Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Mol Carcinog ; 53(1): 38-48, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22911661

RESUMEN

microRNAs (miRNA) are regulators of cellular pathways and alterations of normal miRNA expression levels have been shown to increase tumorigenesis. miR-24 has been demonstrated as having both tumor suppressive and oncogenic properties depending on cell context. Here, we demonstrate a possible role for pre-miR-24-2 as a tumor suppressor in the MCF-7 breast cancer cell line through the preferential processing of mature miR-24-2* over miR-24. Specifically, we show that the ectopic expression of miR-24-2* in MCF-7 breast cancer cells results in a suppression of cellular survival both in vivo and in vitro. Notably, the overexpression of miR-24-2* results in a dampening of cell survival through the targeted suppression of PKCα. In addition, a similar biological change is observed in vivo where MCF-7 cells overexpressing pre-miR-24-2 have decreased tumorigenicity and tumor incidence. Taken together our data demonstrate that when overexpressed biogenesis of the pre-miR-24-2 favors miR-24-2* in the MCF-7 breast cancer cell line and suggests a tumor suppressive role for miR-24-2* observed through the inhibition of PKCα-mediated cellular survival.


Asunto(s)
Neoplasias de la Mama/genética , MicroARNs/genética , Proteína Quinasa C-alfa/genética , Animales , Emparejamiento Base , Secuencia de Bases , Sitios de Unión , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Isoenzimas , Células MCF-7 , Ratones , MicroARNs/química , MicroARNs/metabolismo , Proteína Quinasa C-alfa/química , Proteína Quinasa C-alfa/metabolismo , Interferencia de ARN
2.
Bioorg Med Chem ; 22(4): 1412-20, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24457089

RESUMEN

Multidrug-resistance is a major cause of cancer chemotherapy failure in clinical treatment. Evidence shows that multidrug-resistant cancer cells are as sensitive as corresponding regular cancer cells under the exposure to anticancer ceramide analogs. In this work we designed five new ceramide analogs with different backbones, in order to test the hypothesis that extending the conjugated system in ceramide analogs would lead to an increase of their anticancer activity and selectivity towards resistant cancer cells. The analogs with the 3-ketone-4,6-diene backbone show the highest apoptosis-inducing efficacy. The most potent compound, analog 406, possesses higher pro-apoptotic activity in chemo-resistant cell lines MCF-7TN-R and NCI/ADR-RES than the corresponding chemo-sensitive cell lines MCF-7 and OVCAR-8, respectively. However, this compound shows the same potency in inhibiting the growth of another pair of chemo-sensitive and chemo-resistant cancer cells, MCF-7 and MCF-7/Dox. Mechanism investigations indicate that analog 406 can induce apoptosis in chemo-resistant cancer cells through the mitochondrial pathway. Cellular glucosylceramide synthase assay shows that analog 406 does not interrupt glucosylceramide synthase in chemo-resistant cancer cell NCI/ADR-RES. These findings suggest that due to certain intrinsic properties, ceramide analogs' pro-apoptotic activity is not disrupted by the normal drug-resistance mechanisms, leading to their potential use for overcoming cancer multidrug-resistance.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Bencenoacetamidas/química , Ceramidas/química , Ceramidas/farmacología , Cetonas/química , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Bencenoacetamidas/síntesis química , Bencenoacetamidas/farmacología , Línea Celular Tumoral , Ceramidas/síntesis química , Resistencia a Antineoplásicos/efectos de los fármacos , Glucosiltransferasas/antagonistas & inhibidores , Glucosiltransferasas/metabolismo , Humanos , Isomerismo , Células MCF-7 , Conformación Molecular
3.
Biochim Biophys Acta ; 1825(1): 37-48, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22020294

RESUMEN

While conventional MAP kinase pathways are one of the most highly studied signal transduction molecules, less is known about the MEK5 signaling pathway. This pathway has been shown to play a role in normal cell growth cycles, survival and differentiation. The MEK5 pathway is also believed to mediate the effects of a number of oncogenes. MEK5 is the upstream activator of ERK5 in many epithelial cells. Activation of the MEK-MAPK pathway is a frequent event in malignant tumor formation and contributes to chemoresistance and anti-apoptotic signaling. This pathway may be involved in a number of more aggressive, metastatic varieties of cancer due to its role in cell survival, proliferation and EMT transitioning. Further study of this pathway may lead to new prognostic factors and new drug targets to combat more aggressive forms of cancer.


Asunto(s)
MAP Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Neoplasias/metabolismo , Diferenciación Celular , Supervivencia Celular , Transición Epitelial-Mesenquimal , Humanos , MAP Quinasa Quinasa 5/antagonistas & inhibidores , Proteína Quinasa 7 Activada por Mitógenos/antagonistas & inhibidores
4.
Cancer Invest ; 30(2): 135-48, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22250588

RESUMEN

With an estimated 207,090 patients diagnosed with breast cancer in 2010, the role of chemotherapy-induced cognitive impairment is of growing importance. Studies to determine the impact of chemotherapy-induced cognitive impairment have been hindered by difficulties in study-design, in particular, study methodology. Here, we present a review of existing studies and discuss several mechanisms for chemotherapy-induced neurocognitive impairment in breast cancer patients, such as direct neurotoxic injury, telomere shortening, oxidative stress, cytokine dysregulation, estrogen-mediated effects, and the role of certain genetic polymorphisms. Decreased estrogen levels may serve as a link between multiple mechanisms potentiating the effects of the chemotherapy-induced cognitive impairment.


Asunto(s)
Antineoplásicos/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Trastornos del Conocimiento/inducido químicamente , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/psicología , Trastornos del Conocimiento/psicología , Femenino , Humanos , Pruebas Neuropsicológicas , Estudios Prospectivos
5.
Bioorg Med Chem Lett ; 22(7): 2624-8, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22366655

RESUMEN

The ceramide-sphingosine-1-phosphate rheostat is a promising therapeutic target. Here, the novel ceramide analog (S)-2-(benzylideneamino)-3-hydroxy-N-tetrade-cylpropanamide is shown to block proliferation and enhance the efficacy of the clinical chemotherapeutics, etoposide and doxorubicin. These results demonstrate the therapeutic potential of this compound in treating both endocrine resistant and chemoresistant breast cancer.


Asunto(s)
Antineoplásicos/síntesis química , Ceramidas/síntesis química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ceramidas/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Etopósido/farmacología , Femenino , Humanos , Antígeno Ki-67/biosíntesis , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Estereoisomerismo , Relación Estructura-Actividad
6.
Mol Cancer ; 9: 295, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21087507

RESUMEN

BACKGROUND: Adult human mesenchymal stem cells (hMSC) have been shown to home to sites of carcinoma and affect biological processes, including tumour growth and metastasis. Previous findings have been conflicting and a clear understanding of the effects of hMSCs on cancer remains to be established. Therefore, we set out to investigate the impact of hMSCs on the oestrogen receptor positive, hormone-dependent breast carcinoma cell line MCF-7. RESULTS: In this study, we show the effects of hMSCs on cancer cells are mediated through a secreted factor(s) which are enhanced by cancer cell-hMSC contact/communication. In addition to enhanced proliferation when in co-culture with hMSCs, MCF-7 cells were found to have increased migration potential in vitro. Inhibition of ER signalling by the pure anti-oestrogen ICI 182,780 decreased the effect of hMSCs on MCF-7 cell proliferation and migration supporting a role for ER signalling in the hMSC/MCF-7 cell interaction. Additionally, hMSCs have been shown to secrete a wide variety of growth factors and chemokines including stromal cell-derived factor-1 (SDF-1). This coupled with the knowledge that SDF-1 is an ER-mediated gene linked with hormone-independence and metastasis led to the investigation of the SDF-1/CXCR4 signalling axis in hMSC-MCF-7 cell interaction. Experiments revealed an increase in SDF-1 gene expression both in vivo and in vitro when MCF-7 cells were cultured with hMSCs. SDF-1 treatment of MCF-7 cells alone increased proliferation to just below that seen with hMSC co-culture. Additionally, blocking SDF-1 signalling using a CXCR4-specific inhibitor decreased hMSC induced proliferation and migration of MCF-7. However, the combined treatment of ICI and AMD3100 reduced MCF-7 cell proliferation and migration below control levels, indicating targeting both the ER and CXCR4 pathways is effective in decreasing the hMSCs induction of MCF-7 cell proliferation and migration. CONCLUSIONS: The sum of these data reveals the relationship between tumour microenvironment and tumour growth and progression. Better understanding of the mechanisms involved in this tumour stroma cell interaction may provide novel targets for the development of treatment strategies for oestrogen receptor positive, hormone-independent, and endocrine-resistant breast carcinoma.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Quimiocina CXCL12/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptores CXCR4/metabolismo , Receptores de Estrógenos/metabolismo , Bencilaminas , Neoplasias de la Mama/patología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/farmacología , Técnicas de Cocultivo , Ciclamas , Femenino , Citometría de Flujo , Compuestos Heterocíclicos/farmacología , Humanos , Microscopía Fluorescente , Receptores CXCR4/antagonistas & inhibidores , Receptores de Estrógenos/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
7.
Breast Cancer Res Treat ; 121(2): 293-300, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19597705

RESUMEN

Adult human mesenchymal stem cells (hMSCs) have been shown to home to sites of breast cancer and integrate into the tumor stroma. We demonstrate here the effect of hMSCs on primary breast tumor growth and the progression of these tumors to hormone independence. Co-injection of bone marrow-derived hMSCs enhances primary tumor growth of the estrogen receptor-positive, hormone-dependent breast carcinoma cell line MCF-7 in the presence or absence of estrogen in SCID/beige mice. We also show hormone-independent growth of MCF-7 cells when co-injected with hMSCs. These effects were found in conjunction with increased immunohistochemical staining of the progesterone receptor in the MCF-7/hMSC tumors as compared to MCF-7 control tumors. This increase in PgR expression indicates a link between MCF-7 cells and MSCs through ER-mediated signaling. Taken together, our data reveal the relationship between tumor microenvironment and tumor growth and the progression to hormone independence. This tumor stroma-cell interaction may provide a novel target for the treatment of estrogen receptor-positive, hormone-independent, and endocrine-resistant breast carcinoma.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Células Madre Mesenquimatosas/patología , Animales , Antineoplásicos Hormonales/farmacología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones SCID , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Pharmacol Exp Ther ; 332(1): 35-45, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19797619

RESUMEN

Glyceollins, a group of novel phytoalexins isolated from activated soy, have recently been demonstrated to be novel antiestrogens that bind to the estrogen receptor (ER) and inhibit estrogen-induced tumor progression. Our previous publications have focused specifically on inhibition of tumor formation and growth by the glyceollin mixture, which contains three glyceollin isomers (I, II, and III). Here, we show the glyceollin mixture is also effective as a potential antiestrogenic, therapeutic agent that prevents estrogen-stimulated tumorigenesis and displays a differential pattern of gene expression from tamoxifen. By isolating the individual glyceollin isomers (I, II, and III), we have identified the active antiestrogenic component by using competition binding assays with human ERalpha and in an estrogen-responsive element-based luciferase reporter assay. We identified glyceollin I as the active component of the combined glyceollin mixture. Ligand-receptor modeling (docking) of glyceollin I, II, and III within the ERalpha ligand binding cavity demonstrates a unique type II antiestrogenic confirmation adopted by glyceollin I but not isomers II and III. We further compared the effects of glyceollin I to the antiestrogens, 4-hydroxytamoxifen and ICI 182,780 (fulvestrant), in MCF-7 breast cancer cells and BG-1 ovarian cancer cells on 17beta-estradiol-stimulated expression of progesterone receptor and stromal derived factor-1alpha. Our results establish a novel inhibition of ER-mediated gene expression and cell proliferation/survival. Glyceollin I may represent an important component of a phytoalexin-enriched food (activated) diet in terms of chemoprevention as well as a novel therapeutic agent for hormone-dependent tumors.


Asunto(s)
Anticarcinógenos/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Glycine max/química , Pterocarpanos/farmacología , Terpenos/farmacología , Animales , Anticarcinógenos/química , Anticarcinógenos/aislamiento & purificación , Anticarcinógenos/uso terapéutico , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Moduladores de los Receptores de Estrógeno/química , Moduladores de los Receptores de Estrógeno/aislamiento & purificación , Moduladores de los Receptores de Estrógeno/uso terapéutico , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/biosíntesis , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Ratones , Ratones Desnudos , Estructura Molecular , Trasplante de Neoplasias , Pterocarpanos/química , Pterocarpanos/aislamiento & purificación , Pterocarpanos/uso terapéutico , Sesquiterpenos , Estereoisomerismo , Tamoxifeno/farmacología , Terpenos/química , Terpenos/aislamiento & purificación , Terpenos/uso terapéutico , Transcripción Genética/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Fitoalexinas
9.
Bioorg Med Chem ; 18(14): 5316-22, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20639111

RESUMEN

A group of novel L-serinamides, substituted (S)-2-(benzylideneamino)-3-hydroxy-N-tetradecylpropanamides (3a-o) and substituted (S)-2-(benzylamino)-3-hydroxy-N-tetradecyl propanamides (4c, 4i, 4l, and 4o), were synthesized as potential anti-tumor lead compounds. In vitro cell viability assay results indicate treatment with 3a-o compounds resulted in significant inhibition of cell viability in the chemoresistant breast cancer cell line, MCF-7TN-R. Compounds 3i and 3l, both ortho-substituted analogs, show the greatest efficacy with IC50 values of 10.3 microM and 12.5 microM, respectively. The SAR analysis indicate that the imine functional group of 3a-o is critical for the cellular anti-viability effect, and the partial atomic charge (PAC) value of imine C atom is a valuable structural parameter for predicting the activity of these ceramide analogs.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Ceramidas/química , Ceramidas/farmacología , Antineoplásicos/síntesis química , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ceramidas/síntesis química , Resistencia a Antineoplásicos , Femenino , Humanos , Concentración 50 Inhibidora , Relación Estructura-Actividad
10.
J Cell Biol ; 167(3): 469-78, 2004 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-15534001

RESUMEN

In the lactating breast, ERBB4 localizes to the nuclei of secretory epithelium while regulating activities of the signal transducer and activator of transcription (STAT) 5A transcription factor essential for milk-gene expression. We have identified an intrinsic ERBB4 NLS (residues 676-684) within the ERBB4 intracellular domain (4ICD) that is essential for nuclear accumulation of 4ICD. To determine the functional significance of 4ICD nuclear translocation in a physiologically relevant system, we have demonstrated that cotransfection of ERBB4 and STAT5A in a human breast cancer cell line stimulates beta-casein promoter activity. Significantly, nuclear localization of STAT5A and subsequent stimulation of the beta-casein promoter requires nuclear translocation of 4ICD. Moreover, 4ICD and STAT5A colocalize within nuclei of heregulin beta 1 (HRG)-stimulated cells and both proteins bind to the endogenous beta-casein promoter in T47D breast cancer cells. Together, our results establish a novel molecular mechanism of transmembrane receptor signal transduction involving nuclear cotranslocation of the receptor intracellular domain and associated transcription factor. Subsequent binding of the two proteins at transcription factor target promoters results in activation of gene expression.


Asunto(s)
Transporte Activo de Núcleo Celular , Proteínas de Unión al ADN/metabolismo , Receptores ErbB/fisiología , Regulación de la Expresión Génica , Proteínas de la Leche/metabolismo , Chaperonas Moleculares/fisiología , Transactivadores/metabolismo , Secuencia de Aminoácidos , Neoplasias de la Mama/patología , Caseínas/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Humanos , Proteínas de la Leche/genética , Neurregulina-1/metabolismo , Señales de Localización Nuclear , Regiones Promotoras Genéticas , Unión Proteica , Receptor ErbB-4 , Factor de Transcripción STAT5 , Transactivadores/genética , Transfección , Proteínas Supresoras de Tumor
11.
Artículo en Inglés | MEDLINE | ID: mdl-19439190

RESUMEN

Previous studies have suggested that the lactate dehydrogenase-B gene (Ldh-B) of the Atlantic killifish, Fundulus heteroclitus, is a hypoxia-responsive gene. Here, we demonstrate that the F. heteroclitus Ldh-B promoter confers hypoxia-dependence upon reporter gene expression in transiently transfected mammalian (Hep3B) and fish (RTG-2 and RTH-149) cells in culture. Mutation and deletion analyses identified a putative hypoxia-response element (HRE) between 109 and 90 nucleotides upstream of the major start site. This HRE is characterized by the sequence 5'-GATGTG-3' spaced by 8 nucleotides from a perfect inverted repeat, and both sites are necessary for hypoxic induction of reporter gene expression in mammalian and fish cells. This HRE differs from the canonical sequence at one nucleotide position that is invariant among HREs from a wide range of hypoxia-sensitive genes. In fish cells, maximal induction of reporter gene expression driven by this HRE occurred at the lowest oxygen level tested (0.5%), took 48 h to 96 h, and was independent of glucose concentration (between 5.6 and 25 mM). Under all conditions tested, hypoxic induction of gene expression was lower in RTH-149 cells than in RTG-2, suggesting a potential defect in hypoxia signaling in RTH-149 cells. These results demonstrate that the F. heteroclitus Ldh-B promoter contains a novel HRE that is capable of driving reporter gene expression in a sequence-specific and oxygen-, time-, and cell line-dependent manner.


Asunto(s)
Fundulidae/genética , L-Lactato Deshidrogenasa/genética , Elementos de Respuesta/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Genes Reporteros/efectos de los fármacos , Glucosa/farmacología , Humanos , Hipoxia/genética , Isoenzimas/genética , Luciferasas/biosíntesis , Elementos de Respuesta/efectos de los fármacos
12.
Breast Cancer Res ; 10(6): R105, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19087274

RESUMEN

INTRODUCTION: Despite intensive study of the mechanisms of chemotherapeutic drug resistance in human breast cancer, few reports have systematically investigated the mechanisms that underlie resistance to the chemotherapy-sensitizing agent tumor necrosis factor (TNF)-alpha. Additionally, the relationship between TNF-alpha resistance mediated by MEK5/Erk5 signaling and epithelial-mesenchymal transition (EMT), a process associated with promotion of invasion, metastasis, and recurrence in breast cancer, has not previously been investigated. METHODS: To compare differences in the proteome of the TNF-alpha resistant MCF-7 breast cancer cell line MCF-7-MEK5 (in which TNF-alpha resistance is mediated by MEK5/Erk5 signaling) and its parental TNF-a sensitive MCF-7 cell line MCF-7-VEC, two-dimensional gel electrophoresis and high performance capillary liquid chromatography coupled with tandem mass spectrometry approaches were used. Differential protein expression was verified at the transcriptional level using RT-PCR assays. An EMT phenotype was confirmed using immunofluorescence staining and gene expression analyses. A short hairpin RNA strategy targeting Erk5 was utilized to investigate the requirement for the MEK/Erk5 pathway in EMT. RESULTS: Proteomic analyses and PCR assays were used to identify and confirm differential expression of proteins. In MCF-7-MEK5 versus MCF-7-VEC cells, vimentin (VIM), glutathione-S-transferase P (GSTP1), and creatine kinase B-type (CKB) were upregulated, and keratin 8 (KRT8), keratin 19 (KRT19) and glutathione-S-transferase Mu 3 (GSTM3) were downregulated. Morphology and immunofluorescence staining for E-cadherin and vimentin revealed an EMT phenotype in the MCF-7-MEK5 cells. Furthermore, EMT regulatory genes SNAI2 (slug), ZEB1 (delta-EF1), and N-cadherin (CDH2) were upregulated, whereas E-cadherin (CDH1) was downregulated in MCF-7-MEK5 cells versus MCF-7-VEC cells. RNA interference targeting of Erk5 reversed MEK5-mediated EMT gene expression. CONCLUSIONS: This study demonstrates that MEK5 over-expression promotes a TNF-alpha resistance phenotype associated with distinct proteomic changes (upregulation of VIM/vim, GSTP1/gstp1, and CKB/ckb; and downregulation of KRT8/krt8, KRT19/krt19, and GSTM3/gstm3). We further demonstrate that MEK5-mediated progression to an EMT phenotype is dependent upon intact Erk5 and associated with upregulation of SNAI2 and ZEB1 expression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Epitelio/patología , MAP Quinasa Quinasa 5/metabolismo , Mesodermo/patología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Proteómica , Factor de Necrosis Tumoral alfa/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Cromatografía Liquida , Electroforesis en Gel Bidimensional , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , MAP Quinasa Quinasa 5/genética , Proteína Quinasa 7 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 7 Activada por Mitógenos/genética , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masas en Tándem , Ensayo de Tumor de Célula Madre
13.
Int J Oncol ; 31(4): 705-11, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17786300

RESUMEN

Recent investigations have demonstrated that polyphenolic catechins inhibit cancer cell proliferation and tumor growth. However, how the major active component of tea catechins, epigallocatechin-3 gallate (EGCG), mediates anticancerous effects has not been extensively examined. We have investigated the cell growth inhibitory effects of EGCG on cell growth of the human breast cancer cell line MCF-7, and the mechanism of its action with emphasis on the regulation of tumor cell survival. A significant EGCG dose-dependent growth inhibition was observed coordinated with EGCG-induced apoptosis. Analysis of survivin expression after addition of EGCG showed that both survivin mRNA and protein were decreased. The survivin-promoter luciferase activity in EGCG-treated cells was significantly inhibited by 91+/-2.0% (P<0.001), compared with the control. Interestingly, EGCG strongly inhibited the basal activation of phospho-AKT and AKT kinase activity as early as 30 min after treatment. Furthermore, inhibition of AKT kinase activity by EGCG preceded the suppression of survivin (1 h post treatment), followed by increased caspase-9 activity (6 h post treatment). A dominant negative AKT or the phosphatidylinositol 3-kinase inhibitor, LY294002, also strongly inhibited survivin promoter activity, providing further evidence to support the hypothesis that the inhibitory effect of EGCG on survivin is mediated via the AKT pathway. Therefore, EGCG is a potent proapoptotic agent in MCF-7 breast cancer cells that targets survivin expression via suppression of the AKT pathway.


Asunto(s)
Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Catequina/análogos & derivados , Regulación Neoplásica de la Expresión Génica , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Western Blotting , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Catequina/farmacología , Línea Celular Tumoral , Ensayo de Unidades Formadoras de Colonias , Humanos , Proteínas Inhibidoras de la Apoptosis , Luciferasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Inhibidores de Proteasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Survivin
14.
Int J Oncol ; 30(1): 113-22, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17143519

RESUMEN

Based on cDNA microarray results, integrin-linked kinase (ILK) emerged as an interesting candidate in hypoxia-mediated survival mechanisms employed by cancer cells. This notion was confirmed here by the following observations: the 5' promoter region of the ilk gene contains hypoxia responsive elements (HRE) that bind hypoxia-inducible factor (HIF) transcription factor complexes and drive HRE-luciferase gene expression in reporter assays; ILK protein and kinase activity are induced following hypoxia; downstream targets of ILK signaling are induced following hypoxia treatment; inhibition of ILK leads to increased apoptosis; and HIF and ILK are co-localized within human cancer tissues. The identification of ILK as a player in hypoxia survival signaling employed by cancer cells further validates ILK as a unique target for cancer therapy.


Asunto(s)
Hipoxia/enzimología , Proteínas Serina-Treonina Quinasas/genética , Apoptosis , Neoplasias de la Mama/enzimología , Carcinoma Hepatocelular , Línea Celular Tumoral , ADN de Neoplasias/genética , Femenino , Genes Reporteros , Humanos , Inmunohistoquímica , Neoplasias Renales/enzimología , Neoplasias Hepáticas , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/enzimología , Proteínas Serina-Treonina Quinasas/biosíntesis , Transfección
15.
Clin Cancer Res ; 12(23): 7159-64, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17145841

RESUMEN

PURPOSE: We have identified the phytoalexin compounds glyceollins I, II, and III, which exhibit marked antiestrogenic effects on estrogen receptor function and estrogen-dependent tumor growth in vivo. The purpose of this study was to investigate the interactions among the induced soy phytoalexins glyceollins I, II, and III on the growth of estrogen-dependent MCF-7 breast cancer and BG-1 ovarian cancer cells implanted in ovariectomized athymic mice. EXPERIMENTAL DESIGN: Four treatment groups for each cell line were used: vehicle control, 20 mg/kg/mouse/d glyceollin mixture injection, 0.72 mg estradiol (E2) implant, and E2 implant + 20 mg/kg/mouse/d glyceollin injection. RESULTS: Treatment with glyceollin suppressed E2-stimulated tumor growth of MCF-7 cells (-53.4%) and BG-1 cells (-73.1%) in ovariectomized athymic mice. These tumor-inhibiting effects corresponded with significantly lower E2-induced progesterone receptor expression in the tumors. In contrast to tamoxifen, the glyceollins had no estrogen-agonist effects on uterine morphology and partially antagonized the uterotropic effects of estrogen. CONCLUSIONS: These findings identify glyceollins as antiestrogenic agents that may be useful in the prevention or treatment of breast and ovarian carcinoma.


Asunto(s)
Benzopiranos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Moduladores de los Receptores de Estrógeno/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Animales , Benzopiranos/química , Benzopiranos/farmacología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Estradiol/administración & dosificación , Moduladores de los Receptores de Estrógeno/química , Moduladores de los Receptores de Estrógeno/farmacología , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/patología , Valor Predictivo de las Pruebas , Pterocarpanos , Receptores de Progesterona/biosíntesis , Receptores de Progesterona/efectos de los fármacos , Relación Estructura-Actividad , Trasplante Heterólogo , Útero/efectos de los fármacos , Útero/patología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Lett ; 392: 51-59, 2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28153789

RESUMEN

Mitogen-activated protein kinases (MAPKs) regulate diverse cellular processes including proliferation, cell survival, differentiation, and apoptosis. While conventional MAPK constituents have well-defined roles in oncogenesis, the MEK5 pathway has only recently emerged in cancer research. In this review, we consider the MEK5 signaling cascade, focusing specifically on its involvement in drug resistance and regulation of aggressive cancer phenotypes. Moreover, we explore the role of MEK5/ERK5 in tumorigenesis and metastatic progression, discussing the discrepancies in preclinical studies and assessing its viability as a therapeutic target for anti-cancer agents.


Asunto(s)
MAP Quinasa Quinasa 5/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Diferenciación Celular , Proliferación Celular , Resistencia a Antineoplásicos , Humanos , MAP Quinasa Quinasa 5/antagonistas & inhibidores , MAP Quinasa Quinasa 5/genética , Terapia Molecular Dirigida , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/efectos de los fármacos
17.
Int J Oncol ; 26(3): 763-8, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15703835

RESUMEN

The ability of peptide hormones, as well as the protein kinase C (PKC)-activating phorbol ester (PMA), to protect cells from apoptosis has been demonstrated to occur through activation of cellular signaling pathways such as the mitogen-activated protein kinase (MAPK) and phosphatidyl-inositol-3 kinase (PI3K) families. Here we demonstrate that tumor necrosis factor alpha (TNF)-induced apoptosis is suppressed by treatment with PMA in MCF-7 breast carcinoma cells. Reversal of the PMA survival effect with the classical isoform-specific PKC inhibitor, Go 6976, or the selective mitogen-activated protein kinase kinase (MEK) inhibitor, PD 098059, suggested a partial requirement for PKCalpha and the Erk cascade in MCF-7 cell survival. The ability of these agents to block PMA-mediated cell survival was also correlated with a suppression of PMA-induced AP-1 activity. Some naturally occurring flavonoid compounds such as apigenin can function to block cell signaling cascades such as MAPK. The ability of apigenin to block PMA-mediated cell survival was similarly correlated with suppression of PMA-stimulated AP-1 activity. Our results strongly suggest that PKC- and Erk-dependent pathways are critical components of the cell survival cascade function in suppression of TNF-induced apoptosis in MCF-7 cells. The ability of natural dietary flavonoids such as apigenin to affect cell survival pathways may represent an important aspect of the proposed anti-tumor effects of these compounds.


Asunto(s)
Apoptosis , Neoplasias de la Mama/patología , Supervivencia Celular , MAP Quinasa Quinasa 1/farmacología , Proteína Quinasa C/farmacología , Apigenina/farmacología , Dieta , Femenino , Flavonas/farmacología , Flavonoides/farmacología , Humanos , Ésteres del Forbol/farmacología , Transducción de Señal , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología
18.
Toxicol Lett ; 155(2): 227-38, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15603917

RESUMEN

Some xenobiotics, such as dichlorodiphenyltrichloroethane (DDT), bind to and activate estrogen receptors (ERs), eliciting estrogenic effects in both wildlife and humans. However, our laboratory and others have demonstrated that DDT and DDT-like compounds target non-ER pathways. In search for a molecular mechanism we recently established that DDT and its metabolites stimulate activator protein-1 (AP-1)-mediated gene expression through the p38 mitogen-activated protein kinase (MAPK) cascade. Here, we determined that DDT-induced p38 activity produces a novel environmental signaling pathway in endometrial Ishikawa and human embryonic kidney (HEK) 293 cells. Xenobiotic exposure stimulates expression of the death ligand, tumor necrosis factor-alpha (TNF-alpha) as demonstrated using RT-PCR and reporter gene assays. Furthermore, DDT-induced p38 activity led to the release of cytochrome c from the mitochondria and activation of caspase-3/7. Ultimately, DDT-treated cells underwent cell death. Taken together, these data demonstrate DDT induces both the expression of the death ligand TNF-alpha and apoptosis through a p38 MAPK-dependent mechanism.


Asunto(s)
Apoptosis/efectos de los fármacos , DDT/toxicidad , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Xenobióticos/toxicidad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Caspasa 3 , Caspasa 7 , Caspasas/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Citocromos c/metabolismo , Humanos , Isomerismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Exp Hematol ; 30(12): 1419-27, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12482504

RESUMEN

OBJECTIVE: The aim of this study was to further define the signal transduction pathways leading to hypoxia-inducible factor-1 (HIF-1) erythropoietin (EPO) gene expression. MATERIALS AND METHODS: Human hepatocellular carcinoma cells (Hep3B) were exposed to hypoxia (1% oxygen) and examined for mRNA expression, as well as gene transactivation with RT-PCR and luciferase reporter gene assays, respectively. RESULTS: Treatment with LY294002 (a selective pharmacological inhibitor of phosphatidylinositol 3-kinase) significantly inhibited EPO protein and mRNA expression in Hep3B cells exposed to hypoxia for 24 hours, while treatment with PD098059 or SB203580 (selective pharmacological inhibitors of the MEK and p38 mitogen-activated protein kinase pathways, respectively) had no significant effects. The activity of AKT, a downstream target of PI3K, was increased by hypoxia and was also inhibited by LY294002. Genetic inhibition of AKT resulted in significant inhibition of NF-kappaB and HIF-1-mediated transactivation, as well as EPO gene expression, in response to hypoxia. Overexpression of constitutively active AKT resulted in increased NF-kappaB and HIF-1 transactivation. The selective inhibitor of NF-kappaB, pyrrolidine dithiocarbamate (PDTC), significantly blocked HIF-1 protein expression. Inhibition of NF-kappaB with a superrepressor dominant negative IkappaBalpha genetic construct also significantly blocked NF-kappaB and HIF-1 transactivation, as well as EPO gene expression. CONCLUSION: We propose a key role for NF-kappaB in EPO gene regulation in response to hypoxia.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Eritropoyetina/genética , FN-kappa B/fisiología , Proteínas Nucleares/fisiología , Proteínas Serina-Treonina Quinasas , Factores de Transcripción , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Hipoxia/metabolismo , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , FN-kappa B/metabolismo , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Activación Transcripcional , Células Tumorales Cultivadas
20.
Int J Oncol ; 24(6): 1473-80, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15138590

RESUMEN

The mitogen-activated protein kinase (MAPK) cascade is a critical component in the regulation of cell survival and proliferation decisions. In breast carcinoma cells, activation of the p38-MAPK member of this family occurs in response to pro-inflammatory cytokines and cellular stress. The involvement of p38-MAPK in the activation of the transcription factor, NF-kappaB, suggests a potential role and mechanism for regulation of cell survival and drug resistance. Generation of the resistant MCF-7 variant (MCF-7TN-R) was achieved by prolonged exposure of MCF-7N cells to increasing concentrations of TNF. Differences in MAPK activation and function in the MCF-7 cell variants were determined. The role of the p38-MAPK pathway in regulation of resistance was determined using pharmacological (SB 203580) or molecular [Dominant Inhibitory (DI)-p38] inhibition. The effect of p38 inhibition on NF-kappaB transcriptional activation was analyzed. As compared to the sensitive MCF-7N parent cell line, the MCF-7TN-R cell line displayed significant resistance to TNF- and TRAIL-induced cell death. Analysis of the expression and phosphorylation of members of the MAPK family revealed an increased basal activation of p38 in the MCF-7TN-R variant. The p38-mediated phosphorylation and transcriptional activity were suppressed by pharmacologic inhibition with SB 230580. Treatment of MCF-7TN-R cells with SB partially restored sensitivity to TNF-induced cell death. In addition, use of a DI-p38 construct with or without the addition to TNF induced cell death, thus restoring TNF-sensitivity to these cells. The ability of p38 inhibition to restore apoptotic sensitivity was correlated with suppression of the TNF-induced cell survival pathway, NF-kappaB. The increased activation of p38-MAPK in MCF-7TN-R cells demonstrates that this signaling pathway through activation of NF-kappaB is an important route for control of resistance to cell death in breast carcinoma. Molecular and pharmacological inhibition of p38-MAPK signaling may represent a mechanism for sensitizing cancer cells to chemotherapeutic regimens and restoration of apoptotic signaling.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Glicoproteínas de Membrana/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis , Neoplasias de la Mama/patología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Genes Dominantes , Humanos , Luciferasas/metabolismo , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF , Transcripción Genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA