempo , Resultado del Tratamiento
4.
J Mol Cell Cardiol ; 44(2): 270-80, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18191142

RESUMEN

Impaired glucose uptake is associated with both cardiac hypertrophy and contractile dysfunction, but whether there are common underlying mechanisms linking these conditions is yet to be determined. Using a 'gene dose' Cre-Lox GLUT4-deficient murine model, we examined the effect of suppressed glucose availability on global myocardial gene expression and glycolysis substrate bypass on the function of isolated perfused hearts. Performance of hearts from 22- to 60-week-old male GLUT4 knockout (KO, >95% reduction in GLUT4), GLUT4 knockdown (KD, 85% reduction in cardiac GLUT4) and C57Bl/6 wild-type (WT) controls was measured ex vivo in Langendorff mode perfusion. DNA microarray was used to profile mRNA expression differences between GLUT4-KO and GLUT4-KD hearts. At 22 weeks, GLUT4-KO hearts exhibited cardiac hypertrophy and impaired contractile function ex vivo, characterized by a 40% decrease in developed pressure. At 60 weeks, dysfunction was accentuated in GLUT4-KO hearts and evident in GLUT4-KD hearts. Exogenous pyruvate (5 mM) restored systolic pressure to a level equivalent to WT (GLUT4-KO, 176.8+/-13.2 mmHg vs. WT, 146.4+/-9.56 mmHg) in 22-week-old GLUT4-KO hearts but not in 60-week-old GLUT4-KO hearts. In GLUT4-KO, DNA microarray analysis detected downregulation of a number of genes centrally involved in mitochondrial oxidation and upregulation of other genes indicative of a shift to cytosolic beta-oxidation of long chain fatty acids. A direct link between cardiomyocyte GLUT4 deficiency, hypertrophy and contractile dysfunction is demonstrated. These data provide mechanistic insight into the myocardial metabolic adaptations associated with short and long-term insulin resistance and indicate a window of opportunity for substrate intervention and functional 'rescue'.


Asunto(s)
Transportador de Glucosa de Tipo 4/deficiencia , Glucosa/metabolismo , Miocardio/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Frecuencia Cardíaca/efectos de los fármacos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica/efectos de los fármacos , Miocardio/patología , Tamaño de los Órganos/efectos de los fármacos , Ácido Pirúvico/farmacología , Especificidad por Sustrato/efectos de los fármacos , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
5.
Br J Cancer ; 99(3): 473-80, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18665191

RESUMEN

We investigated the antitumour effect and ability to overcome the resistance to anti-EGFR drugs of enzastaurin, an inhibitor of VEGFR-dependent PKCbeta signalling. Enzastaurin was evaluated alone and in combination with the EGFR inhibitor gefitinib, on growth and signalling protein expression in human cancer cells sensitive and resistant to anti-EGFR drugs, both in vitro and in nude mice. We demonstrated the marked inhibitory activity of enzastaurin against GEO colon and PC3 prostate cancer cells and their gefitinib-resistant counterparts GEO-GR and PC3-GR, accompanied by inhibition of pAkt and its effector pp70S6K, pGSK3beta and VEGF expression and secretion. Moreover, enzastaurin showed a cooperative effect with gefitinib in parental and in gefitinib-resistant cells. Remarkably, these results were confirmed in vivo, where enzastaurin showed antitumour activity and cooperativity with gefitinib in mice grafted with GEO and GEO-GR tumours, incrementing their median survival and inhibiting the aforesaid protein expression and secretion in tumour specimens. In conclusion, enzastaurin by interfering with signalling proteins implicated in EGFR drug resistance markedly cooperates with gefitinib in sensitive and gefitinib-resistant tumours, thus overcoming and reverting such resistance and providing a rational basis for its development in patients resistant to anti-EGFR drugs.


Asunto(s)
Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Indoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Medios de Cultivo Condicionados , Ensayos de Selección de Medicamentos Antitumorales , Ensayo de Inmunoadsorción Enzimática , Gefitinib , Humanos , Ratones , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/metabolismo , Quinazolinas/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Hum Gene Ther ; 18(9): 821-36, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17845130

RESUMEN

Successful gene therapy approaches for metachromatic leukodystrophy (MLD), based either on hematopoietic stem/progenitor cells (HSPCs) or direct central nervous system (CNS) gene transfer, highlighted a requirement for high levels of arylsulfatase A (ARSA) expression to achieve correction of disease manifestations in the mouse model. Full assessment of the safety of ARSA expression above physiological levels thus represents a prerequisite for clinical translation of these approaches. Here, using lentiviral vectors (LVs), we generated two relevant models for the stringent evaluation of the consequences of ARSA overexpression in transduced cells. We first demonstrated that ARSA overexpression in human HSPCs does not affect their clonogenic and multilineage differentiation capacities in clonogenic assays and in a neonatal hematochimeric mouse model. Further, we studied ARSA overexpression in all body tissues by generating transgenic mice overexpressing the ARSA enzyme by LV up to 15-fold above the normal range and carrying multiple copies of LV in their genome. Characterization of these mice demonstrated the safety of ARSA overexpression in two main gene therapy targets, HSPCs and neurons, with maintenance of the complex functions of the hematopoietic and nervous system in the presence of supraphysiological enzyme levels. The activity of other sulfatases dependent on the same common activator, sulfatase-modifying factor-1 (SUMF1), was tested in ARSA-overexpressing HSPCs and in transgenic mice, excluding the occurrence of saturation phenomena. Overall, these data indicate that from the perspective of clinical translation, therapeutic levels of ARSA overexpression can be safely achieved. Further, they demonstrate an experimental platform for the preclinical assessment of the safety of new gene therapy approaches.


Asunto(s)
Cerebrósido Sulfatasa/metabolismo , Terapia Genética , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/terapia , Animales , Animales Recién Nacidos , Antígenos CD34/inmunología , Antígenos CD34/metabolismo , Southern Blotting , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Cerebrósido Sulfatasa/efectos adversos , Cerebrósido Sulfatasa/análisis , Ensayo de Unidades Formadoras de Colonias , Estudios de Factibilidad , Vectores Genéticos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Lentivirus/genética , Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/patología , Ratones , Ratones Transgénicos , Modelos Animales , Neuronas/citología , Neuronas/metabolismo , Reacción en Cadena de la Polimerasa , Bazo/citología , Transducción Genética
7.
J Natl Cancer Inst ; 90(14): 1087-94, 1998 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-9672257

RESUMEN

BACKGROUND: The expression of epidermal growth factor receptor (EGFR) and type I cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKAI) is associated with neoplastic transformation. By use of human renal cancer cell lines (i.e., 769-P, ACHN, A498, and SW839), we investigated the antiproliferative activity and the antitumor activity of an anti-EGFR humanized chimeric mouse monoclonal antibody, MAb C225, and a novel mixed backbone 18-mer antisense oligonucleotide, HYB 190, that targets expression of the RIalpha regulatory subunit of PKAI. METHODS: The antiproliferative activity of MAb C225 and oligonucleotide HYB 190, alone or in combination, on different renal cancer cell lines was determined by monitoring cell growth in soft agar. In addition, the induction of apoptosis by treatment with the anti-EGFR antibody and/or antisense PKAI oligonucleotides was evaluated by flow cytometric analysis of fragmented DNA. The antitumor activity of MAb C225 and oligonucleotide HYB 190 was determined in athymic mice bearing established ACHN tumor xenografts. Cell proliferation and tumor growth data were evaluated for statistical significance using Student's t test; reported P values are two-sided. RESULTS: MAb C225 and oligonucleotide HYB 190 inhibited colony formation in soft agar in a dose-dependent manner for all renal cancer cell lines tested. We observed a potentiation of growth inhibition and induction of apoptosis when 769-P cells and ACHN cells were treated with both agents. Combination treatment with MAb C225 and oligonucleotide HYB 190 caused regression of ACHN tumor xenografts, whereas single-agent treatment only delayed tumor growth. CONCLUSION: The combination of anti-EGFR MAb C225 and ited cooperative antiproliferative effects and cooperative antitumor effects on EGFR and PKAI-expressing human renal cancer cell lines.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Receptores ErbB/antagonistas & inhibidores , Terapia Genética/métodos , Inmunoterapia/métodos , Neoplasias Renales/terapia , Oligonucleótidos Antisentido/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados , Apoptosis , Cetuximab , Terapia Combinada , Citometría de Flujo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/genética , Neoplasias Renales/inmunología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Recombinantes de Fusión/uso terapéutico , Células Tumorales Cultivadas
8.
Cancer Res ; 51(23 Pt 1): 6263-7, 1991 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-1657383

RESUMEN

Site-selective cyclic AMP (cAMP) analogues have been shown to inhibit growth and induce differentiation in several human leukemia cell lines. However, detailed studies of the effects exerted by cAMP analogues on cell cycle kinetics have been lacking. We have examined the effects of 8-Cl-cAMP and N6-benzyl-cAMP on the cell cycle kinetics of the HL-60 human promyelocytic leukemia cell line. A cell cycle study was performed by univariate DNA analysis after 24-72 h of treatment with noncytotoxic concentrations of 8-Cl-cAMP and N6-benzyl-cAMP capable of inducing 50-60% growth inhibition in these cells. HL-60 cells treated with 5 microM 8-Cl-cAMP showed no significant change in the cell distribution in the cycle as compared to the untreated control cells, whereas the treatment with 10 microM N6-benzyl-cAMP transiently increased the percentage of cells in the G0/G1 phase after 48 h, followed by a partial recovery at 72 h. Combined treatment with low doses of 8-Cl-cAMP and N6-benzyl-cAMP, each of which alone produced 20% growth inhibition, exerted a growth inhibitory effect of 65% and delayed increase of the G0/G1 phase by 72 h. To better understand the cell cycle effects induced by 8-Cl-cAMP, flow cytometric analysis of bromodeoxyuridine incorporation was also performed. 8-Cl-cAMP treatment exhibited a slowing down of the cell cycle; thus, the delayed appearance of the G0/G1 cell accumulation after combined treatment could be due to this effect of 8-Cl-cAMP on the HL-60 cell cycle. At a toxic dose, 8-Cl-cAMP brought about a G2M block, whereas N6-benzyl-cAMP brought about an increase of the G0/G1 compartment. G2M block produced by toxic doses of 8-Cl-cAMP was not related to its adenosine metabolite since 8-Cl-adenosine did not produce any specific block in the cell cycle. Our results show, for the first time, that these site-selective cAMP analogues could affect cell cycle kinetics at different points. These data may provide the basis for combination treatments involving cAMP analogues and other agents in the treatment of human leukemia.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Antineoplásicos/farmacología , AMP Cíclico/análogos & derivados , Leucemia Promielocítica Aguda/patología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Ciclo Celular/efectos de los fármacos , AMP Cíclico/farmacología , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Factores de Tiempo , Células Tumorales Cultivadas/patología
9.
Cancer Res ; 53(22): 5548-53, 1993 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-8106149

RESUMEN

Amplification of erbB-2 gene and overexpression of gp185erbB-2 gene product is found in approximately one-third of primary human breast and ovarian cancer. Overexpression of gp185erbB-2 was recently found in human papillary thyroid carcinomas, but not in thyroid follicular carcinomas or adenomas. The erbB-2 gene encodes a cell surface growth factor receptor with intrinsic tyrosine kinase activity. Wild type human erbB-2 has been shown to act as a potent oncogene when overexpressed in mouse fibroblasts. To test whether overexpression of normal human erbB-2 gene can transform epithelial differentiated rat thyroid cells, these cells were infected with a recombinant retroviral expression vector containing the erbB-2 protooncogene. Rat thyroid cells expressing high levels of gp185erbB-2 do not display a fully transformed and tumorigenic phenotype. However, the isolated cell clones that overexpress gp185erbB-2, show changes in their growth properties if compared to normal thyroid cells, since they can grow in absence of thyrotropin, the main growth factor controlling thyroid cell proliferation in vitro, and do not respond to the growth inhibitory effect of transforming growth factor beta.


Asunto(s)
Transformación Celular Neoplásica/genética , Cinamatos , Proteínas Oncogénicas Virales/análisis , Proto-Oncogenes , Glándula Tiroides/patología , Animales , Western Blotting , Diferenciación Celular , División Celular/efectos de los fármacos , Resistencia a Medicamentos , Higromicina B/análogos & derivados , Higromicina B/farmacología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Oncogénicas Virales/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Ratas , Ratas Endogámicas F344 , Receptor ErbB-2 , Receptores de Tirotropina/análisis , Glándula Tiroides/química , Factor de Crecimiento Transformador beta/farmacología
10.
Cancer Res ; 52(9): 2504-8, 1992 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-1314695

RESUMEN

A set of adenosine 3':5'-monophosphate (cAMP) analogues that combine exocyclic sulfur substitutions in the equatorial (Rp) or the axial (Sp) position of the cyclophosphate ring with modifications in the adenine base of cAMP were tested for their effect on the growth of HL-60 human promyelocytic leukemia cells and LS-174T human colon carcinoma cells. Both diasteromeres of the phosphorothioate derivatives were growth inhibitory, exhibiting a concentration inhibiting 50% of cell proliferation of 3-100 microM. Among the analogues tested, Rp-8-Cl-cAMPS and Sp-8-Br-cAMPS were the two most potent. Rp-8-Cl-cAMPS was 5- to 10-fold less potent than 8-Cl-cAMP while Sp-8-Br-cAMPS was approximately 6-fold more potent than 8-Br-cAMP. The growth inhibition was not due to a block in a specific phase of the cell cycle or due to cytotoxicity. Rp-8-Cl-cAMPS enhanced its growth-inhibitory effect when added together with 8-Cl-cAMP and increased differentiation in combination with N6-benzyl-cAMP. The binding kinetics data showed that these Sp and Rp modifications brought about a greater decrease in affinity for Site B than for Site A of RI (the regulatory subunit of type I cAMP-dependent protein kinase) and a substantial decrease of affinity for Site A of RII (the regulatory subunit of type II protein kinase) but only a small decrease in affinity for Site B of RII, indicating the importance of the Site B binding of RII in the growth-inhibitory effect. These results show that the phosphorothioate analogues of cAMP are useful tools to investigate the mechanism of action of cAMP in growth control and differentiation and may have practical implication in the suppression of malignancy.


Asunto(s)
AMP Cíclico/análogos & derivados , Receptores de AMP Cíclico/metabolismo , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Neoplasias del Colon/patología , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Isomerismo , Leucemia Promielocítica Aguda/inmunología , Leucemia Promielocítica Aguda/patología , Proteínas Quinasas/biosíntesis , Células Tumorales Cultivadas
11.
Cancer Res ; 53(12): 2846-51, 1993 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8389245

RESUMEN

Alterations of p53 are one of the most common molecular changes found in all types of lung tumors, suggesting a crucial role for p53 in bronchial carcinogenesis. However, the prognostic significance of p53 abnormalities in lung cancer patients is still unclear. By using genetic and immunohistochemical methods we have found p53 alterations in 40 of 53 (75%) primary, resected non-small cell lung cancer. A strong association (P = 0.0015) was found between deletions on chromosome region 17p13.3 and p53 mutations suggesting that loss of the wild-type p53 allele might be necessary for tumorigenesis. Correlations to clinicopathological parameters showed that p53 alterations (structural aberration of the gene and/or nuclear accumulation of the protein) are significantly linked with metastatic involvement of hilar and mediastinal lymph nodes (P < 0.01). Since the latter are well established prognostic factors for non-small cell lung cancer, p53 aberrations may also be a predictor of tumor aggressiveness.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Exones/genética , Genes p53/genética , Neoplasias Pulmonares/genética , Mutación Puntual/genética , Anciano , Alelos , Secuencia de Bases , Carcinoma de Pulmón de Células no Pequeñas/patología , Deleción Cromosómica , Cromosomas Humanos Par 17 , Humanos , Neoplasias Pulmonares/patología , Metástasis Linfática , Persona de Mediana Edad , Datos de Secuencia Molecular , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa/métodos , Pronóstico
12.
Cancer Res ; 51(18): 4898-902, 1991 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-1893380

RESUMEN

Reversal of the drug resistance phenotype by the use of agents which induce cell differentiation offers an experimental approach to the study of chemoresistance. In numerous in vitro models, alpha-interferon (alpha-IFN) has been shown to induce phenotypical changes and to modulate the growth of cancer cells. The aim of the present study was to define the effect of alpha-IFN on the Adriamycin sensitivity of the human colon adenocarcinoma cell line, LoVo, and its Adriamycin-resistant variant, LoVo/DX. Pretreatment of LoVo/DX cells with 500 units/ml of alpha-IFN increased sensitivity to low doses of Adriamycin. Similar treatment conditions did not change the sensitivity of the parental cell line. Following treatment of the LoVo/DX cells with alpha-IFN plus 100 ng/ml Adriamycin for 1 h, 30% of the cells survived compared to 100% of untreated cells. This effect was not related to changes in cell cycle kinetics induced by alpha-IFN treatment and did not result from variations in the expression of P-glycoprotein at the cell surface, as assessed by flow cytometric analysis using monoclonal antibody MRK16. Adriamycin accumulation was increased by alpha-IFN as assessed by spectrofluorometric analysis. Thus, the data suggest that in LoVo/DX cells, alpha-IFN increased Adriamycin cytotoxicity through modulation of the multidrug resistance phenotype.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Doxorrubicina/farmacología , Interferón Tipo I/farmacología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Doxorrubicina/farmacocinética , Resistencia a Medicamentos/genética , Ensayos de Selección de Medicamentos Antitumorales , Expresión Génica , Humanos , Glicoproteínas de Membrana/genética , Fosfoproteínas/genética , Proteínas Recombinantes , Células Tumorales Cultivadas
13.
Cancer Res ; 54(15): 4123-8, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-7518350

RESUMEN

We have shown that a mutant derivative of Chinese hamster ovary CHO-K1 cells, ADR-5, which shows hypersensitivity to topoisomerase II (topo II)-inhibitory drugs, is cross-sensitive to the site-selective cyclic AMP analogue 8-chloro-cyclic AMP. We tested the hypothesis that overexpression of the type I alpha regulatory subunit of protein kinase A may represent a common element conferring hypersensitivity to both topo II inhibitors and 8-chloro-cyclic AMP in ADR-5 cells. We have demonstrated that ADR-5 cells overexpress RI alpha protein, compared to parental CHO-K1 cells. Moreover, retroviral vector-mediated transfer of the RI alpha gene into CHO-K1 cells was able to confer a drug-hypersensitive phenotype similar to that exhibited by ADR-5 cells. Analysis of topo II protein levels and activity revealed no differences between parental and infected cells, suggesting that protein kinase A may be involved in the downstream processing of topo II-mediated events.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Antineoplásicos/farmacología , Bleomicina/farmacología , Células CHO/efectos de los fármacos , Células CHO/enzimología , Cisplatino/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Etopósido/farmacología , Tenipósido/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Células CHO/patología , División Celular/efectos de los fármacos , Cricetinae , Ensayos de Selección de Medicamentos Antitumorales
14.
Cancer Res ; 57(22): 5107-11, 1997 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-9371510

RESUMEN

8-Chloro-cAMP (8-Cl-cAMP) is a novel agent that is able to inhibit the growth of a wide variety of cancer cell types in vitro and in vivo and, at doses devoid of toxicity, to achieve plasma concentrations in cancer patients in a range effective for cancer cell growth inhibition. In this study, we have demonstrated that 8-Cl-cAMP, at a dose causing mild or no growth inhibition, synergistically increased the growth-inhibitory effect of paclitaxel or cisplatin in a wide series of cell lines including human breast, lung, ovary, colon, and head carcinomas and melanoma. A similar effect was also observed with another taxane, docetaxel, and with the platinum-derivative carboplatin. 8-Cl-cAMP also markedly enhanced apoptotic cell death induced by each cytotoxic drug. A cooperative antitumor effect was also observed in vivo, because treatment with paclitaxel followed by 8-Cl-cAMP markedly inhibited the growth of GEO human colon cancer xenografts as compared to paclitaxel alone without signs of toxicity. These data demonstrate that 8-Cl-cAMP synergistically increases the antiproliferative activity of taxanes and platinum-derived compounds and provide a rationale to use 8-Cl-cAMP in combination with taxanes and platinum-derived compounds.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Paclitaxel/farmacología , Ensayo de Tumor de Célula Madre/métodos , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Apoptosis/genética , División Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Fase G2 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitosis , Trasplante de Neoplasias , Trasplante Heterólogo , Células Tumorales Cultivadas/efectos de los fármacos
15.
Oncogene ; 9(11): 3233-40, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7936647

RESUMEN

MCF-10A, a nontransformed mammary epithelial cell line, requires a mixture of hormones and growth factors for optimal cell proliferation. In this report we show that when MCF-10A cells are cultured in serum free medium they become quiescent and accumulate in G0/G1 phases of the cell cycle. Following addition of complete medium to quiescent cells, MCF-10A cells enter into the S phase within 15-18 h and resume the cell cycle distribution of proliferating cells within 24 h. Measurement of RI alpha subunit of the cAMP-dependent protein kinase (PKA) shows a 10- to 15-fold increase in protein levels at 6 h following complete medium addition, thus preceding cell entry into the S phase. Retroviral vector-mediated overexpression of RI alpha, but not of RII beta or C alpha subunits of PKA, enables MCF-10A cells to grow in serum-free medium. In addition, RI alpha downregulation by specific antisense oligodeoxynucleotide treatment or following infection with a retroviral vector containing the RI alpha cDNA in antisense orientation determines growth arrest of proliferating MCF-10A cells and is able to partially block S phase entry of quiescent MCF-10A cells following complete medium addition. These results suggest that RI alpha/PKAI is involved in the control of cell cycle progression of mammary epithelial cells at a G1 to S transition border, and that its overexpression is able to overcome serum and growth factors requirement for cell proliferation.


Asunto(s)
Ciclo Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glándulas Mamarias Animales/citología , Animales , Sangre , Línea Celular , Clonación Molecular , Medio de Cultivo Libre de Suero , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/genética , ADN Complementario , Epitelio , Vectores Genéticos , Humanos , Retroviridae/genética
16.
Oncogene ; 15(8): 927-36, 1997 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-9285688

RESUMEN

The teratocarcinoma-derived growth factor-1 (TDGF-1) gene codes for a 188-aminoacid glycoprotein that shares structural homology with the epidermal growth factor (EGF) family of growth factors. TDGF-1 is highly expressed in the undifferentiated embryonal carcinoma stem cell line NTERA2 clone D1 (NT2/D1) and its expression is downregulated in response to differentiating agents such as retinoic acid (RA) and hexamethylen-bisacetamide (HMBA). To assess the role of TDGF-1 in the onset and/or progression of human germ cell tumors, we analysed TDGF-1 expression by Northern blot and immunostaining in a panel of 59 human germ cell tumors of different histological origins. We show that TDGF-1 expression is markedly elevated in a subset of human testicular germ cell tumors as compared to normal testes. TDGF-1 overexpression occurs in about 100% of tumors with non-seminomatous phenotype, such as embryonal carcinomas and malignant undifferentiated teratocarcinomas. To address the questions of how TDGF-1 (previously called CRIPTO) may affect the growth and/or the differentiation of embryonal carcinoma cells, we have characterized the effects of exogenous recombinant TDGF-1 protein on the proliferation rate and differentiation 'potential of NT2/D1. Exogenous TDGF-1 protein stimulated DNA synthesis and cell proliferation in both undifferentiated and differentiated NT2/D1 cells. However, TDGF-1 protein treatment was unable to block differentiation induced by both RA and HMBA. These results suggest that TDGF-1 growth factor may represent an autocrine growth factor that may be involved in the process of development of testicular neoplasms.


Asunto(s)
Factor de Crecimiento Epidérmico , Germinoma/metabolismo , Sustancias de Crecimiento/biosíntesis , Glicoproteínas de Membrana , Proteínas de Neoplasias/biosíntesis , Neoplasias Testiculares/metabolismo , Secuencia de Aminoácidos , Biomarcadores de Tumor/biosíntesis , Northern Blotting , Carcinoma Embrionario/metabolismo , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Proteínas Ligadas a GPI , Expresión Génica/efectos de los fármacos , Sustancias de Crecimiento/genética , Sustancias de Crecimiento/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/farmacología , Proteínas Recombinantes/farmacología , Teratocarcinoma/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
17.
Oncogene ; 18(46): 6241-51, 1999 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-10597222

RESUMEN

Hexamethylen-bisacetamide (HMBA) represents the prototype of a group of hybrid polar compounds, which induce differentiation in a variety of transformed cells including human embryonal carcinoma cells. Therefore, HMBA has been used in the differentiation therapy of cancer for patients with both hematological and solid malignancies. Upon HMBA treatment, the embryonal carcinoma cell line NTERA-2 clone D1 (NT2/D1) accumulates in G1 and undergoes terminal differentiation. Here we demonstrate that growth arrest and differentiation of NT2/D1 cells induced by HMBA involve increased expression of the cyclin-dependent kinase inhibitor p27, enhanced association of p27 with cyclin E/CDK2 complexes and suppression of kinase activity associated to cyclin E/CDK2 (but not to cyclin D3/CDK4). When HMBA differentiation was induced in the presence of p27 antisense oligonucleotides, NT2/D1 cells failed to arrest growth properly and, in parallel with the reduction of the anti-apoptotic Bcl-2 gene expression, cells underwent massive programmed cell death. Conversely, constitutive expression of p27 into NT2/D1 cells induced a marked reduction in the growth potential of these cells and partially reproduced HMBA-induced modification of surface antigen expression (down-regulation of SSEA-3 expression and up-regulation of VINIS-53 expression). Expression of p21 induced growth arrest but not differentiation. Likewise, inhibition of CDK2 by transfection of a dominant negative CDK2 in NT2/D1 cells or treatment with the kinase inhibitor olomucine induced growth arrest but not differentiation. Therefore, we propose that p27 represents a crucial molecule in HMBA signaling that cannot be replaced by p21. Furthermore, the results obtained with CDK2 inhibitors demonstrate that the block of CDK2 activity is sufficient for growth arrest but not for cell differentiation and suggest that, at least in these cells, growth arrest and differentiation are regulated by two overlapping but different pathways.


Asunto(s)
Apoptosis/fisiología , Quinasas CDC2-CDC28 , Carcinoma Embrionario/patología , Proteínas de Ciclo Celular , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Supresoras de Tumor , Acetamidas/farmacología , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Antígenos de Superficie/biosíntesis , Antígenos de Superficie/genética , Antígenos de Carbohidratos Asociados a Tumores , Apoptosis/efectos de los fármacos , Carcinoma Embrionario/metabolismo , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Ciclinas/fisiología , Regulación Neoplásica de la Expresión Génica , Glicoesfingolípidos/biosíntesis , Glicoesfingolípidos/genética , Humanos , Cinetina , Sustancias Macromoleculares , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Purinas/farmacología , Proteína de Retinoblastoma/metabolismo , Roscovitina , Antígenos Embrionarios Específico de Estadio , Células Tumorales Cultivadas
18.
Circulation ; 102(19 Suppl 3): III319-25, 2000 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-11082408

RESUMEN

BACKGROUND: Ca(2+) overload plays an important role in the pathogenesis of cardioplegic ischemia-reperfusion injury. The standard technique to control Ca(2+) overload has been to reduce Ca(2+) in the cardioplegic solution (CP). Recent reports suggest that Na(+)/H(+) exchange inhibitors can also prevent Ca(2+) overload. We compared 4 crystalloid CPs that might minimize Ca(2+) overload in comparison with standard Mg(2+)-containing CP: (1) low Ca(2+) CP (0.25 mmol/L), (2) citrate CP/normal Mg(2+) (1 mmol/L Mg(2+)), (3) citrate CP/high Mg(2+) (9 mmol/L Mg(2+)), and (4) the addition of the Na(+)/H(+) exchange inhibitor HOE-642 (Cariporide). We also tested the effect of citrate titration in vitro on the level of free Ca(2+) and Mg(2+) in CPs. METHODS AND RESULTS: Isolated working rat heart preparations were perfused with oxygenated Krebs-Henseleit buffer and subjected to 60 minutes of 37 degrees C arrest and reperfusion with CPs with different Ca(2+) concentrations. Cardiac performance, including aortic flow (AF), was measured before and after ischemia. Myocardial high-energy phosphates were measured after reperfusion. The in vitro addition of citrate to CP (2%, 21 mmol/L) produced parallel reductions in Mg(2+) and Ca(2+). Because only Ca(2+) was required to be low, the further addition of Mg(2+) increased free Mg(2+), but the highest level achieved was 9 mmol/L. Citrate CP significantly impaired postischemic function (AF 58.3+/-2. 5% without citrate versus 41.6+/-3% for citrate with normal Mg(2+), P:<0.05, versus 22.4+/-6.2% for citrate with high Mg(2+), P:<0.05). Low-Ca(2+) CP (0.25 mmol/L Ca(2+)) significantly improved the recovery of postischemic function in comparison with standard CP (1.0 mmol/L Ca(2+)) (AF 47.6+/-1.7% versus 58.3+/-2.5%, P:<0.05). The addition of HOE-642 (1 micromol/L) to CP significantly improved postischemia function (47.6+/-1.7% without HOE-642 versus 62.4+/-1. 7% with HOE-642, P:<0.05). Postischemia cardiac high-energy phosphate levels were unaffected by Ca(2+) manipulation. CONCLUSIONS: (1) A lowered Ca(2+) concentration in CP is beneficial in Mg(2+)-containing cardioplegia. (2) The use of citrate to chelate Ca(2+) is detrimental in the crystalloid-perfused isolated working rat heart, especially with high Mg(2+). (3) The mechanism of citrate action is complex, and its use limits precise simultaneous control of Ca(2+) and Mg(2+). (4) HOE-642 in CP is as efficacious in preservation of the ischemic myocardium as is the direct reduction in Ca(2+).


Asunto(s)
Calcio/metabolismo , Soluciones Cardiopléjicas/metabolismo , Ácido Cítrico/metabolismo , Magnesio/metabolismo , Daño por Reperfusión/prevención & control , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Soluciones Cardiopléjicas/química , Ácido Cítrico/farmacología , Guanidinas/farmacología , Corazón/efectos de los fármacos , Pruebas de Función Cardíaca/efectos de los fármacos , Técnicas In Vitro , Ácido Láctico/metabolismo , Magnesio/farmacología , Masculino , Miocardio/metabolismo , Fosfocreatina/metabolismo , Ratas , Ratas Sprague-Dawley , Sulfonas/farmacología , Volumetría
19.
Cell Death Differ ; 10(2): 218-29, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12700650

RESUMEN

The mechanisms of tumor cell resistance to interferon-alpha (IFNalpha) are at present mostly unsolved. We have previously demonstrated that IFNalpha induces apoptosis on epidermoid cancer cells and EGF antagonizes this effect. We have also found that IFNalpha-induced apoptosis depends upon activation of the NH(2)-terminal Jun kinase-1 (Jnk-1) and p(38) mitogen-activated protein kinase, and that these effects are also antagonized by EGF. At the same time, IFNalpha increases the expression and function of the epidermal growth factor receptor (EGF-R). Here we report that the apoptosis induced by IFNalpha occurs together with activation of caspases 3, 6 and 8 and that EGF also antagonizes this effect. On the basis of these results, we have hypothesized that the increased EGF-R expression and function could represent an inducible survival response that might protect tumor cells from apoptosis caused by IFNalpha via extracellular signal regulated kinase 1 and 2 (Erk-1/2) cascades. We have found an increased activity of Ras and Raf-1 in IFNalpha-treated cells. Moreover, IFNalpha induces a 50% increase of the phosphorylated isoforms and enzymatic activity of Erk-1/2. We have also demonstrated that the inhibition of Ras activity induced by the transfection of the dominant negative Ras plasmid RASN17 and the inhibition of Mek-1 with PD098059 strongly potentiates the apoptosis induced by IFNalpha. Moreover, the selective inhibition of this pathway abrogates the counteracting effect of EGF on the IFNalpha-induced apoptosis. All these findings suggest that epidermoid tumor cells counteract the IFNalpha-induced apoptosis through a survival pathway that involves the hyperactivation of the EGF-dependent Ras->Erk signalling. The selective targeting of this pathway appears to be a promising approach in order to enhance the antitumor activity of IFNalpha.


Asunto(s)
Apoptosis/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Interferón-alfa/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas ras/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Caspasas/metabolismo , Supervivencia Celular , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/efectos de los fármacos , Receptores ErbB/metabolismo , Flavonoides/farmacología , Humanos , Interferón-alfa/metabolismo , Células KB , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/análisis , Proteínas Proto-Oncogénicas c-raf/efectos de los fármacos
20.
Cell Death Differ ; 6(8): 773-80, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10467351

RESUMEN

We have demonstrated that interferon-alpha2-recombinant (IFNalpha) at growth inhibitory concentrations enhances the expression and signalling activity of the epidermal growth factor receptor (EGF-R) in human epidermoid carcinoma KB cells. Here we report that KB cells exposed to IFNalpha underwent apoptotic cell death and this effect was antagonized by EGF. We have also found that IFNalpha enhanced the expression of heat shock proteins (HSP) HSP-70, HSP-90 and HSP-27 and activated the NH2-terminal Jun kinase-1 (JNK-1) and p38 mitogen activated protein kinase, the target enzymes of a stress-dependent intracellular transduction pathway. Moreover, the overexpression of the wild-type JNK-1, obtained through plasmid transfection of KB cells, induced apoptosis which was potentiated by the exposure of wild-type JNK-1 (JNK-1wt)-transfected cells to IFNalpha. All these effects were neutralized by the addition of EGF to parental and JNK-1wt-transfected KB cells exposed to IFNalpha. In conclusion, EGF has a protective effect on KB cells from apoptosis while antagonizing a stress response elicited by IFNalpha and targeted on the stress pathway terminal kinases.


Asunto(s)
Apoptosis , Factor de Crecimiento Epidérmico/metabolismo , Proteínas de Choque Térmico , Interferón-alfa/farmacología , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Proteínas de Choque Térmico HSP27 , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas HSP90 de Choque Térmico/biosíntesis , Humanos , Interferón-alfa/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Chaperonas Moleculares , Proteínas de Neoplasias/biosíntesis , Transfección , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA