Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Cell Physiol ; 234(10): 17959-17974, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30847933

RESUMEN

Obesity is considered as an independent risk factor for breast cancer (BCa) and plays a major role in the breast tumor microenvironment. The etiology and mechanisms by which obesity contributes to BCa development is not yet understood. Herein, we show that in vitro coculture of BCa cells with mature adipocytes (MA-BCa) increased proliferation, migration, and invasive phenotype of BCa cells. MA-BCa coculture led to increased production of proinflammatory cytokines and chemokines. To identify microRNAs (miRNAs) in BCa cells that are modulated by the presence of adipocytes, we used small RNA sequencing analysis. Sequencing data revealed that 98 miRNAs were differentially expressed in MA-BCa. Among them, miR-3184-5p and miR-181c-3p were found to be the most upregulated and downregulated miRNAs, and direct targets are FOXP4 and PPARα, respectively. In vitro functional assays using a combination of miR-3184-5p inhibitor and miR-181c-3p mimic synergistically decreased adipocytes-induced cell proliferation and invasive capacity of BCa cells. Gene Set Enrichment analysis indicated that transcription factors were highly enriched followed by protein kinases, oncogene, and protein regulators in MA-BCa. GeneGo Metacore pathway analysis uncovered "NOTCH-induced EMT pathway" was found to be the most abundant in MA-BCa. Consistently, epithelial-mesenchymal transition-associated markers were also increased in MA-BCa. The disease enrichment analysis of the predict target genes revealed that diabetes mellitus was significantly affected disease in MA-BCa. Taken together, our data suggest that miRNA-based regulatory mechanism associated with deregulation of pathways and biological functions orchestrated by adipocytes-secreted factors might drive the BCa progression and metastasis in obese patients.


Asunto(s)
Adenocarcinoma/metabolismo , Adipocitos/metabolismo , Neoplasias de la Mama/metabolismo , MicroARNs/metabolismo , Obesidad/metabolismo , Microambiente Tumoral , Células 3T3 , Adenocarcinoma/genética , Adenocarcinoma/secundario , Adipocitos/patología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Comunicación Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Técnicas de Cocultivo , Citocinas/genética , Citocinas/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Ratones , MicroARNs/genética , Invasividad Neoplásica , Obesidad/genética , Obesidad/patología , PPAR alfa/genética , PPAR alfa/metabolismo , Transducción de Señal
2.
J Biol Chem ; 291(53): 27122-27133, 2016 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-27875298

RESUMEN

The antineoplastic agent benzyl isothiocyanate (BITC) acts by targeting multiple pro-oncogenic pathways/genes, including signal transducer and activator of transcription 3 (STAT3); however, the mechanism of action is not well known. As reported previously, BITC induced reactive oxygen species (ROS) in Panc1, MiaPaCa2, and L3.6pL pancreatic cancer cells. This was accompanied by induction of apoptosis and inhibition of cell growth and migration, and these responses were attenuated in cells cotreated with BITC plus glutathione (GSH). BITC also decreased expression of specificity proteins (Sp) Sp1, Sp3, and Sp4 transcription factors (TFs) and several pro-oncogenic Sp-regulated genes, including STAT3 and phospho-STAT3 (pSTAT3), and GSH attenuated these responses. Knockdown of Sp TFs by RNA interference also decreased STAT3/pSTAT3 expression. BITC-induced ROS activated a cascade of events that included down-regulation of c-Myc, and it was also demonstrated that c-Myc knockdown decreased expression of Sp TFs and STAT3 These results demonstrate that in pancreatic cancer cells, STAT3 is an Sp-regulated gene that can be targeted by BITC and other ROS inducers, thereby identifying a novel therapeutic approach for targeting STAT3.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Isotiocianatos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factores de Transcripción Sp/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/uso terapéutico , Factores de Transcripción Sp/genética , Factores de Transcripción Sp/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Phytother Res ; 30(11): 1723-1732, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27384261

RESUMEN

Naturally occurring anticancer agents and their derivatives act on multiple pathways to inhibit carcinogenesis and their inhibition of migration, invasion, growth, survival, and metastasis is associated with downregulation of genes associated with these responses. Several phytochemical-derived anticancer drugs including curcumin, betulinic acid, phenethylisothiocyanate and celastrol, and many others induce reactive oxygen species, and their effects on gene regulation show some overlap in various cancer cell lines. We hypothesize that reactive oxygen species-inducing anticancer agents and many other natural products target a common pathway in cancer cells, which initially involves downregulation of specificity protein 1 (Sp1), Sp3, and Sp4, which are highly expressed in tumors/cell lines derived from solid tumors. This hypothesis is supported by several published reports showing that a large number of phytochemical-derived anticancer agents downregulate Sp1, Sp3, Sp4, and pro-oncogenic Sp-regulated genes involved in cell growth (cyclin D1 and growth factor receptors), survival (bcl-2 and survivin), angiogenesis and migration (MMP-9, vascular endothelial growth factor and its receptors), and inflammation (NF-kB). The contribution of this pathway to the anticancer activity of drugs such as curcumin, celastrol, betulinic acid, and phenethylisothiocyanate must be determined in order to optimize clinical applications of drug combinations containing these compounds. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Antineoplásicos/química , Productos Biológicos/química , Factores de Transcripción Sp/metabolismo , Antineoplásicos/farmacología , Productos Biológicos/farmacología , Humanos
4.
J Biol Chem ; 289(18): 12217-31, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24619425

RESUMEN

Fe65 is a multidomain adaptor with established functions in neuronal cells and neurodegeneration diseases. It binds to the C terminus of the Aß amyloid precursor protein and is involved in regulating gene transcription. The present studies show that Fe65 is expressed in breast cancer (BCa) cells and acts as an ERα transcriptional coregulator that is recruited by 17ß-estradiol to the promoters of estrogen target genes. Deletion analyses mapped the ERα binding domain to the phosphotyrosine binding domain 2 (PTB2). Ectopic Fe65 increased the transcriptional activity of the ERα in a PTB2-dependent manner in reporter assays. Fe65 knockdown decreased, whereas its stable expression increased the transcriptional activity of endogenous ERα in BCa cells and the ability of estrogens to stimulate target gene expression, ERα, and coactivator recruitment to target gene promoters and cell growth. Furthermore, Fe65 expression decreased the antagonistic activity of tamoxifen (TAM), suggesting a role for Fe65 in TAM resistance. Overall, the studies define a novel role for the neuronal adaptor in estrogen actions in BCa cells.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Sitios de Unión/genética , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Células Epiteliales/metabolismo , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Inmunohistoquímica , Células MCF-7 , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Mutación , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tamoxifeno/farmacología
5.
J Biol Chem ; 287(49): 41297-309, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23055531

RESUMEN

Telomerase is an essential enzyme that counteracts the telomere attrition accompanying DNA replication during cell division. Regulation of the promoter activity of the gene encoding its catalytic subunit, the telomerase reverse transcriptase, is established as the dominant mechanism conferring the high telomerase activity in proliferating cells, such as embryonic stem and cancer cells. This study reveals a new mechanism of telomerase regulation through non-coding small RNA by showing that microRNA-498 (miR-498) induced by 1,25-dihydroxyvitamin D3 (1,25(OH)(2)D(3)) decreases the mRNA expression of the human telomerase reverse transcriptase. MiR-498 was first identified in a microarray analysis as the most induced microRNA by 1,25(OH)(2)D(3) in ovarian cancer cells and subsequently validated by quantitative polymerase chain reaction assays in multiple human cancer types. A functional vitamin D response element was defined in the 5-prime regulatory region of the miR-498 genome, which is occupied by the vitamin D receptor and its coactivators. Further studies showed that miR-498 targeted the 3-prime untranslated region of human telomerase reverse transcriptase mRNA and decreased its expression. The levels of miR-498 expression were decreased in malignant human ovarian tumors as well as human ovarian cancer cell lines. The ability of 1,25(OH)(2)D(3) to decrease human telomerase reverse transcriptase mRNA and to suppress ovarian cancer growth was compromised when miR-498 was depleted using the sponges in cell lines and mouse tumor models. Taken together, our studies define a novel mechanism of telomerase regulation by small non-coding RNAs and identify miR-498 as an important mediator for the anti-tumor activity of 1,25(OH)(2)D(3).


Asunto(s)
Calcitriol/farmacología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , MicroARNs/biosíntesis , Neoplasias/metabolismo , Telomerasa/antagonistas & inhibidores , Telomerasa/biosíntesis , Animales , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Femenino , Genoma , Humanos , Ratones , Ratones Desnudos , MicroARNs/fisiología , Mutagénesis , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN no Traducido/metabolismo
6.
J Food Biochem ; 46(9): e14211, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35488719

RESUMEN

An absolute interlinks between inflammation and obesity with scarce investigations on the role of lutein in inflammation-induced obesity motivated us to explore the protective mechanism of lutein on adipogenesis-mediated inflammation in vitro by culturing RAW264.7 macrophages in adipocyte conditioned medium. The RAW264 macrophage cells were cultured with adipocyte-conditioned media, and the potency of lutein on the expression of adipocyte inflammation-associated protein markers (IL-1ß, MCP-1, TNF-α, IL-6, NF-κB, and IKKα/ß) were analyzed by western blotting. The data revealed that lutein effectively reduces the protein levels of major inflammatory markers such as NF-κB, IL-1ß, MCP-1, and TNF-α in differentiated adipocytes. Interestingly, lutein hampered inflammation in the RAW264 cells that were cultured in adipocyte-conditioned media by lowering the protein expression of IL-1ß, MCP-1, and TNF-α. The blockage of inflammation by lutein in both differentiated adipocytes, and adipogenesis-induced macrophages is associated with suppression of IKK α/ß phosphorylation. These data suggest that lutein potentially alters adipocyte differentiation-mediated inflammation by regulating the NF-κB signaling pathway. Thus, lutein could be utilized as a potent nutraceutical agent in the management of obesity and associated inflammation. PRACTICAL APPLICATIONS: Lutein isolated from a dietary source exhibited an inhibitory effect in adipogenesis-induced inflammations. The findings of this study authenticate the diversified prospective of lutein in regulating obesity and other inflammation-related diseases. Thus, it is understood that continuous intake of lutein-rich food or dietary intervention of lutein may reduce the risk of developing obesity.


Asunto(s)
Adipogénesis , Luteína , Animales , Medios de Cultivo Condicionados , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Luteína/farmacología , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Obesidad , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/genética
7.
Mol Cancer ; 9: 311, 2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21138557

RESUMEN

BACKGROUND: MCT-1 oncoprotein accelerates p53 protein degradation via a proteosome pathway. Synergistic promotion of the xenograft tumorigenicity has been demonstrated in circumstance of p53 loss alongside MCT-1 overexpression. However, the molecular regulation between MCT-1 and p53 in tumor development remains ambiguous. We speculate that MCT-1 may counteract p53 through the diverse mechanisms that determine the tumorigenic outcomes. RESULTS: MCT-1 has now identified as a novel target gene of p53 transcriptional regulation. MCT-1 promoter region contains the response elements reactive with wild-type p53 but not mutant p53. Functional p53 suppresses MCT-1 promoter activity and MCT-1 mRNA stability. In a negative feedback regulation, constitutively expressed MCT-1 decreases p53 promoter function and p53 mRNA stability. The apoptotic events are also significantly prevented by oncogenic MCT-1 in a p53-dependent or a p53-independent fashion, according to the genotoxic mechanism. Moreover, oncogenic MCT-1 promotes the tumorigenicity in mice xenografts of p53-null and p53-positive lung cancer cells. In support of the tumor growth are irrepressible by p53 reactivation in vivo, the inhibitors of p53 (MDM2, Pirh2, and Cop1) are constantly stimulated by MCT-1 oncoprotein. CONCLUSIONS: The oppositions between MCT-1 and p53 are firstly confirmed at multistage processes that include transcription control, mRNA metabolism, and protein expression. MCT-1 oncogenicity can overcome p53 function that persistently advances the tumor development.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Mutagénesis Sitio-Dirigida , Proteínas Oncogénicas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/genética
8.
Mol Cancer Res ; 7(4): 536-48, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19372582

RESUMEN

MCT-1 oncoprotein accelerates p53 degradation by means of the ubiquitin-dependent proteolysis. Our present data show that induction of MCT-1 increases chromosomal translocations and deregulated G(2)-M checkpoint in response to chemotherapeutic genotoxin. Remarkably, increases in chromosome copy number, multinucleation, and cytokinesis failure are also promoted while MCT-1 is induced in p53-deficient cells. In such a circumstance, the Ras-mitogen-activated protein kinase/extracellular signal-regulated kinase kinase-mitogen-activated protein kinase signaling activity and the expression of metastatic molecules are amplified. Given a p53-silencing background, MCT-1 malignantly transforms normal breast epithelial cells that are satisfactory for stimulating cell migration/adhesion and tumorigenesis. Detailed analyses of MCT-1 oncogenicity in H1299 p53-null lung cancer cells have shown that ectopically expressed MCT-1 advances xenograft tumorigenicity and angiogenesis, which cannot be completely suppressed by induction of p53. MCT-1 counteracts mutually with p53 at transcriptional levels. Clinical validations confirm that MCT-1 mRNA levels are differentially enriched in comparison between human lung cancer and nontumorigenic tissues. The levels of p53 mRNA are comparatively reduced in a subset of cancer specimens, which highly present MCT-1 mRNA. Our results indicate that synergistic promotions of chromosomal imbalances and oncogenic potency as a result of MCT-1 expression and p53 loss play important roles in tumor development.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Proteínas de Ciclo Celular/genética , Inestabilidad Cromosómica , Neoplasias Pulmonares/genética , Proteínas Oncogénicas/genética , Proteína p53 Supresora de Tumor/genética , Aneuploidia , Animales , Antineoplásicos Fitogénicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Adhesión Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular/fisiología , Proliferación Celular , Análisis Citogenético , Sinergismo Farmacológico , Etopósido/farmacología , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Técnicas para Inmunoenzimas , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Microscopía Fluorescente , Mutágenos/farmacología , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Translocación Genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
9.
Sci Rep ; 10(1): 4456, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32157137

RESUMEN

Edible nanoparticles (ENPs) are nano-sized vesicles derived from edible plants. These ENPs are loaded with plant derived microRNAs, protein, lipids and phytochemicals. Recently, ginger derived ENPs was shown to prevent inflammatory bowel diseases and colon cancer, in vivo, highlighting their therapeutic potential. Conventionally, differential centrifugation with an ultra-centrifugation step is employed to purify these ENPs which imposes limitation on the cost-effectiveness of their purification. Herein, we developed polyethylene glycol-6000 (PEG6000) based ginger ENP purification (PEG-ENPs) method, which eliminates the need for expensive ultracentrifugation. Using different PEG6000 concentrations, we could recover between 60% to 90% of ENPs compared to ultracentrifugation method. PEG-ENPs exhibit near identical size and zeta potential similar to ultra-ENPs. The biochemical composition of PEG-ENPs, such as proteins, lipids, small RNAs and bioactive content is comparable to that of ultra-ENPs. In addition, similar to ultra-ENPs, PEG-ENPs are efficiently taken up by the murine macrophages and protects cells from hydrogen peroxide induced oxidative stress. Since PEG has been approved as food additive, the PEG method described here will provide a cost-effective alternative to purify ENPs, which can be directly used as a dietary supplement in therapeutic formulations.


Asunto(s)
Macrófagos/citología , Nanopartículas/administración & dosificación , Nanopartículas/economía , Polietilenglicoles/química , Rizoma/química , Zingiber officinale/química , Animales , Proliferación Celular , Células Cultivadas , Análisis Costo-Beneficio , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Nanopartículas/química
10.
Mol Cancer Res ; 17(3): 794-805, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30610105

RESUMEN

Methyl 2-trifluoromethyl-3,11-dioxo-18ß-olean-1,12-dien-3-oate (CF3DODA-Me) is derived synthetically from glycyrrhetinic acid, a major component of licorice, and this compound induced reactive oxygen species (ROS) in RD and Rh30 rhabdomyosarcoma (RMS) cells. CF3DODA-Me also inhibited growth and invasion and induced apoptosis in RMS cells, and these responses were attenuated after cotreatment with the antioxidant glutathione, demonstrating the effective anticancer activity of ROS in RMS. CF3DODA-Me also downregulated expression of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 and prooncogenic Sp-regulated genes including PAX3-FOXO1 (in Rh30 cells). The mechanism of CF3DODA-Me-induced Sp-downregulation involved ROS-dependent repression of c-Myc and cMyc-regulated miR-27a and miR-17/20a, and this resulted in induction of the miRNA-regulated Sp repressors ZBTB4, ZBTB10, and ZBTB34. The cell and tumor growth effects of CF3DODA-Me further emphasize the sensitivity of RMS cells to ROS inducers and their potential clinical applications for treating this deadly disease. IMPLICATIONS: CF3DODA-Me and HDAC inhibitors that induce ROS-dependent Sp downregulation could be developed for clinical applications in treating rhabdomyosarcoma.


Asunto(s)
Especies Reactivas de Oxígeno/metabolismo , Rabdomiosarcoma/tratamiento farmacológico , Factores de Transcripción Sp/genética , Triterpenos/uso terapéutico , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Desnudos , Rabdomiosarcoma/patología , Transfección , Triterpenos/farmacología
12.
DNA Repair (Amst) ; 6(9): 1319-32, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17416211

RESUMEN

Tumor suppressor p53 protein mediates checkpoint controls and the apoptotic program that are critical for maintaining genomic integrity and preventing tumorigenesis. Forced-induction of MCT-1 decreased p53 expression before and after genomic insults. While inhibiting protein synthesis, the levels of ubiquinated-p53 and the phospho-MDMA2 were significantly increased in ectopic MCT-1 cells. Abrogation of the proteosome degradation process attenuated p53 destabilization and p21 down-regulation by MCT-1. Concomitantly, MCT-1 overexpression enhanced the phosphorylation status of MAPK (ERK1/ERK2). While MCT-1 gene knockdown or MEK/ERK pathway inhibition dramatically reduced MAPK phosphorylation, the genotoxin-induced p53 and p21 production were noticeably elevated. Upon Etoposide treatment, ectopic MCT-1 cells relaxed S-phase and G2/M checkpoints followed by G1 phase progressing. Moreover, cells inducing with MCT-1 abridged accumulations of G2/M populations in the response to gamma-irradiation. The polyploidy (DNA content>4N) populations were increased in association with p53 loss in MCT-1 oncogenic cells. Alkaline comet assay validated that ectopic MCT-1 cells were less susceptibility to the genotoxicity. Furthermore, the allocation of nuclear MCT-1 induced by the genotoxic stress was moderately coincided with gamma-H2AX appearances. Throughout damage-repairing process, ectopic MCT-1 cells displayed many larger chromosomes and multiple chromosomal fusions compared to the controls that showed increase in chromosomal breaks/gaps and minute chromosomal fragments. Spectral karyotyping analysis precisely identified the acquisition of a single extra copy of chromosome 14 together with a complex genome organizations in ectopic MCT-1 cells, including extra copies of chromosome segments that had been translocated to derivative chromosomes 6 [der(6)] and 9 [der(9)]. In conclusion, MCT-1 deregulates p53-p21 network and impairs the damage checkpoints those are robustly connected to oncogenic chromosomal abnormalities.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Inestabilidad Cromosómica , Daño del ADN , Proteínas Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/genética , Células Cultivadas , Ensayo Cometa , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Replicación del ADN , Regulación hacia Abajo , Fibroblastos/citología , Fibroblastos/metabolismo , Fase G1 , Fase G2 , Histonas/metabolismo , Humanos , Cariotipificación , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Oncogénicas/genética , Fase S , Translocación Genética
13.
Adv Nutr ; 8(6): 868-888, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29141971

RESUMEN

Breast cancer is the most common malignancy diagnosed in women, and the incidence of breast cancer is increasing every year. Obesity has been identified as one of the major risk factors for breast cancer progression. The mechanisms by which obesity contributes to breast cancer development is not yet understood; however, there are a few mechanisms counted as potential producers of breast cancer in obesity, including insulin resistance, chronic inflammation and inflammatory cytokines, adipokines, and sex hormones. Recent emerging evidence suggests that alterations in microRNA (miRNA) expressions are found in several diseases, including breast cancer and obesity; however, miRNA roles in obesity-linked breast cancer are beginning to unravel. miRNAs are thought to be potential noninvasive biomarkers for diagnosis and prognosis of cancer patients with comorbid conditions of obesity as well as therapeutic targets. Recent studies have evidenced that nutrients and other dietary factors protect against cancer and obesity through modulation of miRNA expressions. Herein, we summarize a comprehensive overview of up-to-date information related to miRNAs and their molecular targets involved in obesity-associated breast cancer. We also address the mechanisms by which dietary factors modulate miRNA expression and its protective roles in obesity-associated breast cancer. It is hoped that this review would provide new therapeutic strategies for the treatment of obesity-associated breast cancer to reduce the burden of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Dieta/efectos adversos , MicroARNs/metabolismo , Obesidad/complicaciones , Neoplasias de la Mama/etiología , Femenino , Humanos , Obesidad/genética
14.
Cancer Prev Res (Phila) ; 10(8): 467-477, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28673967

RESUMEN

Piperlongumine is a natural product found in the plant species Piper longum, and this compound exhibits potent anticancer activity in multiple tumor types and has been characterized as an inducer of reactive oxygen species (ROS). Treatment of Panc1 and L3.6pL pancreatic, A549 lung, 786-O kidney, and SKBR3 breast cancer cell lines with 5 to 15 µmol/L piperlongumine inhibited cell proliferation and induced apoptosis and ROS, and these responses were attenuated after cotreatment with the antioxidant glutathione. Piperlongumine also downregulated expression of Sp1, Sp3, Sp4, and several pro-oncogenic Sp-regulated genes, including cyclin D1, survivin, cMyc, EGFR and hepatocyte growth factor receptor (cMet), and these responses were also attenuated after cotreatment with glutathione. Mechanistic studies in Panc1 cells showed that piperlongumine-induced ROS decreased expression of cMyc via an epigenetic pathway, and this resulted in downregulation of cMyc-regulated miRNAs miR-27a, miR-20a, and miR-17 and induction of the transcriptional repressors ZBTB10 and ZBTB4. These repressors target GC-rich Sp-binding sites to decrease transactivation. This pathway observed for piperlongumine in Panc1 cells has previously been reported for other ROS-inducing anticancer agents and shows that an important underlying mechanism of action of piperlongumine is due to downregulation of Sp1, Sp3, Sp4, and pro-oncogenic Sp-regulated genes. Cancer Prev Res; 10(8); 467-77. ©2017 AACR.


Asunto(s)
Antineoplásicos/farmacología , Dioxolanos/farmacología , Especies Reactivas de Oxígeno , Factores de Transcripción Sp/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Factores de Transcripción Sp/biosíntesis
15.
J Steroid Biochem Mol Biol ; 173: 157-167, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28159673

RESUMEN

Receptor interacting protein kinase 1 (RIPK1) is an enzyme acting downstream of tumor necrosis factor alpha to control cell survival and death. RIPK1 expression has been reported to cause drug resistance in cancer cells, but so far, no published studies have investigated the role of RIPK1 in vitamin D signaling. In the present study, we investigated whether RIPK1 plays any roles in 1,25-dihydroxyvitamin D3 (1,25D3)-induced growth suppression. In our studies, RIPK1 decreased the transcriptional activity of vitamin D receptor (VDR) in luciferase reporter assays independent of its kinase activity, suggesting a negative role of RIPK1 in 1,25D3 action. RIPK1 also formed a complex with VDR, and deletion analyses mapped the RIPK1 binding region to the C-terminal ligand-binding domain of the VDR. Subcellular fractionation analyses indicated that RIPK1 increased VDR retention in the cytoplasm, which may account for its inhibition of VDR transcriptional activity. Consistent with the reporter analyses, 1,25D3-induced growth suppression was more pronounced in RIPK1-null MEFs and RIPK1-knockdown ovarian cancer cells than in control cells. Our studies have defined RIPK1 as a VDR repressor, projecting RIPK1 depletion as a potential strategy to increase the potency of 1,25D3 and its analogs for cancer intervention.


Asunto(s)
Proliferación Celular , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , Línea Celular Tumoral , Citoplasma/química , Citoplasma/genética , Citoplasma/metabolismo , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Unión Proteica , Dominios Proteicos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Receptores de Calcitriol/química , Receptores de Calcitriol/genética , Vitamina D/metabolismo
16.
J Steroid Biochem Mol Biol ; 148: 138-47, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25448740

RESUMEN

Epithelial ovarian cancer (EOC) is the leading cause of gynecological cancer death in women, mainly because it has spread to intraperitoneal tissues such as the omentum in the peritoneal cavity by the time of diagnosis. In the present study, we established in vitro assays, ex vivo omental organ culture system and syngeneic animal tumor models using wild type (WT) and vitamin D receptor (VDR) null mice to investigate the effects of 1α,25-dihydroxyvitamin D3 (1,25D3) and VDR on EOC invasion. Treatment of human EOC cells with 1,25D3 suppressed their migration and invasion in monolayer scratch and transwell assays and ability to colonize the omentum in the ex vivo system, supporting a role for epithelial VDR in interfering with EOC invasion. Furthermore, VDR knockdown in OVCAR3 cells increased their ability to colonize the omentum in the ex vivo system in the absence of 1,25D3, showing a potential ligand-independent suppression of EOC invasion by epithelial VDR. In syngeneic models, ID8 tumors exhibited an increased ability to colonize omenta of VDR null over that of WT mice; pre-treatment of WT, not VDR null, mice with EB1089 reduced ID8 colonization, revealing a role for stromal VDR in suppressing EOC invasion. These studies are the first to demonstrate a role for epithelial and stromal VDR in mediating the activity of 1,25D3 as well as a 1,25D3-independent action of the VDR in suppressing EOC invasion. The data suggest that VDR-based drug discovery may lead to the development of new intervention strategies to improve the survival of patients with EOC at advanced stages. This article is part of a Special Issue entitled "Vitamin D Workshop".


Asunto(s)
Calcitriol/farmacología , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Epiplón/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Receptores de Calcitriol/metabolismo , Vitaminas/farmacología , Animales , Carcinoma Epitelial de Ovario , Femenino , Humanos , Ratones , Invasividad Neoplásica , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Epiplón/metabolismo , Epiplón/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología
17.
Cancer Res ; 74(21): 6194-204, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25252917

RESUMEN

Obesity is a pandemic and major risk factor for cancers. The reduction of obesity would have been an effective strategy for cancer prevention, but the reality is that worldwide obesity has kept increasing for decades, remaining a major avoidable cancer risk secondary only to smoke. The present studies suggest that vitamin D may be an effective agent to reduce obesity-associated cancer risks in women. Molecular analyses showed that leptin increased human telomerase reverse transcriptase (hTERT) mRNA expression and cell growth through estrogen receptor-α (ERα) activation in ovarian cancer cells, which was suppressed by 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3]. The suppression was compromised when miR-498 induction by the hormone was depleted with microRNA (miRNA) sponges. In mice, high-fat diet (HFD) stimulation of ovarian tumor growth was remarkably suppressed by 1,25(OH)2D3 analogue EB1089, which was also compromised by miR-498 sponges. EB1089 did not alter HFD-induced increase in serum leptin levels but increased miR-498 and decreased the diet-induced hTERT expression in tumors. Quantitative RT-PCR analyses revealed an inverse correlation between hTERT mRNA and miR-498 in response to 1,25(OH)2D3 in estrogen-sensitive ovarian, endometrial, and breast cancers. The studies suggest that miR-498-mediated hTERT downregulation is a key event mediating the anti-leptin activity of 1,25(OH)2D3 in estrogen-sensitive tumors in women.


Asunto(s)
Neoplasias de la Mama/genética , Leptina/biosíntesis , MicroARNs/biosíntesis , Neoplasias Ováricas/genética , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dieta Alta en Grasa , Receptor alfa de Estrógeno/biosíntesis , Receptor alfa de Estrógeno/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leptina/metabolismo , Ratones , MicroARNs/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Telomerasa/biosíntesis , Telomerasa/genética , Vitamina D/administración & dosificación
18.
Mol Cell Endocrinol ; 338(1-2): 58-67, 2011 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-21458521

RESUMEN

1alpha,25-dihydroxyvitamin D3, 1,25(OH)(2)D(3), regulates gene expression through the vitamin D receptor. The present studies identify the epidermal growth factor receptor, EGFR, as a target gene suppressed by 1,25(OH)(2)D(3) in human ovarian cancer cells. The suppression was detected at both mRNA and protein levels in vitamin D-sensitive human ovarian cancer cells. A novel vitamin D response element was identified in intron 1 of the EGFR genome, a known hotspot for its transcriptional regulation. Chromatin immunoprecipitations and reporter gene analyses showed that the intronic DNA element bound to vitamin D receptor and a co-repressor and was functional in mediating transcriptional suppression of EGFR promoter by 1,25(OH)(2)D(3) under stable transfection conditions. Consistent with the EGFR down regulation, 1,25(OH)(2)D(3) suppressed activation of the external signal regulated kinase by epidermal growth factors. Over expression of an active EGFR in vitamin D sensitive ovarian cancer cells caused resistance to 1,25(OH)(2)D(3)-induced growth suppression and diminished the hormonal regulation of cyclin D1, cyclin E, Skp2 and p27, a group of cell cycle regulators that mediate 1,25(OH)(2)D(3)-induced cell cycle arrest at G1-S checkpoint. Taken together, our studies demonstrate that 1,25(OH)(2)D(3) suppresses the response of human ovarian cancer cells to mitogenic growth factors and couple the suppression to the cell cycle arrest at G1-S checkpoint by the hormone.


Asunto(s)
Calcitriol/farmacología , Regulación hacia Abajo/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Ováricas/fisiopatología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclinas/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/agonistas , Receptores ErbB/genética , Femenino , Genes Reporteros , Humanos , Intrones , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Transducción de Señal , Elemento de Respuesta a la Vitamina D
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA