Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Cancer ; 14: 281, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24758355

RESUMEN

BACKGROUND: Zalypsis(®) is a marine compound in phase II clinical trials for multiple myeloma, cervical and endometrial cancer, and Ewing's sarcoma. However, the determinants of the response to Zalypsis are not well known. The identification of biomarkers for Zalypsis activity would also contribute to broaden the spectrum of tumors by selecting those patients more likely to respond to this therapy. METHODS: Using in vitro drug sensitivity data coupled with a set of molecular data from a panel of sarcoma cell lines, we developed molecular signatures that predict sensitivity to Zalypsis. We verified these results in culture and in vivo xenograft studies. RESULTS: Zalypsis resistance was dependent on the expression levels of PDGFRα or constitutive phosphorylation of c-Kit, indicating that the activation of tyrosine kinase receptors (TKRs) may determine resistance to Zalypsis. To validate our observation, we measured the levels of total and active (phosphorylated) forms of the RTKs PDGFRα/ß, c-Kit, and EGFR in a new panel of diverse solid tumor cell lines and found that the IC50 to the drug correlated with RTK activation in this new panel. We further tested our predictions about Zalypsis determinants for response in vivo in xenograft models. All cells lines expressing low levels of RTK signaling were sensitive to Zalypsis in vivo, whereas all cell lines except two with high levels of RTK signaling were resistant to the drug. CONCLUSIONS: RTK activation might provide important signals to overcome the cytotoxicity of Zalypsis and should be taken into consideration in current and future clinical trials.


Asunto(s)
Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Sarcoma/tratamiento farmacológico , Sarcoma/genética , Biomarcadores Farmacológicos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Receptores ErbB/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Proto-Oncogénicas c-kit/biosíntesis , ARN Mensajero/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Sarcoma/patología , Tetrahidroisoquinolinas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
2.
ACS Chem Biol ; 8(9): 2084-94, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-23859655

RESUMEN

We have investigated the target and mechanism of action of a new family of cytotoxic small molecules of marine origin. PM050489 and its dechlorinated analogue PM060184 inhibit the growth of relevant cancer cell lines at subnanomolar concentrations. We found that they are highly potent microtubule inhibitors that impair mitosis with a distinct molecular mechanism. They bind with nanomolar affinity to unassembled αß-tubulin dimers, and PM050489 binding is inhibited by known Vinca domain ligands. NMR TR-NOESY data indicated that a hydroxyl-containing analogue, PM060327, binds in an extended conformation, and STD results define its binding epitopes. Distinctly from vinblastine, these ligands only weakly induce tubulin self-association, in a manner more reminiscent of isohomohalichondrin B than of eribulin. PM050489, possibly acting like a hinge at the association interface between tubulin heterodimers, reshapes Mg(2+)-induced 42 S tubulin double rings into smaller 19 S single rings made of 7 ± 1 αß-tubulin dimers. PM060184-resistant mutants of Aspergillus nidulans map to ß-tubulin Asn100, suggesting a new binding site different from that of vinblastine at the associating ß-tubulin end. Inhibition of assembly dynamics by a few ligand molecules at the microtubule plus end would explain the antitumor activity of these compounds, of which PM060184 is undergoing clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Policétidos/farmacología , Pironas/farmacología , Moduladores de Tubulina/farmacología , Animales , Antineoplásicos/química , Línea Celular Tumoral , Humanos , Mitosis/efectos de los fármacos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Policétidos/química , Poríferos/química , Pironas/química , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química
3.
Int J Clin Exp Pathol ; 6(2): 199-211, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23330005

RESUMEN

XPG (Xeroderma pigmentosum group G complementing factor) is a protein associated with DNA repair and transcription. Point mutations in ERCC5, the gene coding for XPG, cause the cancer-prone disorder xeroderma pigmentosum (XP) while truncation mutations give rise to individuals with the combined clinical features of XP and Cockayne syndrome. Polymorphisms of ERCC5 or alterations in XPG mRNA expression were also associated to an increase risk of different cancers types and to prognosis of cancer patients. However, the expression of XPG protein in different normal or tumor human tissues is not well known. In the present work, we have validated an immunohistochemistry (IHC) assay for detection of expression levels of XPG protein in FFPE human tissue samples. We have also tested this IHC assay in different normal and tumor human tissues. On a microarray containing 28 normal cores, positive staining was observed in 60% of the samples. The highest staining was detected in adrenal gland, breast, colon, heart, kidney, thyroid and tongue. In tumors, positive staining was observed in 9 of 10 breast cancer samples and in all 5 ovarian cancer and 5 sarcomas samples. Subcellular localization was predominantly nuclear. The use of this validated methodology would help to interpret the role of XPG in tumorogenesis and its use as a possible prognostic or predictive factor.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/metabolismo , Glándulas Suprarrenales/fisiología , Mama/fisiología , Neoplasias de la Mama/genética , Síndrome de Cockayne/genética , Colon/fisiología , Femenino , Células HeLa , Corazón/fisiología , Humanos , Inmunohistoquímica/métodos , Inmunohistoquímica/normas , Riñón/fisiología , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/genética , Reproducibilidad de los Resultados , Sarcoma/genética , Glándula Tiroides/fisiología , Lengua/fisiología
4.
PLoS One ; 6(4): e19042, 2011 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-21556352

RESUMEN

Irvalec is a marine-derived antitumor agent currently undergoing phase II clinical trials. In vitro, Irvalec induces a rapid loss of membrane integrity in tumor cells, accompanied of a significant Ca(2+) influx, perturbations of membrane conductivity, severe swelling and the formation of giant membranous vesicles. All these effects are not observed in Irvalec-resistant cells, or are significantly delayed by pretreating the cells with Zn(2+). Using fluorescent derivatives of Irvalec it was demonstrated that the compound rapidly interacts with the plasma membrane of tumor cells promoting lipid bilayer restructuration. Also, FRET experiments demonstrated that Irvalec molecules localize in the cell membrane close enough to each other as to suggest that the compound could self-organize, forming supramolecular structures that likely trigger cell death by necrosis through the disruption of membrane integrity.


Asunto(s)
Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Depsipéptidos/farmacología , Neoplasias/patología , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Humanos
5.
Eur J Med Chem ; 45(1): 134-41, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19853978

RESUMEN

Despite some initial research that reported a lack of activity of trans geometry, complexes with general formula trans-[PtCl2(L)(L')] exhibit an important cytotoxic activity in cisplatin-sensitive and resistant cell lines. Based on the proposed mechanism of action for the trans-platinum compounds, they might form DNA adducts initiating a DNA-damage response and ultimately ending in the activation of the p53 protein. In the present work, we have studied the biochemical properties of the trans-[PtCl2(isopropylamine)(L)] complexes (where L is 3- or 4-(hydroxymethyl)-pyridine) against several cell lines and the relationship between cytotoxicity and the protein p53. Both complexes showed different antitumoral properties depending on the presence or absence of protein p53 in isogenic colon carcinoma HCT116 cell lines. Cell cycle studies with the complexes in these cell lines were performed to investigate their antitumoral activity. Apoptosis was observed to be launched from G1 or G2/M accumulations. Confocal microscopy showed the different behaviour of isogenic tumoral cell lines treated with the trans-platinum complexes. Our data suggest that small differences in the carrier ligands could play an important role in the overall biological effects. The body of the research regarding structure-activity relationships such as the different position of groups in the carrier ligands will provide new rational basis for the design of new platinum antitumor drugs.


Asunto(s)
Alcohol Nicotinílico/química , Compuestos Organoplatinos/química , Compuestos Organoplatinos/farmacología , Propilaminas/química , Piridinas/química , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , ADN/metabolismo , Humanos , Compuestos Organoplatinos/metabolismo , Estereoisomerismo
6.
Mol Carcinog ; 48(11): 1038-47, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19526460

RESUMEN

Mutational activation of RAS proteins occurs in nearly 30% of all human tumors. To date direct pharmacological inhibition of RAS oncoproteins has not been possible. As a consequence, current strategies are focusing on the development of inhibitors that target those kinases acting downstream of RAS proteins, including those of the RAF/MEK/ERK and PI3K/AKT pathways. Most of these inhibitors have undesired off-target effects that mask the potential therapeutic effect of blocking their targeted kinases. To facilitate the screening of selective inhibitors, we have generated lines of mouse embryonic fibroblasts that lack endogenous Ras proteins. These cells proliferate due to ectopic expression of either Ras oncoproteins that selectively activate the Raf/Mek/Erk pathway such as H-Ras(G12V/D38E) or constitutively active kinases such as B-Raf and Mek1. These cell lines were exposed to inhibitors against the RAF, MEK, and AKT kinases as well as inhibitors of other kinases known to crosstalk with RAS signaling such as JNK and p38. Amongst all compounds tested, only the MEK inhibitors U0126 and PD0325901, showed the expected specificity pattern. Yet, PD0325901, but not U0126, was able to inhibit a cell line lacking Ras proteins that owed its proliferative properties to loss of p53. Thus, suggesting unexpected off-target activities for this compound. The use of cell lines whose proliferative properties exclusively depend on selective targets provide a novel strategy to analyze the specificity of selective inhibitors designed against molecular targets implicated in human cancer.


Asunto(s)
Descubrimiento de Drogas , Proteínas ras/genética , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Proliferación Celular , Cartilla de ADN , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Biochem Pharmacol ; 78(2): 162-70, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19427997

RESUMEN

Zalypsis is a new synthetic alkaloid tetrahydroisoquinoline antibiotic that has a reactive carbinolamine group. This functionality can lead to the formation of a covalent bond with the amino group of selected guanines in the DNA double helix, both in the absence and in the presence of methylated cytosines. The resulting complex is additionally stabilized by the establishment of one or more hydrogen bonds with adjacent nucleotides in the opposite strand as well as by van der Waals interactions within the minor groove. Fluorescence-based thermal denaturation experiments demonstrated that the most favorable DNA triplets for covalent adduct formation are AGG, GGC, AGC, CGG and TGG, and these preferences could be rationalized on the basis of molecular modeling results. Zalypsis-DNA adducts eventually give rise to double-strand breaks, triggering S-phase accumulation and apoptotic cell death. The potent cytotoxic activity of Zalypsis was ascertained in a 24 cell line panel. The mean IC(50) value was 7nM and leukemia and stomach tumor cell lines were amongst the most sensitive. Zalypsis administration in four murine xenograft models of human cancer demonstrates significant tumor growth inhibition that is highest in the Hs746t gastric cancer cell line with no weight loss of treated animals. Taken together, these results indicate that the potent antitumor activity of Zalypsis supports its current development in the clinic as an anticancer agent.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Tetrahidroisoquinolinas/farmacología , Animales , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Dioxoles/química , Dioxoles/farmacología , Dioxoles/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Ratones , Tetrahidroisoquinolinas/química , Tetrahidroisoquinolinas/uso terapéutico , Trabectedina , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Chem Biodivers ; 5(10): 2090-2100, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18972499

RESUMEN

trans-Pt(II) Complexes with aliphatic amines and planar amines such as (hydroxymethyl)pyridines, and pyridine-3- and pyridine-4-carboxylic acids were synthesized and screened for their potential cytotoxic activity in different cancer cell lines used at the NCI for in vitro screens, i.e., MCF7, NCIH460, and SF268. The complexes studied were designed to differ in geometrical parameters such as the position of the phenyl-group substituents and the nature of the substituents themselves for gathering information about the structure-activity relationships in the trans-complexes. The variation of the substituents turns to be crucial for their biological activity, as both pyridine-3- and pyridine-4-carboxylic acids in trans-position to both amine and isopropylamine ligands provided complexes which displayed no specificity toward any type of cell tested, while (hydroxymethyl)pyridine in trans-position to isopropylamine ligands led to complexes that were clearly more effective against the cell lines tested.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Carboxílicos/química , Compuestos Organoplatinos/farmacología , Compuestos de Platino/farmacología , Propilaminas/química , Piridinas/química , Aminas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Enlace de Hidrógeno , Ligandos , Estructura Molecular , Compuestos Organoplatinos/síntesis química , Compuestos Organoplatinos/química , Compuestos de Platino/síntesis química , Compuestos de Platino/química , Estereoisomerismo , Relación Estructura-Actividad
9.
Mol Cancer Ther ; 6(4): 1310-6, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17431109

RESUMEN

Aplidin (plitidepsin) is a novel anticancer drug isolated from the marine tunicate Aplidium albicans. Aplidin shows potent antitumor activity in preclinical models against a wide variety of human tumors. Aplidin is currently in phase II clinical trials in a variety of solid tumors and hematologic malignancies. Moreover, clinical studies of Aplidin in combination with other agents are ongoing because it generally lacks cross-resistance with other known cytotoxic drugs. The mode of action of Aplidin in tumor cells is only partially understood. Aplidin induces an early oxidative stress response, which results in a rapid and sustained activation of the epidermal growth factor receptor, the nonreceptor protein tyrosine kinase Src, and the serine threonine kinases c-Jun NH(2)-terminal kinase and p38 mitogen-activated protein kinase. Here, we show that sensitivity to Aplidin correlates inversely with the levels of expression of the cyclin-dependent kinase inhibitor p27(kip1) (p27) in a panel of low passaged human sarcoma cell lines. Aplidin induces p27 through an oxidation-dependent mechanism and the reduction of p27 levels by specific short hairpin RNA increases Aplidin sensitivity. We confirmed these results in p27 null mouse embryonic fibroblasts corroborating the specificity of the p27 role in Aplidin response because p21(waf1) null mouse embryonic fibroblasts do not show this increased sensitivity. We propose a mechanism of action of Aplidin involving p27 and support the analysis of p27 in the response to Aplidin in currently ongoing clinical trials to establish the levels of this protein as response predictor.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Depsipéptidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Ensayos de Selección de Medicamentos Antitumorales , Flavonoides/farmacología , Humanos , Ratones , Péptidos Cíclicos , Piperidinas/farmacología , Vinblastina/farmacología
10.
J Inorg Biochem ; 101(4): 551-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17257684

RESUMEN

The new trans-Pt complexes, derived from trans-[PtCl2(amine)(dimethylamine)] and trans-[PtCl2(OH)2(amine)(dimethylamine)], were synthesized and characterized studying the structure-activity relationship and testing their antiproliferative activity. Their evaluation as cytotoxic agents towards different cancer and normal cell lines is presented. These compounds are active in a panel of tumor cell lines at low micromolar range. Compounds seems to be more active in tumoral than in normal primary human cell lines. Cytotoxic activity is closely related to the amine ligand. Cyclohexylamine ligand was the most active among the amine-ligands tested. Cytotoxic activity correlates with an increase in annexin V positive cells indicating an apoptotic effect of the compounds. Mechanistically, the antitumor activity correlates with a blockade of the cell cycle in S phase and a complete abolishment of G2/M checkpoint arrest suggesting physical interaction of compound with DNA inhibiting S phase transition.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Ciclohexilaminas/farmacología , Compuestos Organometálicos/farmacología , Compuestos Organoplatinos/farmacología , Platino (Metal)/química , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Ciclohexilaminas/química , Humanos , Compuestos Organometálicos/química , Compuestos Organoplatinos/química , Relación Estructura-Actividad
11.
J Cell Biochem ; 100(2): 339-48, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16888811

RESUMEN

Yondelis (Trabectedin, ET-743) is a marine anticancer agent currently in Phase II/III development in patients with advanced pretreated soft tissue sarcoma. In the present study, we generated a panel of low passaged tumor cell lines from samples explanted from chemonaive sarcoma patients with different tumor types. We assessed in vitro sensitivity/resistance to Trabectedin and doxorubicin in a panel of sarcoma cell lines and examined the correlation between molecular alterations in DNA repair genes and sensitivity to Trabectedin. We treated cell lines with Trabectedin and doxorubicin in both 96-h and clonogenic assays. In both assays, well-defined groups of resistant and sensitive cell lines were observed. Resistance to Trabectedin did not correlate with resistance to doxorubicin, indicating that the two drugs may have different mechanisms of resistance. p53 mutations and deletions correlated with extreme sensitivity (IC50 < 1 nM) to Trabectedin (P < 0.01). In a pair of isogenic cell lines differing only in the presence or absence of wild-type p53, the absence of p53 rendered cells threefold more sensitive to Trabectedin.


Asunto(s)
Dioxoles/farmacología , Sarcoma/metabolismo , Tetrahidroisoquinolinas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Proliferación Celular/efectos de los fármacos , Dioxoles/toxicidad , Doxorrubicina/farmacología , Humanos , Cariotipificación , Mutación/genética , ARN Interferente Pequeño/genética , Sarcoma/genética , Sarcoma/patología , Sensibilidad y Especificidad , Tetrahidroisoquinolinas/toxicidad , Trabectedina , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
12.
Anticancer Drugs ; 16(9): 977-87, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16162974

RESUMEN

Apoptotic pathways, including the phosphatidylinositol-3-kinase (PI3K)/AKT survival pathway, are altered in most cancer cells in relation to their normal counterparts and these differences may present an excellent therapeutic window. To gain insight into the relevance of the PI3K pathway as a target for drug discovery we generated tumor cell lines from different tumor samples that we maintained at low passage. The characterization of these cell lines indicates that all of them have constitutively activated the PI3K pathway through different mechanisms. All cell lines were differentially sensitive to the PI3K inhibitor LY294002. Our data also support previous work indicating that PI3K inhibition might help classical chemotherapeutic treatments such as gemcitabine and strengthen suboptimal doses that might be effective for these purposes in decreasing the risk of side-effects. Finally, the analysis of the molecular markers that might be implicated in the synergism between LY294002 and gemcitabine suggests that PI3K inhibition might aid chemotherapeutic treatment, leading to changes in the balance between anti- and pro-apoptotic molecules of the Bcl-2 family, Bcl-XL and Bax. These results facilitate the exploration of potential synergism between chemotherapeutic treatment and the search for others that can account for similar molecular mechanisms of cooperation.


Asunto(s)
Cromonas/farmacología , Desoxicitidina/análogos & derivados , Mitocondrias/efectos de los fármacos , Morfolinas/farmacología , Neoplasias/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteína X Asociada a bcl-2/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Proteínas de Unión al ADN/genética , Desoxicitidina/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Genes Supresores de Tumor , Humanos , Concentración 50 Inhibidora , Leiomiosarcoma/genética , Leiomiosarcoma/metabolismo , Leiomiosarcoma/patología , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Mitocondrias/metabolismo , Neoplasias/genética , Neoplasias/patología , Proteínas Nucleares/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor , Proteína bcl-X/metabolismo , Gemcitabina
13.
Mol Cancer Ther ; 4(5): 814-23, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15897246

RESUMEN

Ecteinascidin 743 (ET-743; Yondelis, Trabectedin) is a marine anticancer agent that induces long-lasting objective remissions and tumor control in a subset of patients with pretreated/resistant soft-tissue sarcoma. Drug-induced tumor control is achievable in 22% of such patients, but there is no clear indication of the molecular features correlated with clinical sensitivity/resistance to ET-743. Nine low-passage, soft-tissue sarcoma cell lines, explanted from chemo-naive patients with different patterns of sensitivity, have been profiled with a cDNA microarray containing 6,700 cancer-related genes. The molecular signature of these cell lines was analyzed at baseline and at four different times after ET-743 exposure. The association of levels of TP53 mutation and TP73 expression with ET-743 sensitivity and cell cycle kinetics after treatment was also analyzed. Gene expression profile analysis revealed up-regulation of 86 genes and down-regulation of 244 genes in response to ET-743. The ET-743 gene expression signature identified a group of genes related with cell cycle control, stress, and DNA-damage response (JUNB, ATF3, CS-1, SAT, GADD45B, and ID2) that were up-regulated in all the cell lines studied. The transcriptional signature 72 hours after ET-743 administration, associated with ET-743 sensitivity, showed a more efficient induction of genes involved in DNA-damage response and apoptosis, such as RAD17, BRCA1, PAR4, CDKN1A, and P53DINP1, in the sensitive cell line group. The transcriptional signature described here may lead to the identification of ET-743 downstream mediators and transcription regulators and the proposal of strategies by which ET-743-sensitive tumors may be identified.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Dioxoles/farmacología , Resistencia a Antineoplásicos , Perfilación de la Expresión Génica , Isoquinolinas/farmacología , Sarcoma/metabolismo , Transcripción Genética , Ciclo Celular , Humanos , Cinética , Mutación/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Sarcoma/genética , Tetrahidroisoquinolinas , Trabectedina , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
14.
J Cell Biochem ; 92(3): 514-24, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15156563

RESUMEN

Oncogenic activation in primary murine fibroblasts initiates a senescence-like cell cycle arrest that depends on the p53 tumor suppressor pathway. Conditional p53 activation efficiently induced a reversible cell cycle arrest but was unable to induce features of senescence. In contrast, coexpression of oncogenic ras with p53 produced an irreversible cell cycle arrest that displayed features of cellular senescence. Introduction of a conditional murine p53 allele (p53val135) into double p53/p21-null mouse embryonic fibroblasts showed that p21waf1 was not required for this effect, since p53-/-;p21-/- double-null cells undergo terminal growth arrest with features of senescence following coexpression of oncogenic Ras and p53. Our results indicate that oncogenic activation of the Ras pathway in murine fibroblasts converts p53 into a senescence inducer through a p21waf1-independent mechanism.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Senescencia Celular/fisiología , Embrión de Mamíferos/citología , Fibroblastos/citología , Proteína Oncogénica p21(ras)/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Células Cultivadas , Senescencia Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Proteína Oncogénica p21(ras)/genética , Proteína p53 Supresora de Tumor/genética
15.
J Cell Biochem ; 89(3): 484-99, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12761882

RESUMEN

Although activated macrophages destroy cancer cells more effectively than normal cells, the facility to escape activated macrophages is a characteristic of tumor cells. One of the mechanisms responsible for the specific killing of tumor cells by macrophages is the production of the cytokine tumor necrosis factor alpha (TNF). Therefore, resistance to TNF may provide such cancer cells a selective advantage against host elimination. In the present work we explore the possibility that cyclin D1 overrides the cytostatic effect of TNF. We show that TNF induces p21(waf1) protein in malignant melanoma A375 cells and its binding to CDK2/4 and 6 proteins, and thereby inhibiting the activity of these complexes. This inhibition leads the cells to a G1 arrest. Overexpression of cyclin D1 in these cells makes them insensitive to TNF treatment with the recovery of CDK activity, however, is unable to overcome the inhibitory action of etoposide blocking the cells on G2/M. The bypass of TNF-induced G1 arrest seems to be related to the increase in the stability of cyclin D bound CDK complexes, increasing the total amount of CDK2/4 and 6 complexes and leading to a functional down titration of the p21(waf1) molecules. In these conditions the TNF-induced increase of p21(waf1) is not sufficient to inhibit the high amount of cyclin D-bound complexes. This hypothesis is supported by the fact that a reduction in the levels of p21(waf1) protein, induced by the expression of a mRNA antisense against p21(waf1), is also able to bypass of TNF-induced arrest. Our results confirm that p21(waf1) has an essential role in TNF-induced arrest and that the deregulation of cyclin D1 may be one of the mechanisms to escape physiological signals to restrict tumoral growth.


Asunto(s)
División Celular/fisiología , Ciclina D1/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Ciclina D1/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Humanos , Procesamiento Postranscripcional del ARN , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...