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










Base de datos
Intervalo de año de publicación
1.
Clin Transl Oncol ; 24(1): 127-144, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34370182

RESUMEN

Metaplastic breast carcinoma (MBC) is a rare breast cancer subtype with rapid growth, high rates of metastasis, recurrence and drug resistance, and diverse molecular and histological heterogeneity. Patient-derived xenografts (PDXs) provide a translational tool and physiologically relevant system to evaluate tumor biology of rare subtypes. Here, we provide an in-depth comprehensive characterization of a new PDX model for MBC, TU-BcX-4IC. TU-BcX-4IC is a clinically aggressive tumor exhibiting rapid growth in vivo, spontaneous metastases, and elevated levels of cell-free DNA and circulating tumor cell DNA. Relative chemosensitivity of primary cells derived from TU-BcX-4IC was performed using the National Cancer Institute (NCI) oncology drug set, crystal violet staining, and cytotoxic live/dead immunofluorescence stains in adherent and organoid culture conditions. We employed novel spheroid/organoid incubation methods (Pu·MA system) to demonstrate that TU-BcX-4IC is resistant to paclitaxel. An innovative physiologically relevant system using human adipose tissue was used to evaluate presence of cancer stem cell-like populations ex vivo. Tissue decellularization, cryogenic-scanning electron microscopy imaging and rheometry revealed consistent matrix architecture and stiffness were consistent despite serial transplantation. Matrix-associated gene pathways were essentially unchanged with serial passages, as determined by qPCR and RNA sequencing, suggesting utility of decellularized PDXs for in vitro screens. We determined type V collagen to be present throughout all serial passage of TU-BcX-4IC tumor, suggesting it is required for tumor maintenance and is a potential viable target for MBC. In this study we introduce an innovative and translational model system to study cell-matrix interactions in rare cancer types using higher passage PDX tissue.


Asunto(s)
Antineoplásicos/uso terapéutico , Modelos Biológicos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Adipocyte ; 7(2): 96-105, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29411671

RESUMEN

Stromal/stem cell differentiation is controlled by a vast array of regulatory mechanisms. Included within these are methods of mRNA gene regulation that occur at the level of epigenetic, transcriptional, and/or posttranscriptional modifications. Current studies that evaluate the posttranscriptional regulation of mRNA demonstrate microRNAs (miRNAs) as key mediators of stem cell differentiation through the inhibition of mRNA translation. miRNA expression is enhanced during both adipogenic and osteogenic differentiation; however, the mechanism by which miRNA expression is altered during stem cell differentiation is less understood. Here we demonstrate for the first time that adipose-derived stromal/stem cells (ASCs) induced to an adipogenic or osteogenic lineage have differences in strand preference (-3p and -5p) for miRNAs originating from the same primary transcript. Furthermore, evaluation of miRNA expression in ASCs demonstrates alterations in both miRNA strand preference and 5'seed site heterogeneity. Additionally, we show that during stem cell differentiation there are alterations in expression of genes associated with the miRNA biogenesis pathway. Quantitative RT-PCR demonstrated changes in the Argonautes (AGO1-4), Drosha, and Dicer at intervals of ASC adipogenic and osteogenic differentiation compared to untreated ASCs. Specifically, we demonstrated altered expression of the AGOs occurring during both adipogenesis and osteogenesis, with osteogenesis increasing AGO1-4 expression and adipogenesis decreasing AGO1 gene and protein expression. These data demonstrate changes to components of the miRNA biogenesis pathway during stromal/stem cell differentiation. Identifying regulatory mechanisms for miRNA processing during ASC differentiation may lead to novel mechanisms for the manipulation of lineage differentiation of the ASC through the global regulation of miRNA as opposed to singular regulatory mechanisms.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , MicroARNs/biosíntesis , Células Cultivadas , Humanos
3.
Oncogenesis ; 4: e168, 2015 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-26436950

RESUMEN

Liver kinase B1 (LKB1), also known as serine/threonine kinase 11 (STK11), has been identified as a tumor suppressor in many cancers including breast. Low LKB1 expression has been associated with poor prognosis of breast cancer patients, and we report here a significant association between loss of LKB1 expression and reduced patient survival specifically in the basal subtype of breast cancer. Owing to the aggressive nature of the basal subtype as evidenced by high incidences of metastasis, the purpose of this study was to determine if LKB1 expression could regulate the invasive and metastatic properties of this specific breast cancer subtype. Induction of LKB1 expression in basal-like breast cancer (BLBC)/triple-negative breast cancer cell lines, MDA-MB-231 and BT-549, inhibited invasiveness in vitro and lung metastatic burden in an orthotopic xenograft model. Further analysis of BLBC cells overexpressing LKB1 by unbiased whole transcriptomics (RNA-sequencing) revealed striking regulation of metastasis-associated pathways, including cell adhesion, extracellular matrix remodeling, and epithelial-to-mesenchymal transition (EMT). In addition, LKB1 overexpression inhibited EMT-associated genes (CDH2, Vimentin, Twist) and induced the epithelial cell marker CDH1, indicating reversal of the EMT phenotype in the MDA-MB-231 cells. We further demonstrated marked inhibition of matrix metalloproteinase 1 expression and activity via regulation of c-Jun through inhibition of p38 signaling in LKB1-expressing cells. Taken together, these data support future development of LKB1 inducing therapeutics for the suppression of invasion and metastasis of BLBC.

4.
Oncogene ; 30(9): 1082-97, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21057537

RESUMEN

Fulvestrant is a selective estrogen receptor downregulator (SERD) and highly effective antagonist to hormone-sensitive breast cancers following failure of previous tamoxifen or aromatase inhibitor therapies. However, after prolonged fulvestrant therapy, acquired resistance eventually occurs in the majority of breast cancer patients, due to poorly understood mechanisms. To examine a possible role(s) of aberrantly expressed microRNAs (miRNAs) in acquired fulvestrant resistance, we compared antiestrogen-resistant and -sensitive breast cancer cells, revealing the overexpression of miR-221/222 in the SERD-resistant cell lines. Fulvestrant treatment of estradiol (E2)- and fulvestrant-sensitive MCF7 cells resulted in increased expression of endogenous miR-221/222. Ectopic upregulation of miR-221/222 in estrogen receptor-α (ERα)-positive cell lines counteracted the effects of E2 depletion or fulvestrant-induced cell death, thus also conferring hormone-independent growth and fulvestrant resistance. In cells with acquired resistance to fulvestrant, miR-221/222 expression was essential for cell growth and cell cycle progression. To identify possible miR-221/222 targets, miR-221- or miR-222- induced alterations in global gene expression profiles and target gene expression at distinct time points were determined, revealing that miR-221/222 overexpression resulted in deregulation of multiple oncogenic signaling pathways previously associated with drug resistance. Activation of ß-catenin by miR-221/222 contributed to estrogen-independent growth and fulvestrant resistance, whereas TGF-ß-mediated growth inhibition was repressed by the two miRNAs. This first in-depth investigation into the role of miR-221/222 in acquired fulvestrant resistance, a clinically important problem, demonstrates that these two 'oncomirs' may represent promising therapeutic targets for treating hormone-independent, SERD-resistant breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Resistencia a Antineoplásicos , Estradiol/análogos & derivados , MicroARNs/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Fulvestrant , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba , beta Catenina/genética
5.
J Steroid Biochem Mol Biol ; 78(5): 409-18, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11738551

RESUMEN

In oestrogen receptor (ER)-positive breast carcinoma cells, 17beta-oestradiol suppresses a dose-dependent induction of cell death by tumour necrosis factor alpha (TNF). The ability of oestrogens to promote cell survival in ER-positive breast carcinoma cells is linked to a coordinate increase in Bcl-2 expression, an effect that is blocked with the pure anti-oestrogen ICI 182,780. The role of Bcl-2 in MCF-7 cell survival was confirmed by stable overexpression of Bcl-2 which resulted in suppression of apoptosis induced by doxorubicin (DOX), paclitaxel (TAX) and TNF as compared to vector-control cells. The pure anti-oestrogen ICI 182,780 in combination with TNF, DOX or TAX potentiated apoptosis in vector-transfected cells. Interestingly, pre-treatment with ICI 182,780 markedly enhanced chemotherapeutic drug- or TNF-induced apoptosis in Bcl-2 expressing cells, an effect that was correlated with ICI 182,780 induced activation of c-Jun N-terminal kinase. Our results suggest that the effects of oestrogens/anti-oestrogens on the regulation of apoptosis may involve coordinate activation of signalling events and Bcl-2 expression.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Estradiol/análogos & derivados , Estradiol/farmacología , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/patología , Factor de Necrosis Tumoral alfa/farmacología , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Interacciones Farmacológicas , Estradiol/administración & dosificación , Moduladores de los Receptores de Estrógeno/administración & dosificación , Moduladores de los Receptores de Estrógeno/farmacología , Femenino , Fulvestrant , Genes bcl-2 , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Transducción de Señal , Transfección , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/administración & dosificación
6.
Pediatr Clin North Am ; 48(5): 1223-40, x, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11579671

RESUMEN

What do pseudohermaphroditic polar bears and girls with premature breast development have in common? Hormones. Sexual differentiation and the initiation of secondary sexual characteristics, such as breast growth, are both under the control of the sex hormones estrogen and androgen. Abnormal differentiation of the internal or external genitalia in bears and early onset of breast development in girls also may have a common element--exposure to environmental hormones.


Asunto(s)
Trastornos del Desarrollo Sexual/fisiopatología , Enfermedades del Sistema Endocrino/etiología , Enfermedades del Sistema Endocrino/fisiopatología , Exposición a Riesgos Ambientales/efectos adversos , Hormonas Esteroides Gonadales/efectos adversos , Pediatría , Pubertad/fisiología , Diferenciación Sexual/fisiología , Adolescente , Caimanes y Cocodrilos , Animales , Aves , Niño , Salud Ambiental , Femenino , Peces , Humanos , Masculino , Phocidae , Ursidae
7.
Nature ; 413(6852): 128-9, 2001 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-11557969

RESUMEN

Nitrogen fixation is a symbiotic process initiated by chemical signals from legumes that are recognized by soil bacteria. Here we show that some endocrine-disrupting chemicals (EDCs), so called because of their effect on hormone-signalling pathways in animal cells, also interfere with the symbiotic signalling that leads to nitrogen fixation. Our results raise the possibility that these phytochemically activated pathways may have features in common with hormonal signalling in vertebrates, thereby extending the biological and ecological impact of EDCs.


Asunto(s)
Hormonas , Fijación del Nitrógeno , Plaguicidas/farmacología , Fenoles/farmacología , Medicago sativa/microbiología , Medicago sativa/fisiología , Plaguicidas/química , Fenoles/química , Rhizobiaceae/fisiología , Transducción de Señal/efectos de los fármacos , Simbiosis
8.
Surgery ; 130(2): 143-50, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11490342

RESUMEN

BACKGROUND: Nuclear factor-kappa B (NF-kappa B) is a known survival pathway, and it may explain differential sensitivity to tumor necrosis factor-alpha (TNF-alpha) and chemotherapeutic-induced apoptosis in apoptotically sensitive (APO+) and apoptotically resistant (APO-) Michigan Cancer Foundation-7 breast cancer cells. METHODS: Crystal violet viability and luciferase reporter gene assays were used to determine the inhibitory concentration of viability at 50% (IC(50)) and the inhibitory concentration of activity at 50% (EC(50)) values in APO- and APO+ cells with the selective NF-kappa B inhibitor, BAY 11-7082 (BAY). The apoptotic reporter assay was used to determine the effects of the transfection of the inhibitory kappa B-dominant negative (I kappa B-DN) construct in conjunction with TNF, paclitaxel, or doxorubicin treatments in these cells. RESULTS: The concentrations at which 50% of cell viability is inhibited (IC(50)) and at which 50% of NF-kappa B activity is inhibited (EC(50)) for BAY in APO- and APO+ cells were 95.24 micromol/L and 1.53 micromol/L, respectively, and 7.62 micromol/L and 2.64 micromol/L, respectively. The IC(50) and the EC(50) values were equivalent for the APO+ cells (P =.665), but not for the APO- cells (P =.025). I kappa B-DN--transfection alone, or with TNF, doxorubicin, or paclitaxel treatments resulted in cell death of both APO- and APO+ cells as compared with vector-control; however, greater cytotoxicity was seen in the APO+ cells. Direct comparison of the APO+ cells versus the APO- cells revealed that these differences were significant (P =.05). CONCLUSIONS: Pharmacologic or molecular inhibition of the NF-kappa B pathway blocked cell survival in MCF-7 APO+ cells, while only molecular inhibition induced cytotoxicity in the APO- cells. Selective manipulation of the NF-kappa B pathway in combination with standard chemotherapeutic agents may lead to an increased potency and efficacy of these agents.


Asunto(s)
Neoplasias de la Mama , Resistencia a Antineoplásicos , FN-kappa B/genética , FN-kappa B/metabolismo , Nitrilos , Compuestos Orgánicos , Sulfonas , Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Doxorrubicina/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Concentración 50 Inhibidora , Luciferasas/genética , FN-kappa B/antagonistas & inhibidores , Paclitaxel/farmacología , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos
9.
APMIS ; 109(4): 263-72, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11469497

RESUMEN

For three decades, we have known that estrogens alter the development of the mammalian reproductive system in predictable ways. In mice exposed prenatally to diethylstilbestrol (DES) or other estrogens, the male offspring exhibit structural malformations including cryptorchidism, epididymal cysts and retained Mullerian ducts. The estrogen-associated alterations in the genital tract phenotype can be usefully considered as a model called Developmental Estrogenization Syndrome. While estrogen treatment during critical periods of morphogenesis of the male reproductive system has been associated with these changes, the mechanisms at the molecular level are still being discovered. Parallel findings on the hormones involved in Mullerian duct regression and testicular descent have helped guide research on the mechanisms of developmental estrogenization of the male. Cellular localization of molecular signals associated with key steps in genital tract development, use of mice with gene disruption, and knowledge of the mechanisms underlying persistent changes in gene expression are beginning to provide a blue print for both the physiological role and pathological effects of estrogens in reproductive tract development. Since many of the same biological principles underlie genital tract morphogenesis in mammals, one may expect some of the same changes in males of other species exposed to estrogen during the appropriate developmental periods.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Dietilestilbestrol/toxicidad , Estrógenos no Esteroides/toxicidad , Genitales Masculinos/anomalías , Anomalías Inducidas por Medicamentos/embriología , Animales , Metilación de ADN/efectos de los fármacos , Femenino , Genes Reporteros/efectos de los fármacos , Impresión Genómica , Humanos , Masculino , Intercambio Materno-Fetal , Ratones , Fenotipo , Embarazo , Diferenciación Sexual/efectos de los fármacos , Síndrome
10.
Mol Cell Endocrinol ; 176(1-2): 67-75, 2001 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11369444

RESUMEN

Leptin is a potential regulator of conceptus development. We have previously suggested that in primate pregnancy, leptin biosynthesis is regulated by estrogen in a tissue-specific manner. Therefore, the objective of the current study was to determine the mechanism of estrogen action on LEP promoter activation in divergent cell types. The effects of estrogen were investigated in estrogen receptor (ER)-positive MCF-7 breast cancer cells and in ER-negative JEG-3 choriocarcinoma cells. Cells were transfected with a leptin-luciferase or an estrogen responsive element (ERE)-luciferase reporter construct, in conjunction with ERalpha, ERbeta, or empty vector expression plasmids. Cells were treated with estradiol and/or the specific estrogen antagonists, ICI-182,780 or 4-hydroxytamoxifen. In MCF-7 cells, estradiol stimulated (P<0.05) ERE-luciferase activity and was inhibited by ICI-182,780, but did not stimulate leptin-luciferase activity. However, leptin-luciferase was stimulated by estradiol (P<0.05) and inhibited by antiestrogens in JEG-3 cells that were co-transfected with ERalpha. Both antiestrogens stimulated leptin-luciferase activity (P<0.05) in JEG-3 cells co-transfected with ERbeta. Results suggested that LEP promoter activation may depend upon co-activators present in leptin-producing cells and may be inhibited by repressors present in non-leptin producing cells. Divergent effects of estrogen may be owed to differences in the type of ER (alpha or beta) expressed in target tissues.


Asunto(s)
Neoplasias de la Mama/metabolismo , Coriocarcinoma/metabolismo , Estradiol/farmacología , Leptina/genética , Regiones Promotoras Genéticas/genética , Receptores de Estrógenos/metabolismo , Activación Transcripcional/efectos de los fármacos , Estradiol/análogos & derivados , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Fulvestrant , Genes Reporteros/genética , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/genética , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Células Tumorales Cultivadas
11.
J Clin Endocrinol Metab ; 86(4): 1750-8, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11297613

RESUMEN

The flavonoid family of phytochemicals, particularly those derived from soy, has received attention regarding their estrogenic activity as well as their effects on human health and disease. In addition to these flavonoids other phytochemicals, including phytostilbene, enterolactone, and lignans, possess endocrine activity. The types and amounts of these compounds in soy and other plants are controlled by both constitutive expression and stress-induced biosynthesis. The health benefits of soy-based foods may, therefore, be dependent upon the amounts of the various hormonally active phytochemicals within these foods. The aim was to identify unique soy phytochemicals that had not been previously assessed for estrogenic or antiestrogenic activity. Here we describe increased biosynthesis of the isoflavonoid phytoalexin compounds, glyceollins, in soy plants grown under stressed conditions. In contrast to the observed estrogenic effects of coumestrol, daidzein, and genistein, we observed a marked antiestrogenic effect of glyceollins on ER signaling, which correlated with a comparable suppression of 17 beta-estradiol-induced proliferation in MCF-7 cells. Further evaluation revealed greater antagonism toward ER alpha than ER beta in transiently transfected HEK 293 cells. Competition binding assays revealed a greater affinity of glyceollins for ER alpha vs. ER beta, which correlated to greater suppression of ER alpha signaling with higher concentrations of glyceollins. In conclusion, we describe the phytoalexin compounds known as glyceollins, which exhibit unique antagonistic effects on ER in both HEK 293 and MCF-7 cells. The glyceollins as well as other phytoalexin compounds may represent an important component of the health effects of soy-based foods.


Asunto(s)
Benzopiranos/farmacología , Glycine max/química , Antagonistas de Hormonas/farmacología , Extractos Vegetales/farmacología , Receptores de Estrógenos/fisiología , Benzopiranos/metabolismo , Unión Competitiva , División Celular/efectos de los fármacos , Línea Celular/citología , Línea Celular/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Humanos , Concentración Osmolar , Pterocarpanos , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos
12.
ScientificWorldJournal ; 1: 653-5, 2001 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-12805768

RESUMEN

Endocrine disrupting chemicals (EDCs) include organochlorine pesticides, plastics manufacturing by-products, and certain herbicides. These chemicals have been shown to disrupt hormonal signaling in exposed wildlife, lab animals, and mammalian cell culture by binding to estrogen receptors (ER-+/- and ER-) and affecting the expression of estrogen responsive genes. Additionally, certain plant chemicals, termed phytoestrogens, are also able to bind to estrogen receptors and modulate gene expression, and as such also may be considered EDCs. One example of phytoestrogen action is genistein, a phytochemical produced by soybeans, binding estrogen receptors, and changing expression of estrogen responsive genes which certain studies have linked to a lower incidence of hormonally related cancers in Japanese populations. Why would plants make compounds that are able to act as estrogens in the human body? Obviously, soybeans do not intentionally produce phytoestrogens to prevent breast cancer in Japanese women.


Asunto(s)
Sistema Endocrino/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/genética , Herbicidas/farmacología , Hidrocarburos Clorados , Insecticidas/farmacología , Simbiosis/efectos de los fármacos , Simbiosis/genética , Animales , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos , Isoflavonas/antagonistas & inhibidores , Isoflavonas/fisiología , Fitoestrógenos , Preparaciones de Plantas/antagonistas & inhibidores , Plantas/química , Simbiosis/fisiología , Activación Transcripcional
13.
J Biol Chem ; 275(36): 27694-702, 2000 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-10874044

RESUMEN

The mouse heme oxygenase-1 (HO-1) gene, ho-1, contains two inducible enhancers, E1 and E2. Of several cell lines tested, induction of an E1/luciferase fusion construct, pE1-luc, by CdCl(2) is most pronounced in MCF-7 cells. In these cells, E1, but not E2, is necessary and sufficient for ho-1 gene activation. Exposure of MCF-7 cells to 10 micrometer CdCl(2) stimulates phosphorylation of ERK, JNK, and p38 mitogen-activated protein kinases, implicating one or more of these signaling pathways in ho-1 gene induction. SB203580, an inhibitor of p38, diminishes cadmium-stimulated pE1-luc expression and HO-1 mRNA levels by up to 70-80%. PD098059, an ERK pathway inhibitor, does not affect HO-1 mRNA induction at the highest concentration (40 micrometer) tested. Similarly, co-expression of a dominant-negative mutant of p38alpha, but not of ERK1, ERK2, JNK1, or JNK2, reduces basal and cadmium-induced pE1-luc activity. E1 contains binding sites for the activator protein-1 (Fos/Jun), Cap'n'Collar/basic leucine zipper (CNC-bZIP), and CCAAT/enhancer-binding protein (C/EBP) families of transcription factors. A dominant-negative mutant of Nrf2 (a CNC-bZIP member), but not of c-Jun or C/EBPbeta, inhibits pE1-luc activation by cadmium. Induction of the endogenous ho-1 gene is also inhibited by the Nrf2 mutant. Mutations of E1 that inhibit cadmium inducibility also suppress the trans-activation and DNA binding activities of Nrf2, and SB203580, but not PD098059, attenuates Nrf2-mediated trans-activation of pE1-luc. Taken together, these results indicate that cadmium induces ho-1 gene expression via sequential activation of the p38 kinase pathway and Nrf2.


Asunto(s)
Cadmio/farmacología , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/enzimología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transactivadores/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Secuencia de Bases , Neoplasias de la Mama , Línea Celular , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Femenino , Flavonoides/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Hemo-Oxigenasa 1 , Humanos , Células L , Proteínas de la Membrana , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteína Quinasa 9 Activada por Mitógenos , Datos de Secuencia Molecular , Factor 2 Relacionado con NF-E2 , Factores de Transcripción/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos
14.
Biochem Biophys Res Commun ; 271(2): 342-5, 2000 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-10799299

RESUMEN

We found that in MCF-7 breast carcinoma cells, PI3K and Akt suppressed a dose-dependent induction of apoptosis by tumor necrosis factor alpha (TNF). PI3K and Akt stimulated NF-kappaB activation in a dose-dependent manner, suggesting a common link between these two pathways. TNF has been shown to activate both an apoptotic cascade, as well as a cell survival signal through NF-kappaB. PI3K and AKT cell survival signaling were correlated with increased TNF-stimulated NF-kappaB activity in MCF-7 cells. We demonstrate that while both TNFR1 and NIK are partially involved in Akt-induced NF-kappaB stimulation, a dominant negative IkappaBalpha completely blocked Akt-NF-kappaB cross-talk. PI3K-Akt signaling activated NF-kappaB through both TNFR signaling-dependent and -independent mechanisms, potentially representing a mechanism by which Akt functions to suppress apoptosis in cancer.


Asunto(s)
Apoptosis , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , Factor de Necrosis Tumoral alfa/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Luciferasas/metabolismo , Plásmidos/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Recombinantes/metabolismo , Transducción de Señal , Transfección , Células Tumorales Cultivadas
15.
Int J Oncol ; 16(6): 1179-87, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10811993

RESUMEN

Widespread use of MCF-7 human breast cancer cells as a model system for breast cancer has lead to variations in these cells between different laboratories. Although several reports have addressed these differences in terms of proliferation and estrogenic response, differences in sensitivity to apoptosis have just begun to be described. Based on the possible differences in apoptotic sensitivity that may arise due to the existence of MCF-7 cell variants, we determined the relative sensitivity of MCF-7 cell variants from three established laboratories (designated M, L and N) to known inducers of apoptosis. Consistent with our previous studies we demonstrate that differences exist among these variants in regards to tumor necrosis factor alpha (TNF)-induced cell death and inhibition of proliferation in a dose-dependent manner. To establish if the difference in apoptotic susceptibility was specific to TNF, the three MCF-7 cell variants were tested for their response to other known inducers of apoptosis: okadaic acid, staurosporine and 4-hydroxy-tamoxifen. Viability and DNA fragmentation analysis revealed a similar pattern of resistance to apoptosis by all agents in the MCF-7 M variant. The MCF-7 L variant was resistant to okadaic acid and 4-hydroxy-tamoxifen but not staurosporine. In contrast, MCF-7 N cells were sensitive to induction of apoptosis by all agents. The role of both protein kinase C (PKC) and estrogen signaling in the regulation of cell survival prompted investigation of these pathways as a mechanism for differential sensitivity of MCF-7 cell variants to apoptosis. While both estrogen receptor alpha (ERalpha) and ERbeta were expressed in MCF-7 M and N cells, the absence of ERbeta in MCF-7 L cells correlated with decreased estrogen responsiveness of the L variant. Variations in estrogenic responsiveness and PKC isoform expression may account for the enhanced susceptibility of both the L and N variants to staurosporine.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Estrógenos/metabolismo , Células Tumorales Cultivadas/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias de la Mama/fisiopatología , Estradiol/farmacología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Humanos , Isoformas de Proteínas/farmacología , Receptores de Estrógenos/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/fisiología
16.
J Biol Chem ; 275(13): 9628-35, 2000 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-10734114

RESUMEN

Peptide hormones act to regulate apoptosis through activation of multiple pro- and anti-apoptotic signaling cascades of which lipid signaling events represent an important facet of the cellular rheostat that determines survival and death decisions. Activation of sphingomyelinase, which generates ceramide, is an intermediate in cellular stress responses and induction of apoptosis in many systems. Conversely, phosphatidylinositol 3-kinase (PI3K) is a critical signaling molecule involved in regulating cell survival and proliferation pathways. In the present study, we investigate cross-talk between the PI3K and sphingomyelinase pathways as a mechanism for regulation of cell survival/death decisions. We show that phorbol ester, insulin-like growth factor 1, and a constitutively active PI3K suppress both tumor necrosis factor-induced apoptosis and ceramide generation. Conversely, inhibition of the PI3K pathway with expression of a kinase-dead PI3K both prevented survival signaling and enhanced tumor necrosis factor-induced ceramide generation. The ability of exogenous sphingomyelinase to induce ceramide generation was partially suppressed by expression of constitutively active PI3K and enhanced by inhibition of PI3K suggesting that cross-talk between PI3K and ceramide generation within cells is regulated subsequent to activation of sphingomyelinase.


Asunto(s)
Apoptosis , Supervivencia Celular , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Esfingomielina Fosfodiesterasa/metabolismo , Apoptosis/fisiología , Supervivencia Celular/fisiología , Ceramidas/antagonistas & inhibidores , Ceramidas/fisiología , Activación Enzimática , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Acetato de Tetradecanoilforbol/farmacología , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/fisiología
17.
Nutr Cancer ; 38(2): 229-44, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11525602

RESUMEN

Members of the flavonoid class of phytochemicals have previously been demonstrated to possess estrogenic activity in a number of hormonally responsive systems. We have performed the present study to characterize the estrogenic and antiestrogenic activity of flavonoids in the estrogen receptor (ER)-positive MCF-7 human breast cancer cell line. Using an ER-dependent reporter gene assay and an ER competition binding assay, we have identified phytochemicals possessing estrogenic and antiestrogenic activities, which appeared to correlate directly with their capacity to displace [3H]estradiol from ER. Several flavonoids, including kaempferide, apigenin, and flavone, were distinct, in that their antiestrogenic activity did not appear to correlate with binding to ER, and therefore their suppression of estrogen-mediated gene transactivation and proliferation may occur independent of direct antagonism of the receptor. Further examination in HEK-293 cells transfected with ERalpha or ERbeta demonstrated potent antagonism with kaempferide and apigenin, while flavone was weakly antagonistic only toward ERP. These results suggest that the receptor binding-independent antiestrogenic chemicals may function through alternate signaling pathways as indirect ER modulators in a receptor- and cell type-specific manner. We conclude that antiestrogenic activities of flavonoid phytochemicals may occur through ER binding-dependent and -independent mechanisms and that the binding-independent antiestrogen activity of certain flavonoids is biologically significant in regulation of breast cancer cell proliferation.


Asunto(s)
Neoplasias de la Mama/metabolismo , Estrógenos no Esteroides/farmacología , Flavonoides/farmacología , Isoflavonas , Neoplasias Hormono-Dependientes/metabolismo , Receptores de Estrógenos/metabolismo , Unión Competitiva , Neoplasias de la Mama/prevención & control , División Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Estradiol/metabolismo , Antagonistas de Estrógenos/clasificación , Antagonistas de Estrógenos/metabolismo , Antagonistas de Estrógenos/farmacología , Estrógenos no Esteroides/clasificación , Estrógenos no Esteroides/metabolismo , Femenino , Flavonoides/clasificación , Flavonoides/metabolismo , Humanos , Luciferasas , Neoplasias Hormono-Dependientes/prevención & control , Fitoestrógenos , Preparaciones de Plantas , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/antagonistas & inhibidores , Células Tumorales Cultivadas
18.
Reprod Toxicol ; 13(6): 473-80, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10613395

RESUMEN

Previously, we have demonstrated that cadmium inhibits progesterone release in cultured human trophoblast cells. In the present study, we investigated potential mechanism(s) by which cadmium may elicit this effect. Cytotrophoblasts were obtained via enzymatic dispersion, purified by density gradient centrifugation, and cultured with increasing concentrations of cadmium. Cadmium-induced suppression of progesterone release seemed to be independent of cell death, as no significant decline in viability was observed with cadmium treatment. Further, immunocytochemical localization of cellular boundaries and nuclei indicated approximately 94% syncytial maturity was attained by both untreated and cadmium-treated cells, demonstrating that cadmium did not inhibit syncytial development. However, the abundance of LDL receptor (LDL-R) mRNA transcripts, as determined by competitive RT-PCR, was reduced (P < 0.05) by cadmium exposure in an apparent dose-dependent manner. Thus, the LDL-R, by which cholesterol substrate is supplied to the syncytiotrophoblast, is one site at which cadmium may interfere with placental progesterone production.


Asunto(s)
Cadmio/toxicidad , ARN Mensajero/metabolismo , Receptores de LDL/biosíntesis , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Fragmentación del ADN/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Embarazo , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de LDL/genética , Sales de Tetrazolio , Tiazoles , Trofoblastos/citología
19.
Carcinogenesis ; 20(11): 2057-61, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10545406

RESUMEN

Environmental estrogens represent a class of compounds which have been shown to mimic the effects or activity of the naturally occurring ovarian hormone 17beta-estradiol. Given the role of 17beta-estradiol in cell survival in a number of systems, we wished to determine if environmental estrogens protect MCF-7 cells from apoptosis. Here we demonstrate that the organochlorine pesticides o, p' DDT and alachlor, like 17beta-estradiol, have the ability to suppress tumor necrosis factor alpha (TNF)-induced apoptosis in estrogen receptor (ER)-positive MCF-7 breast carcinoma cells. These compounds, however, did not affect TNF-induced apoptosis of the ER-negative MDA-MB-231 cell line. The ability of these compounds to suppress apoptosis in MCF-7 cells was correlated with an ER-dependent increase in Bcl-2 expression. Taken together these results demonstrate that estrogenic organochlorine pesticides like o, p' DDT and alachlor may partially mimic the primary endogenous estrogen, 17beta-estradiol, and function to suppress apoptosis in ER-responsive cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Estrógenos no Esteroides/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Neoplasias de la Mama/metabolismo , Humanos , Receptores de Estrógenos/metabolismo , Células Tumorales Cultivadas , Xenobióticos/farmacología
20.
Cancer Res ; 58(21): 4940-6, 1998 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9810003

RESUMEN

Widespread use of MCF-7 human breast carcinoma cells as a model system for breast cancer has led to variations in these cells between different laboratories. Although several reports have addressed these differences in terms of proliferation and estrogenic response, variations in sensitivity to apoptosis have not yet been described. Tumor necrosis factor alpha (TNF-alpha) has been shown to both induce apoptosis and inhibit proliferation in MCF-7 cells. We observed that TNF-alpha inhibited proliferation in MCF-7 cell variants from three different laboratories (designated M, L, and N). MCF-7 M cells were resistant to TNF-alpha-induced apoptosis, whereas MCF-7 L cells were moderately resistant to the effect of TNF-alpha. A third variant, MCF-7 N, underwent apoptosis when exposed to TNF-alpha. Analysis of the p55 TNF-alpha receptor (TNFR) 1 expression revealed the greatest expression in MCF-7 N cells, whereas the MCF-7 L and M cells expressed 89 and 67% of MCF-7 N cell TNFR1 levels, respectively. Ceramide generation occurred in all three variants in response to TNF-alpha treatment, with MCF-7 N cells expressing the greatest increase. Cleavage of the CPP32/caspase 3 substrate poly(ADP-ribose) was observed in MCF-7 N and L cells as early as 3 and 6 h, respectively, but poly(ADP-ribose) cleavage was not observed in MCF-7 M cells. The delayed protease activation in the L variant may represent the mechanism by which these cells display delayed sensitivity to TNF-a-induced apoptosis. Expression of the Bcl-2, Mcl-1, Bcl-X, Bax, and Bak proteins was analyzed to determine whether the differences in MCF-7 cell sensitivity to apoptosis could be correlated to the differential expression of these proteins. Whereas Bak, Bcl-X, and Mcl-1 levels were identical between variants, the levels of Bcl-2 were 3.5-3.8-fold higher and the levels of Bax were 1.5-1.7-fold lower in the resistant variants (M and L) as compared with those of the sensitive variant (N). Taken together, these results suggest that differences in susceptibility to TNF-alpha-induced apoptosis among MCF-7 breast cancer cell variants may be explained by differences in TNFR expression, ceramide generation, differential expression of the Bcl-2 family of proteins, and protease activation.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Factor de Necrosis Tumoral alfa/farmacología , Neoplasias de la Mama/metabolismo , Caspasas/fisiología , Ceramidas/biosíntesis , Femenino , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Receptores del Factor de Necrosis Tumoral/análisis , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA