Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Oncogene ; 34(15): 1928-37, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-24882580

RESUMEN

Lysyl oxidase (LOX) is a multifunctional protein required for normal collagen and elastin biosynthesis and maturation. In addition, LOX has complex roles in cancer in which the lysyl oxidase propeptide (LOX-PP) domain of secreted pro-LOX has tumor-suppressor activity, while the active enzyme promotes metastasis. In prostate cancer cell lines, recombinant LOX-PP (rLOX-PP) inhibits the growth of PC3 cells in vitro by mechanisms that were not characterized, while in DU145 cells rLOX-PP targeted fibroblast growth factor signaling. Because rLOX-PP can enhance effects of a genotoxic chemotherapeutic on breast cancer cell apoptosis, we reasoned that rLOX-PP could target DNA repair pathways typically elevated in cancer. Here we demonstrate for the first time that rLOX-PP inhibits prostate xenograft growth in vivo and that activating phosphorylations of the key DNA repair molecules ataxia-telangiectasia mutated (ATM) and checkpoint kinase 2 (CHK2) are inhibited by rLOX-PP expression in vivo. In addition, in vitro studies showed that rLOX-PP inhibits radiation-induced activating phosphorylations of ATM and CHK2 and that exogenously added rLOX-PP protein can localize to the nucleus in both DU145 and PC3 cells. rLOX-PP pull-down studies resulted in detection of a protein complex with the nuclear DNA repair regulator MRE11 in both cell lines, and rLOX-PP localized to radiation-induced nuclear DNA repair foci. Finally, rLOX-PP was shown to sensitize both DU145 and PC3 cells to radiation-induced cell death determined in colony-formation assays. These data provide evidence that rLOX-PP has a nuclear mechanism of action in which it directly interacts with DNA repair proteins to sensitize prostate cancer cells to the effects of ionizing radiation.


Asunto(s)
Reparación del ADN , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Proteína-Lisina 6-Oxidasa/biosíntesis , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Daño del ADN , Humanos , Masculino , Ratones , Ratones Desnudos , Fosforilación , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteína-Lisina 6-Oxidasa/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Oncogene ; 29(2): 201-13, 2010 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-19838210

RESUMEN

Approximately 80% of breast cancers express the estrogen receptor-alpha (ERalpha) and are treated with anti-estrogens. Resistance to these agents is a major cause of mortality. We have shown that estrogen inhibits Notch, whereas anti-estrogens or estrogen withdrawal activate Notch signaling. Combined inhibition of Notch and estrogen signaling has synergistic effects in ERalpha-positive breast cancer models. However, the mechanisms whereby Notch-1 promotes the growth of ERalpha-positive breast cancer cells are unknown. Here, we demonstrate that Notch-1 increases the transcription of ERalpha-responsive genes in the presence or absence of estrogen via a novel chromatin crosstalk mechanism. Our data support a model in which Notch-1 can activate the transcription of ERalpha-target genes via IKKalpha-dependent cooperative chromatin recruitment of Notch-CSL-MAML1 transcriptional complexes (NTC) and ERalpha, which promotes the recruitment of p300. CSL binding elements frequently occur in close proximity to estrogen-responsive elements (EREs) in the human and mouse genomes. Our observations suggest that a hitherto unknown Notch-1/ERalpha chromatin crosstalk mediates Notch signaling effects in ERalpha-positive breast cancer cells and contributes to regulate the transcriptional functions of ERalpha itself.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/fisiología , Quinasa I-kappa B/genética , Receptor Notch1/fisiología , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Quinasa I-kappa B/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oligopéptidos/farmacología , Regiones Promotoras Genéticas/genética , Receptor Notch1/genética , Receptor Notch1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tamoxifeno/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Oncogene ; 28(38): 3390-400, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19597471

RESUMEN

Enhanced RAS signaling and decreased androgen dependence of prostate cancer cells accompany poor clinical outcomes. Elevated autocrine fibroblast growth factors 2 (FGF-2) signaling promotes prostate cancer cell growth and survival. Expression of lysyl oxidase (LOX) inhibits RAS transforming activity. LOX is secreted as 50 kDa pro-LOX protein and then undergoes extracellular proteolytic processing to form approximately 30 kDa LOX enzyme and approximately 18 kDa propeptide (LOX-PP). We have previously shown that LOX-PP inhibits breast cancer cell transformation and tumor formation, but mechanisms of action of LOX-PP have not been fully elucidated. Here we report that LOX expression is reduced in prostate cancer cell lines and that recombinant LOX-PP protein inhibits serum-stimulated DNA synthesis and MEK/ERK and PI3K/AKT pathways in DU 145 and PC-3 androgen-independent cell lines. In DU 145 cells, treatment with a pharmacologic FGF-receptor inhibitor or a neutralizing anti-FGFR1 antibody mimicked LOX-PP inhibition of serum-stimulated DNA synthesis. FGF-2-stimulated DNA synthesis, ERK1/2, AKT and FRS2alpha activation were found all to be inhibited by LOX-PP in DU 145 cells. LOX-PP reduced specific binding of FGF-2 to DU 145 cells, suggesting that LOX-PP targets FGF signaling at the receptor. Interestingly, PC-3 cells did not respond to FGF-2, consistent with previous reports. We conclude that LOX-PP inhibits proliferation of DU 145 cells by interfering with FGFR(s) binding and signaling, and that LOX-PP has other mechanisms of action in PC-3 cells.


Asunto(s)
Precursores Enzimáticos/farmacología , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Neoplasias de la Próstata/prevención & control , Proteína-Lisina 6-Oxidasa/farmacología , Transducción de Señal , Línea Celular Tumoral , Proliferación Celular , ADN/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Masculino , Próstata/enzimología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes/farmacología
4.
Cell Mol Life Sci ; 66(11-12): 1850-7, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19387549

RESUMEN

CK2 is a highly conserved tetrameric serine/ threonine kinase present in all eukaryotic organisms. It is constitutively active, and appears to be regulated by level of expression and activity, and subcellular localization. In turn, it has been postulated to control the function of many proteins through changes in phosphorylation that affect protein stability, protein-protein interactions, and subcellular localization. Through these mechanisms, CK2 regulates many fundamental cellular properties. An enzyme that carries out such a master regulatory function is likely to be important in organismic development and in cancer. We have shown that overexpression of CK2 catalytic subunits is capable of promoting tumorigenesis, and that loss of CK2 catalytic subunits in development can be lethal. Through studies in cells, mice, and frogs, we and others have identified the Wnt and NF-kappaB pathways as two key signal transduction pathways that are regulated by CK2 activity, in embryonic development and in cancer. These results suggest that inhibiting CK2 could be useful in treating cancer, but dangerous to developing organisms.


Asunto(s)
Quinasa de la Caseína II/fisiología , Transformación Celular Neoplásica/metabolismo , Desarrollo Embrionario/fisiología , FN-kappa B/fisiología , Transducción de Señal/fisiología , Proteínas Wnt/fisiología , Animales , Humanos , Fosforilación
5.
Oncogene ; 27(44): 5833-44, 2008 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-18560356

RESUMEN

Notch-1 inhibits apoptosis in some transformed cells through incompletely understood mechanisms. Notch-1 can increase nuclear factor-kappa B (NF-kappaB) activity through a variety of mechanisms. Overexpression of cleaved Notch-1 in T-cell acute lymphoblastic leukemia cells activates NF-kappaB via interaction with the I kappa B kinase (IKK) signalosome. Concomitant activation of the Notch and NF-kappaB pathways has been described in a large series of cervical cancer specimens. Here, we show that wild-type, spontaneously expressed Notch-1 stimulates NF-kappaB activity in CaSki cervical cancer cells by associating with the IKK signalosome through IKKalpha. A significant fraction of tumor necrosis factor (TNF)-alpha-stimulated IkappaB kinase activity in CaSki cells is Notch-1-dependent. In addition, Notch-1 is found in the nucleus in association with IKKalpha at IKKalpha-stimulated promoters and is required for association of IKKalpha with these promoters under basal and TNF-alpha-stimulated conditions. Notch-1-IKKalpha complexes are found in normal human keratinocytes as well, suggesting that IKK regulation is a physiological function of Notch-1. Both Notch-1 and IKKalpha knockdown sensitize CaSki cells to cisplatin-induced apoptosis to equivalent extents. Our data indicate that Notch-1 regulates NF-kappaB in cervical cancer cells at least in part via cytoplasmic and nuclear IKK-mediated pathways.


Asunto(s)
Quinasa I-kappa B/metabolismo , Receptor Notch1/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Cromatina/metabolismo , Cisplatino/farmacología , Femenino , Humanos , Quinasa I-kappa B/genética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Receptor Notch1/genética , Receptor Notch2/metabolismo , Receptor Notch4 , Receptores Notch/metabolismo
6.
J Cell Biochem ; 82(3): 387-98, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11500915

RESUMEN

Epidemiological evidence suggests tea (Camellia sinensis L.) has chemopreventive effects against various tumors. Green tea contains many polyphenols, including epigallocatechin-3 gallate (EGCG), which possess anti-oxidant qualities. Reduction of chemically induced mammary gland carcinogenesis by green tea in a carcinogen-induced rat model has been suggested previously, but the results reported were not statistically significant. Here we have tested the effects of green tea on mammary tumorigenesis using the 7,12-dimethylbenz(a)anthracene (DMBA) Sprague-Dawley (S-D) rat model. We report that green tea significantly increased mean latency to first tumor, and reduced tumor burden and number of invasive tumors per tumor-bearing animal; although, it did not affect tumor number in the female rats. Furthermore, we show that proliferation and/or viability of cultured Hs578T and MDA-MB-231 estrogen receptor-negative breast cancer cell lines was reduced by EGCG treatment. Similar negative effects on proliferation were observed with the DMBA-transformed D3-1 cell line. Growth inhibition of Hs578T cells correlated with induction of p27(Kip1) cyclin-dependent kinase inhibitor (CKI) expression. Hs578T cells expressing elevated levels of p27(Kip1) protein due to stable ectopic expression displayed increased G1 arrest. Thus, green tea had significant chemopreventive effects on carcinogen-induced mammary tumorigenesis in female S-D rats. In culture, inhibition of human breast cancer cell proliferation by EGCG was mediated in part via induction of the p27(Kip1) CKI.


Asunto(s)
Antineoplásicos/farmacología , Carcinógenos/farmacología , Flavonoides , Neoplasias Mamarias Animales/inducido químicamente , Neoplasias Mamarias Animales/patología , Extractos Vegetales/farmacología , , Proteínas Supresoras de Tumor , 9,10-Dimetil-1,2-benzantraceno/antagonistas & inhibidores , 9,10-Dimetil-1,2-benzantraceno/farmacología , Animales , Antineoplásicos/uso terapéutico , Carcinógenos/antagonistas & inhibidores , Catequina/análogos & derivados , Catequina/farmacología , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Femenino , Citometría de Flujo , Neoplasias Mamarias Animales/enzimología , Fenoles/metabolismo , Extractos Vegetales/uso terapéutico , Polímeros/metabolismo , Probabilidad , Ratas , Células Tumorales Cultivadas
7.
Hepatology ; 34(1): 32-41, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11431731

RESUMEN

NF-kappaB regulates liver cell death during development, regeneration, and neoplastic transformation. For example, we showed that oncogenic Ras- or Raf-mediated transformation of rat liver epithelial cells (RLEs) led to altered NF-kappaB regulation through IKK complex activation, which rendered these cells more resistant to TGF-beta1-induced apoptosis. Thus, based on these findings, we sought to determine whether NF-kappaB could also be involved in tumor growth of liver cells in vivo. Hepatocellular carcinomas (HCCs) derived from bitransgenic mice harboring TGF-alpha and c-myc transgenes targeted specifically to the liver were compared with HCCs from c-myc single transgenic mice. Tumors from bitransgenic mice are characterized by a higher frequency of appearance, lower apoptotic index, and a higher rate of cell proliferation. Here we show that NF-kappaB is activated in HCCs of double TGF-alpha/c-myc transgenic mice, but not of c-myc single transgenic mice, suggesting that TGF-alpha mediates induction of NF-kappaB. Activation of the IKK complex was observed in the HCCs of double TGF-alpha/c-myc transgenic mice, implicating this pathway in NF-kappaB induction. Lastly, activation of the Akt/protein kinase B (PKB), which has recently been implicated in NF-kappaB activation by PDGF, TNF-alpha, and Ras, was also observed. Importantly, human HCC cell lines similarly displayed NF-kappaB activation. Thus, these studies elucidate an anti-apoptotic mechanism by a TGF-alpha-Akt/PKB-IKK pathway, which likely contributes to survival and proliferation, thereby accelerating c-myc-induced liver neoplastic development in vivo.


Asunto(s)
Proteínas Portadoras/fisiología , Neoplasias Hepáticas Experimentales/metabolismo , FN-kappa B/biosíntesis , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas/fisiología , Factor de Crecimiento Transformador alfa/genética , Animales , Apoptosis , División Celular , Activación Enzimática , Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteínas de Unión al ARN , Factor de Crecimiento Transformador alfa/fisiología
8.
Oncogene ; 20(25): 3247-57, 2001 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-11423974

RESUMEN

Protein kinase CK2 is a ubiquitous and evolutionarily conserved serine/threonine kinase that is upregulated in many human cancers and can serve as an oncogene in lymphocytes. Recently, we have demonstrated that CK2 potentiates Wnt/beta-catenin signaling in mammary epithelial cells. To determine whether CK2 overexpression contributes to mammary tumorigenesis, we have performed comparative studies of human and rat breast cancer specimens and we have engineered transgenic mice with dysregulated expression of CK2alpha in the mammary gland. We find that CK2 is highly expressed in human breast tumor specimens and in carcinogen-induced rat mammary tumors. Overexpression of CK2alpha in the mammary gland of transgenic mice, under control of the MMTV-LTR, causes hyperplasia and dysplasia of the female mammary gland. Thirty per cent of the female MMTV-CK2alpha transgenic mice develop mammary adenocarcinomas at a median of 23 months of age, often associated with Wnt pathway activation, as evidenced by upregulation of beta-catenin protein. NF-kappaB activation and upregulation of c-Myc also occur frequently. Thus, in mice, rats, and humans, dysregulated expression of CK2 is associated with and is capable of contributing to mammary tumorigenesis. Targeted inhibition of CK2 could be useful in the treatment of breast cancer.


Asunto(s)
Adenocarcinoma/etiología , Neoplasias de la Mama/etiología , Transformación Celular Neoplásica , Neoplasias Mamarias Animales/etiología , Proteínas Serina-Treonina Quinasas/biosíntesis , Transactivadores , Proteínas de Pez Cebra , Edad de Inicio , Animales , Quinasa de la Caseína II , Proteínas del Citoesqueleto/metabolismo , Femenino , Enfermedad Fibroquística de la Mama , Humanos , Hiperplasia , Lactancia , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/inducido químicamente , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Proteínas Wnt , beta Catenina
9.
Cancer Res ; 61(9): 3810-8, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11325857

RESUMEN

Nuclear factor-kappaB (NF-kappaB)/Rel transcription factors regulate genes that control cell proliferation, survival, and transformation. In normal breast epithelial cells, NF-kappaB/Rel proteins are mainly sequestered in the cytoplasm bound to one of the specific inhibitory IkappaB proteins, whereas in breast cancers they are activated aberrantly. Human breast tumor cell lines, carcinogen-transformed mammary epithelial cells, and the majority of primary human or rodent breast tumor tissue samples express constitutively high levels of nuclear NF-kappaB/REL: To begin to understand the mechanism of this aberrant NF-kappaB/Rel expression, in this study we measured the activity of the major kinases implicated in regulation of IkappaB stability, namely IKKalpha, IKKbeta, and protein kinase, CK2 (formerly casein kinase II). Hs578T, D3-1, and BP-1 breast cancer cell lines displayed higher levels of IKKalpha, IKKbeta, and CK2 activity than untransformed MCF-10F mammary epithelial cells. Inhibition of IKK activity upon expression of dominant negative kinases or of CK2 activity by treatment with selective inhibitors decreased NF-kappaB/Rel activity in breast cancer cells. Inactivation of the IkappaB kinase complex in Hs578T cells via expression of a dominant negative IKKgamma/NF-kappaB essential modulator/IKK-associated protein 1 reduced soft agar colony growth. Thus, the aberrant expression of CK2 or IKK kinases promotes increased nuclear levels of NF-kappaB/Rel and transformation of breast cancer cells. Furthermore, primary human breast cancer specimens that displayed aberrant constitutive expression of NF-kappaB/Rel were found to exhibit increased CK2 and/or IKK kinase activity. These observations suggest these kinases play a similar role in an intracellular signaling pathway that leads to the elevated NF-kappaB/Rel levels seen in primary human mammary tumors and, therefore, represent potential therapeutic targets in the treatment of patients with breast cancer.


Asunto(s)
Neoplasias de la Mama/enzimología , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias de la Mama/patología , Quinasa de la Caseína II , Adhesión Celular/fisiología , División Celular/fisiología , Supervivencia Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Activación Enzimática , Humanos , Quinasa I-kappa B , FN-kappa B/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Células Tumorales Cultivadas
10.
Oncogene ; 20(11): 1287-99, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11313873

RESUMEN

The Nuclear Factor (NF)-kappaB family of transcription factors controls expression of genes which promote cell growth, survival, and neoplastic transformation. Recently we demonstrated aberrant constitutive activation of NF-kappaB in primary human and rat breast cancer specimens and in cell lines. Overexpression of the epidermal growth factor receptor (EGFR) family member Her-2/neu, seen in approximately 30% of breast cancers, is associated with poor prognosis. Previously, Her-2/neu has been shown to signal via a phosphatidylinositol 3 (PI3)-kinase to Akt/protein kinase B (PKB) pathway. Since this signaling pathway was recently shown to activate NF-kappaB, here we have tested the hypothesis that Her-2/neu can activate NF-kappaB in breast cancer. Overexpression of Her-2/neu and EGFR-4 in Ba/F3 cells led to constitutive PI3- and Akt kinase activities, and induction of classical NF-kappaB (p50/p65). Similarly, a tumor cell line and tumors derived from MMTV-Her-2/neu transgenic mice displayed elevated levels of classical NF-kappaB. Engagement of Her-2/neu receptor downregulated the level of NF-kappaB. NF-kappaB binding and activity in the cultured cells was reduced upon inhibition of the PI3- to Akt kinase signaling pathway via ectopic expression of kinase inactive mutants, incubation with wortmannin, or expression of the tumor suppressor phosphatase PTEN. Inhibitors of calpain, but not the proteasome, blocked IkappaB-alpha degradation. Inhibition of Akt did not affect IKK activity. These results indicate that Her-2/neu activates NF-kappaB via a PI3- to Akt kinase signaling pathway that can be inhibited via the tumor suppressor PTEN, and is mediated by calpain rather than the IkappaB kinase complex.


Asunto(s)
Neoplasias de la Mama/metabolismo , Genes Supresores de Tumor , Proteínas I-kappa B , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas , Receptor ErbB-2/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor , Animales , Calpaína/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Transgénicos , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Fosfohidrolasa PTEN , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Células Tumorales Cultivadas
11.
Oncogene ; 20(14): 1688-702, 2001 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-11313917

RESUMEN

Upon engagement of the B Cell Receptor (BCR) of WEHI 231 immature B cells, a drop in c-Myc expression is followed by activation of the cyclin-dependent kinase inhibitor (CKI) p27(Kip1), which induces growth arrest and apoptosis. Here, we report inverse patterns of p27 and c-Myc protein expression follow BCR engagement. We present evidence demonstrating, for the first time, that the p27(Kip1) gene is a target of transcriptional repression by c-Myc. Specifically, the changes in p27 protein levels correlated with changes in p27 mRNA levels, and gene transcription. Induction of p27 promoter activity followed BCR engagement of WEHI 231 cells, and this induction could be repressed upon co-transfection of a c-Myc expression vector. Inhibition of the TATA-less p27 promoter by c-Myc was also observed in Jurkat T cells, vascular smooth muscle, and Hs578T breast cancer cells, extending the observation beyond immune cells. Consistent with a putative Inr element CCAGACC (where +1 is underlined) at the start site of transcription in the p27 promoter, deletion of Myc homology box II reduced the extent of repression. Furthermore, enhanced repression was observed upon transfection of the c-Myc 'super-repressor', with mutation of Phe115 to Leu. The sequences mediating transcriptional activity and c-Myc repression were mapped to bp -20 to +20 of the p27 gene. Finally, binding of Max was shown to facilitate c-Myc binding and repression of p27 promoter activity. Overall, these studies identify the p27 CKI gene as a new target whereby c-Myc can control cell proliferation, survival and neoplastic transformation.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Asociadas a Microtúbulos/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteínas Represoras/fisiología , Factores de Transcripción , Proteínas Supresoras de Tumor , Animales , Anticuerpos Antiidiotipos/inmunología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Transformación Celular Neoplásica , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Proteínas de Unión al ADN/metabolismo , Fase G1 , Humanos , Músculo Liso Vascular/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Receptores de Antígenos de Linfocitos B/fisiología
12.
Endocrinology ; 142(1): 147-56, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11145577

RESUMEN

Dependence of murine pro-B Ba/F3 cells on interleukin-3 can be substituted by GH when cells are stably transfected with the GH receptor (GHR) complementary DNA. Recently, we demonstrated that Ba/F3 cells produce GH, which is responsible for the survival of cells expressing the GHR. This GH effect involves the activation of nuclear factor-kappaB (NF-kappaB). Here, we examined the signaling pathways mediating proliferation of growth factor-deprived Ba/F3 GHR cells. Exogenous GH stimulation of Ba/F3 GHR cells induced cyclins E and A and the cyclin-dependent kinase inhibitor p21(waf1/cip1) and repressed cyclin-dependent kinase inhibitor p27(kip1). The presence of the phosphatidylinositol 3-kinase (PI 3-kinase) inhibitor Ly 294002 abolished proliferation induced by GH, arresting Ba/F3 GHR cells at the G(1)/S boundary, but did not promote apoptosis. Thus, the proliferative effect of GH is closely related to PI 3-kinase activation, whereas PI 3-kinase is not essential for GH-induced cell survival. Addition of Ly 294002 resulted in a moderate decrease in NF-kappaB activation by GH, suggesting a possible link between PI 3-kinase and NF-kappaB signaling by GH. Expression of c-myc was also induced by GH in Ba/F3 GHR cells, and inactivation of either PI 3-kinase or NF-kappaB reduced this induction. Overexpression of the dominant negative repressor mutant c-Myc-RX resulted in an inhibition of the GH proliferative effect, suggesting the involvement of c-myc in GH-induced proliferation. Taken together, these results suggest that the effects of GH on cell survival and proliferation are mediated through two different signaling pathways, NF-kappaB and PI 3-kinase, respectively; although cross-talk between them has not been excluded. NF-kappaB, which has been shown to be responsible for the antiapoptotic effect of GH, could also participate in GH-induced proliferation, as c-myc expression is promoted by PI 3-kinase, in an NF-kappaB-dependent and -independent manner.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Hormona del Crecimiento/farmacología , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Somatotropina/fisiología , Transducción de Señal/fisiología , Animales , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Fase G1 , Genes Reporteros , Genes myc , Luciferasas/genética , Ratones , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Ratas , Receptores de Somatotropina/efectos de los fármacos , Receptores de Somatotropina/genética , Fase S , Transducción de Señal/efectos de los fármacos , Transfección
13.
Mol Cell Biochem ; 227(1-2): 153-65, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11827167

RESUMEN

Breast cancer is a major cause of cancer death in women, and the genetic abnormalities leading to the common sporadic forms of the disease are still under active investigation. CK2 has been reported to be upregulated in human breast cancer, which these studies confirm; CK2 is also upregulated in rat carcinogen-induced breast tumors. Transgenic mice overexpressing CK2alpha in the mammary gland develop mammary hyperplasia, dysplasia, and eventually adenocarcinomas, demonstrating that dysregulated expression of CK2 can contribute to transformation of the mammary epithelium. These mammary tumors have evidence of activation of the Wnt and NFkappaB pathways and upregulation of c-Myc. CK2 is capable of phosphorylating the key signaling molecule in the Wnt pathway, the transcriptional cofactor beta-catenin, and regulating its turnover. CK2 is known to phosphorylate IkappaB and thereby regulate basal NFkappaB levels; in the mammary cell lines and tumors, CK2 activity correlates with NFkappaB levels and inhibition of CK2 downregulates NFkappaB. Thus, CK2 may promote breast cancer through dysregulation of key pathways of transcriptional control in the mammary epithelium, and inhibition of CK2 has a potential role in the treatment of breast and other cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias Mamarias Animales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas de Pez Cebra , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Apigenina , Western Blotting , Quinasa de la Caseína II , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Flavonoides/farmacología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/metabolismo , Neoplasias Experimentales , Fosforilación , Pruebas de Precipitina , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Regulación hacia Arriba , Proteínas Wnt
14.
Oncogene ; 19(48): 5498-506, 2000 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-11114727

RESUMEN

NF-kappaB/Rel transcription factors regulate many genes involved in control of cellular proliferation, neoplastic transformation, and apoptosis, including the c-myc oncogene. Recently, we have observed that levels of NF-kappaB and aryl hydrocarbon receptor (AhR), which mediates malignant transformation by environmental carcinogens, are highly elevated and appear constitutively active in breast cancer cells. Rel factors have been found to functionally interact with other transcription factors. Here we demonstrate a physical and functional association between the RelA subunit of NF-kappaB and AhR resulting in the activation of c-myc gene transcription in breast cancer cells. RelA and AhR proteins were co-immunoprecipitated from cytoplasmic and nuclear extracts of non-malignant MCF-10F breast epithelial and malignant Hs578T breast cancer cells. In transient co-transfection, RelA and AhR gene products demonstrated cooperation in transactivation of the c-myc promoter, which was dependent on the NF-kappaB elements, and in induction of endogenous c-Myc protein levels. A novel AhR/RelA-containing NF-kappaB element binding complex was identified by electrophoretic mobility shift analysis of nuclear extracts from RelA and AhR co-transfected Hs578T cells. Thus, the RelA and AhR proteins functionally cooperate to bind to NF-kappaB elements and induce c-myc gene expression. These findings suggest a novel signaling mechanism whereby the Ah receptor can stimulate proliferation and tumorigenesis of mammary cells.


Asunto(s)
Mama/fisiología , Genes myc/genética , FN-kappa B/fisiología , Receptores de Hidrocarburo de Aril/fisiología , Activación Transcripcional/fisiología , Sitios de Unión , Mama/citología , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , División Celular/genética , Línea Celular , Transformación Celular Neoplásica/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Pruebas de Precipitina , Regiones Promotoras Genéticas/genética , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Factor de Transcripción ReIA , Transfección , Células Tumorales Cultivadas
15.
Breast Cancer Res Treat ; 63(2): 117-31, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11097088

RESUMEN

Exposure to ubiquitous environmental chemicals, such as polycyclic aromatic hydrocarbons (PAH), may contribute to human breast cancer. In animals, PAH induce tumors in part by activating the aryl hydrocarbon receptor (AhR)/transcription factor. Historically, investigations into AhR-regulated carcinogenesis have focused on AhR-dependent transcriptional regulation of cytochrome P450 (CYP) enzymes which oxidize PAH to mutagenic intermediates. However, recent studies suggest that the AhR directly regulates cell growth. Given the postulated role of the AhR in carcinogenesis, we predicted that: (1) tissue predisposed to PAH tumorigenesis would express the AhR and (2) aberrant AhR and/or AhR-regulated gene expression would accompany malignant transformation. To test these hypotheses, AhR and CYP1 protein and/or mRNA levels were evaluated in rat mammary tumors induced with 7, 12-dimethylbenz[a]anthracene (DMBA), a prototypic PAH and AhR ligand. Results indicate modest AhR expression in normal mammary myoepithelial and ductal epithelial cells. In contrast, high AhR levels were detected in DMBA-induced tumors. Nuclear AhR localization in tumors suggested constitutive AhR activation. In situ hybridization and quantitative RT-PCR assays indicated high AhR mRNA levels in neoplastic epithelial cells. While both AhR-regulated CYP1A1 and CYP1B1 mRNAs were induced in breast tissue within 6 h of DMBA gavage, only CYP1B1 mRNA remained elevated in tumors. These results: (1) help explain targeting of breast tissue by carcinogenic PAH, (2) imply that AhR and CYP1B1 hyper-expression represent molecular biomarkers for, at least, PAH-induced mammary cell transformation, and (3) suggest mechanisms through which the AhR may contribute to carcinogenesis well after exogenous AhR ligands have been eliminated.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas , Citocromo P-450 CYP1A1/genética , Sistema Enzimático del Citocromo P-450/genética , Neoplasias Mamarias Experimentales/metabolismo , ARN Mensajero/análisis , Receptores de Hidrocarburo de Aril/análisis , 9,10-Dimetil-1,2-benzantraceno , Animales , Citocromo P-450 CYP1B1 , Femenino , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/inducido químicamente , Ratas , Ratas Sprague-Dawley
16.
Blood ; 96(3): 1013-20, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10910917

RESUMEN

A-myb is a member of the myb family of transcription factors, which regulates proliferation, differentiation, and apoptosis of hematopoietic cells. A-Myb expression is normally restricted to the proliferating B-cell centroblasts and transgenic mice overexpressing A-myb displayed enhanced hyperplasia of the lymph nodes. Because A-Myb is highly expressed in several subtypes of human B-cell neoplasias, we sought to determine whether the A-myb gene promoted proliferation and survival of B lymphocytes, using the WEHI 231 and CH33 murine B-cell lymphomas as models. Here, we show that ectopic expression of A-myb rescues WEHI 231 and CH33 cells from growth arrest and apoptosis induced by anti-IgM treatment. Previously, we demonstrated an essential role of the c-myc gene in promoting cell survival of WEHI 231 cells in response to a variety of apoptotic stimuli. Furthermore, we and others have shown that the c-myc gene is potently transactivated by A-Myb in several cell types. Thus, we sought to determine whether c-Myc would mediate the A-Myb antiapoptotic effect in B cells. Here we show that ectopic expression of A-myb leads to maintenance of c-myc expression, and that expression of antisense c-myc RNA ablates A-Myb-mediated survival signals. Thus, these findings strongly implicate the A-myb gene in the regulation of B-cell survival and confirm the c-myc gene as one of the downstream targets of A-myb in these cells. Overall, our observation suggests that A-myb expression may be relevant to the pathology of human B-cell neoplasias.


Asunto(s)
Apoptosis/genética , Genes myb , Genes myc , Linfoma de Células B/genética , Linfoma de Células B/patología , Receptores Fc/genética , Animales , Apoptosis/inmunología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-myb , Receptores Fc/inmunología , Transactivadores/genética , Transcripción Genética
17.
Mol Cell Biol ; 20(15): 5381-91, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10891479

RESUMEN

NF-kappaB/Rel factors have been implicated in the regulation of liver cell death during development, after partial hepatectomy, and in hepatocytes in culture. Rat liver epithelial cells (RLEs) display many biochemical and ultrastructural characteristics of oval cells, which are multipotent cells that can differentiate into mature hepatocytes. While untransformed RLEs undergo growth arrest and apoptosis in response to transforming growth factor beta1 (TGF-beta1) treatment, oncogenic Ras- or Raf-transformed RLEs are insensitive to TGF-beta1-mediated growth arrest. Here we have tested the hypothesis that Ras- or Raf-transformed RLEs have altered NF-kappaB regulation, leading to this resistance to TGF-beta1. We show that classical NF-kappaB is aberrantly activated in Ras- or Raf-transformed RLEs, due to increased phosphorylation and degradation of IkappaB-alpha protein. Inhibition of NF-kappaB activity with a dominant negative form of IkappaB-alpha restored TGF-beta1-mediated cell killing of transformed RLEs. IKK activity mediates this hyperphosphorylation of IkappaB-alpha protein. As judged by kinase assays and transfection of dominant negative IKK-1 and IKK-2 expression vectors, NF-kappaB activation by Ras appeared to be mediated by both IKK-1 and IKK-2, while Raf-induced NF-kappaB activation was mediated by IKK-2. NF-kappaB activation in the Ras-transformed cells was mediated by both the Raf and phosphatidylinositol 3-kinase pathways, while in the Raf-transformed cells, NF-kappaB induction was mediated by the mitogen-activated protein kinase cascade. Last, inhibition of either IKK-1 or IKK-2 reduced focus-forming activity in Ras-transformed RLEs. Overall, these studies elucidate a mechanism that contributes to the process of transformation of liver cells by oncogene Ras and Raf through the IkappaB kinase complex leading to constitutive activation of NF-kappaB.


Asunto(s)
Transformación Celular Neoplásica/genética , Genes ras , Proteínas I-kappa B/metabolismo , Hígado/patología , Proteínas Oncogénicas de Retroviridae/genética , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Transformada , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Proteínas I-kappa B/antagonistas & inhibidores , Proteínas I-kappa B/genética , Hígado/efectos de los fármacos , FN-kappa B/metabolismo , Proteínas Oncogénicas v-raf , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Ratas , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
18.
Mol Endocrinol ; 14(5): 650-61, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10809229

RESUMEN

The pro-B Ba/F3 cell line requires interleukin-3 and serum for growth, and their removal results in cell apoptosis. Ba/F3 cells transfected with the GH receptor (GHR) cDNA become able to proliferate in response to GH. To investigate the role of GH in the control of apoptosis, Ba/F3 cells expressing either the wild-type rat GHR (Ba/F3 GHR) or a mutated rat GHR (Ba/F3 ILV/T) were used. We show that Ba/F3 GHR cells, but not parental Ba/F3 or Ba/F3 ILV/T cells, were able to survive in the absence of growth factor. Furthermore, an autocrine/paracrine mode of GH action was suggested by the demonstration that Ba/F3 cells produce GH, and that addition of GH antagonists (B2036 and G120K) promotes apoptosis of Ba/F3 GHR cells. Consistent with survival, the levels of both antiapoptotic proteins Bcl-2 and Bag-1 were maintained in Ba/F3 GHR cells, but not in parental Ba/F3 cells upon growth factor deprivation. Constitutive activation of the transcription factor nuclear factor-kappaB (NF-kappaB), which has been shown to promote cell survival, was sustained in Ba/F3 GHR cells, whereas no NF-kappaB activation was detected in parental Ba/F3 cells in the absence of growth factor. Furthermore, addition of GH induced NF-kappaB DNA binding activity in Ba/F3 GHR cells. Overexpression of the mutated IkappaB alpha (A32/36) protein, known to inhibit NF-kappaB activity, resulted in death of growth factor-deprived Ba/F3 GHR cells, and addition of GH was no longer able to rescue these cells from apoptosis. Together, our results provide evidence for a new GH-mediated pathway that initiates a survival signal through activation of the transcription factor NF-kappaB and sustained levels of the antiapoptotic proteins Bcl-2 and Bag-1.


Asunto(s)
Apoptosis/efectos de los fármacos , Hormona del Crecimiento/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Proteínas I-kappa B , Interleucina-3/farmacología , FN-kappa B/fisiología , Receptores de Somatotropina/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Animales , Comunicación Autocrina , Proteínas Portadoras/biosíntesis , Bovinos , Ciclo Celular/efectos de los fármacos , Línea Celular , Medios de Cultivo/farmacología , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Depresión Química , Dimerización , Genes bcl-2 , Hormona del Crecimiento/antagonistas & inhibidores , Células Madre Hematopoyéticas/citología , Ratones , Inhibidor NF-kappaB alfa , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ratas , Receptores de Somatotropina/genética , Receptores de Somatotropina/metabolismo , Factores de Transcripción , Transfección , Proteína bcl-X
19.
Carcinogenesis ; 21(5): 871-9, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10783306

RESUMEN

NF-kappaB/Rel is a family of transcription factors which are expressed in all cells; however, in most non-B cells, they are sequestered in the cytoplasm in inactive complexes with specific inhibitory proteins, termed IkappaBs. We have recently shown that NF-kappaB/Rel factors are aberrantly activated in human breast cancer and rodent mammary tumors, and function to promote tumor cell survival and proliferation. Here, we have examined the time-course of induction of NF-kappaB/Rel factors upon carcinogen treatment of female Sprague-Dawley (S-D) rats in vivo and in human mammary epithelial cells (HMECs) in culture. We observed that NF-kappaB/Rel activation is an early event, occurring prior to malignant transformation. In S-D rats, increased NF-kappaB/Rel binding was detected in nuclear extracts of mammary glands from 40% of animals 3 weeks post-treatment with 15 mg/kg 7,12-dimethylbenz[a]anthracene (DMBA); this is prior to formation of tumors which normally begin to be detected after 7-9 weeks. In non-tumorigenic MCF-10F cells, in vitro malignant transformation upon treatment with either DMBA or benzo[a]pyrene (B[a]P) resulted in a 4- to 12-fold increase in activity of classical NF-kappaB (p65/p50). NF-kappaB induction was corrrelated with a decrease in the stability of the NF-kappaB-specific inhibitory protein IkappaB-alpha. Ectopic expression of the transactivating p65 subunit of NF-kappaB in MCF-10F cells induced the c-myc oncogene promoter, which is driven by two NF-kappaB elements, and endogenous c-Myc levels. Furthermore, reduction mammoplasty-derived HMECs, immortalized following B[a]P exposure, showed dysregulated induction of classical NF-kappaB prior to malignant transformation. Together these findings suggest that activation of NF-kappaB plays an early, critical role in the carcinogen-driven transformation of mammary glands.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias Mamarias Experimentales/metabolismo , FN-kappa B/metabolismo , Animales , Secuencia de Bases , Carcinógenos , Femenino , Genes myc , Humanos , Neoplasias Mamarias Experimentales/patología , Datos de Secuencia Molecular , Fenotipo , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas
20.
J Immunol ; 163(12): 6530-5, 1999 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-10586045

RESUMEN

Engagement of the B cell receptor of WEHI 231 immature B cells leads sequentially to a drop in c-Myc, to induction of the cyclin-dependent kinase inhibitor p27Kip1, and finally to apoptosis. Recently we demonstrated that the drop in c-Myc expression promotes cell death, whereas the induction of p27 has been shown to lead to growth arrest. In this paper, we demonstrate that increased p27 expression also promotes apoptosis of WEHI 231 B cells. The rescue of WEHI 231 cells by CD40 ligand engagement of its receptor prevented the increase in p27 induction. Inhibition of p27-ablated apoptosis induced upon expression of antisense c-myc RNA. Furthermore, specific induction of p27 gene expression resulted in apoptosis of WEHI 231 cells. Lastly, inhibition of expression of c-Myc, upon induction of an antisense c-myc RNA vector, was sufficient to induce increased p27 levels and apoptosis. Thus, these findings define a signaling pathway during B cell receptor engagement in which the drop in c-Myc levels leads to an increase in p27 levels that promotes apoptosis.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Apoptosis/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Proteínas de Ciclo Celular , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/inmunología , Inmunoglobulina M/inmunología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Supresoras de Tumor , Animales , Antígenos CD40/metabolismo , Ligando de CD40 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/fisiología , Inhibidores Enzimáticos/farmacología , Ligandos , Glicoproteínas de Membrana/fisiología , Ratones , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/biosíntesis , Oligonucleótidos Antisentido/biosíntesis , Oligonucleótidos Antisentido/farmacología , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , ARN sin Sentido/biosíntesis , ARN sin Sentido/farmacología , Transducción de Señal/inmunología , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA