Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Sarcoma ; 2011: 598218, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21559267

RESUMEN

Chondrocytes are mesenchymally derived cells that reportedly acquire some epithelial characteristics; however, whether this is a progression through a mesenchymal to epithelial transition (MET) during chondrosarcoma development is still a matter of investigation. We observed that chondrosarcoma cells acquired the expression of four epithelial markers, E-cadherin,desmocollin 3, maspin, and 14-3-3σ, all of which are governed epigenetically through cytosine methylation. Indeed, loss of cytosine methylation was tightly associated with acquired expression of both maspin and 14-3-3σ in chondrosarcomas. In contrast, chondrocyte cells were negative for maspin and 14-3-3σ and displayed nearly complete DNA methylation. Robust activation of these genes was also observed in chondrocyte cells following 5-aza-dC treatment. We also examined the transcription factor snail which has been reported to be an important mediator of epithelial to mesenchymal transitions (EMTs). In chondrosarcoma cells snail is downregulated suggesting a role for loss of snail expression in lineage maintenance. Taken together, these results document an epigenetic switch associated with an MET-like phenomenon that accompanies chondrosarcoma progression.

3.
Otolaryngol Head Neck Surg ; 137(5): 722-8, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17967635

RESUMEN

OBJECTIVE: To determine if epigenetic-modifying drugs can increase iodine uptake in thyroid carcinoma cell lines. STUDY DESIGN: Human thyroid carcinoma cell lines were tested for iodine uptake before and after treatment with epigenetic-modifying agents. RESULTS: Thyroid carcinoma cell lines DRO and 2-7 had high levels of DNA methylation (74% and 80%) compared with normal thyroid tissue (6%) (P < 0.05). This finding correlated with low levels of sodium iodide symporter (NIS) expression in the untreated thyroid carcinoma cell line. Combination treatment with the epigenetic-modifying agents 5-aza-2'-deoxycytidine and sodium butyrate resulted in increases in NIS messenger RNA levels, global histone acetylation, and 9- and 8-fold increases in I(125) uptake for the DRO and 2-7 cells, respectively. CONCLUSIONS: Epigenetic-modifier drugs represent a novel adjuvant treatment for those patients with radioablation-resistant thyroid cancer. SIGNIFICANCE: Epigenetic-modifying agents show potential for treatment of radioablation-resistant thyroid cancer.


Asunto(s)
Azacitidina/análogos & derivados , Butiratos/administración & dosificación , Epigénesis Genética/efectos de los fármacos , Radioisótopos de Yodo/metabolismo , Neoplasias de la Tiroides/radioterapia , Acetilación/efectos de los fármacos , Azacitidina/administración & dosificación , Western Blotting , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Decitabina , Quimioterapia Combinada , Humanos , Radioisótopos de Yodo/uso terapéutico , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Yoduro de Sodio/metabolismo , Simportadores/análisis , Simportadores/genética , Neoplasias de la Tiroides/metabolismo
4.
PLoS One ; 12(10): e0186603, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29040313

RESUMEN

Thoracic aortic aneurysm and dissection are life-threatening complications of Marfan syndrome (MFS). Studies of human and mouse aortic samples from late stage MFS demonstrate increased TGF-ß activation/signaling and diffuse matrix changes. However, the role of the aortic smooth muscle cell (SMC) phenotype in early aneurysm formation in MFS has yet to be fully elucidated. As our objective, we investigated whether an altered aortic SMC phenotype plays a role in aneurysm formation in MFS. We describe previously unrecognized concordant findings in the aortas of a murine model of MFS, mgR, during a critical and dynamic phase of early development. Using Western blot, gelatin zymography, and histological analysis, we demonstrated that at postnatal day (PD) 7, before aortic TGF-ß levels are increased, there is elastic fiber fragmentation/disorganization and increased levels of MMP-2 and MMP-9. Compared to wild type (WT) littermates, aortic SMCs in mgR mice express higher levels of contractile proteins suggesting a switch to a more mature contractile phenotype. In addition, tropoelastin levels are decreased in mgR mice, a finding consistent with a premature switch to a contractile phenotype. Proliferation assays indicate a decrease in the proliferation rate of mgR cultured SMCs compared to WT SMCs. KLF4, a regulator of smooth muscle cell phenotype, was decreased in aortic tissue of mgR mice. Finally, overexpression of KLF4 partially reversed this phenotypic change in the Marfan SMCs. This study indicates that an early phenotypic switch appears to be associated with initiation of important metabolic changes in SMCs that contribute to subsequent pathology in MFS.


Asunto(s)
Aorta Torácica/patología , Aneurisma de la Aorta Torácica/patología , Matriz Extracelular/patología , Síndrome de Marfan/patología , Miocitos del Músculo Liso/patología , Animales , Aorta Torácica/metabolismo , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/metabolismo , Diferenciación Celular , Proliferación Celular , Tejido Elástico/metabolismo , Tejido Elástico/patología , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/metabolismo , Fenotipo , Cultivo Primario de Células , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Tropoelastina/genética , Tropoelastina/metabolismo
5.
Leuk Res ; 37(8): 963-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23537707

RESUMEN

This study evaluates the role of scavenger receptor class A member 3 (SCARA3) in multiple myeloma (MM). SCARA3 expression was induced upon treatment with oxidative stressors (ionizing radiation and chemotherapeutic drugs). An epigenetic inactivation of SCARA3 was noted in MM.1S myeloma cells. Myeloma cell killing by dexamethasone and bortezomib was inhibited by up-regulation of SCARA3 while SCARA3 knockdown sensitized myeloma cells to the drugs. Clinical samples showed an inverse correlation between SCARA3 gene expression, myeloma progression, and favorable clinical prognosis. In MM, SCARA3 protects against oxidative stress-induced cell killing and can serve as predictor of MM progression and therapeutic response.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Proteínas de Choque Térmico/genética , Mieloma Múltiple/genética , Receptores Depuradores de Clase A/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular , Línea Celular Tumoral , Decitabina , Dexametasona/farmacología , Progresión de la Enfermedad , Inhibidores Enzimáticos/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Técnicas de Silenciamiento del Gen , Proteínas de Choque Térmico/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Ácidos Hidroxámicos/farmacología , Mieloma Múltiple/patología , Mieloma Múltiple/terapia , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidantes/farmacología , Pronóstico , Pirazinas/farmacología , Radiación Ionizante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptores Depuradores de Clase A/metabolismo
6.
Mol Cancer Res ; 10(1): 40-51, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22064654

RESUMEN

Extracellular superoxide dismutase (EcSOD) is an important superoxide scavenger in the lung in which its loss, sequence variation, or abnormal expression contributes to lung diseases; however, the role of EcSOD in lung cancer has yet to be studied. We hypothesized that EcSOD loss could affect malignant progression in lung, and could be either genetic or epigenetic in nature. To test this, we analyzed EcSOD expression, gene copy number, promoter methylation, and chromatin accessibility in normal lung and carcinoma cells. We found that normal airway epithelial cells expressed abundant EcSOD and had an unmethylated promoter, whereas EcSOD-negative lung cancer cells displayed aberrant promoter hypermethylation and decreased chromatin accessibility. 5-aza-dC induced EcSOD suggesting that cytosine methylation was causal, in part, to silencing. In 48/50 lung tumors, EcSOD mRNA was significantly lower as early as stage I, and the EcSOD promoter was hypermethylated in 8/10 (80%) adenocarcinomas compared with 0/5 normal lung samples. In addition, 20% of the tumors showed loss of heterozygosity (LOH) of EcSOD. Reexpression of EcSOD attenuated the malignant phenotype of lung carcinoma cells by significantly decreasing invasion and survival. Finally, EcSOD decreased heparanase and syndecan-1 mRNAs in part by reducing NF-κB. By contrast, MnSOD and CuZnSOD showed no significant changes in lung tumors and had no effect on heparanase expression. Taken together, the loss of EcSOD expression is unique among the superoxide dismutases in lung cancer and is the result of EcSOD promoter methylation and LOH, suggesting that its early loss may contribute to ECM remodeling and malignant progression.


Asunto(s)
Adenocarcinoma/patología , Matriz Extracelular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa/fisiología , Adenocarcinoma/genética , Células Cultivadas , Metilación de ADN/genética , Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Matriz Extracelular/fisiología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/fisiología , Silenciador del Gen/fisiología , Homeostasis/genética , Humanos , Pérdida de Heterocigocidad/fisiología , Neoplasias Pulmonares/genética , Invasividad Neoplásica , Fenotipo , Regiones Promotoras Genéticas , Regulación hacia Arriba/genética
7.
PLoS One ; 6(1): e14617, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21297970

RESUMEN

BACKGROUND: The prolyl-hydroxylase domain family of enzymes (PHD1-3) plays an important role in the cellular response to hypoxia by negatively regulating HIF-α proteins. Disruption of this process can lead to up-regulation of factors that promote tumorigenesis. We observed decreased basal expression of PHD3 in prostate cancer tissue and tumor cell lines representing diverse tissues of origin. Furthermore, some cancer lines displayed a failure of PHD3 mRNA induction when introduced to a hypoxic environment. This study explores the mechanism by which malignancies neither basally express PHD3 nor induce PHD3 under hypoxic conditions. METHODOLOGY/PRINCIPAL FINDINGS: Using bisulfite sequencing and methylated DNA enrichment procedures, we identified human PHD3 promoter hypermethylation in prostate, breast, melanoma and renal carcinoma cell lines. In contrast, non-transformed human prostate and breast epithelial cell lines contained PHD3 CpG islands that were unmethylated and responded normally to hypoxia by upregulating PHD3 mRNA. Only treatment of cells lines containing PHD3 promoter hypermethylation with the demethylating drug 5-aza-2'-deoxycytidine significantly increased the expression of PHD3. CONCLUSIONS/SIGNIFICANCE: We conclude that expression of PHD3 is silenced by aberrant CpG methylation of the PHD3 promoter in a subset of human carcinoma cell lines of diverse origin and that this aberrant cytosine methylation status is the mechanism by which these cancer cell lines fail to upregulate PHD3 mRNA. We further show that a loss of PHD3 expression does not correlate with an increase in HIF-1α protein levels or an increase in the transcriptional activity of HIF, suggesting that loss of PHD3 may convey a selective advantage in some cancers by affecting pathway(s) other than HIF.


Asunto(s)
Islas de CpG/genética , Metilación de ADN , Dioxigenasas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Regiones Promotoras Genéticas/genética , Línea Celular Tumoral , Dioxigenasas/deficiencia , Femenino , Silenciador del Gen , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Masculino , Neoplasias/patología , ARN Mensajero/análisis
8.
Radiat Res ; 174(3): 290-6, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20726720

RESUMEN

Trypanothione is a unique diglutathionyl-spermidine conjugate found in abundance in trypanosomes but not in other eukaryotes. Because trypanothione is a naturally occurring polyamine thiol reminiscent of the synthetic drug amifostine, it may be a useful protector against radiation and oxidative stress. For these reasons we hypothesized that trypanothione might serve as a radioprotective agent when produced in bacteria. To accomplish this objective, the trypanothione synthetase and reductase genes from T. cruzi were introduced into E. coli and their expression was verified by qPCR and immunoblotting. Trypanothione synthesis in bacteria, detected by HPLC, resulted in decreased intracellular levels of reactive oxygen species as determined by H(2)DCFDA oxidation. Moreover, E. coli genomic DNA was protected from radiation-induced DNA damage by 4.6-fold in the presence of trypanothione compared to control bacteria. Concordantly, the transgenic E. coli expressing trypanothione were 4.3-fold more resistant to killing by (137)Cs gamma radiation compared to E. coli devoid of trypanothione expression. Thus we have shown for the first time that E. coli can be genetically engineered to express the trypanothione biosynthetic pathway and produce trypanothione, which results in their radioresistance. These results warrant further research to explore the possibility of developing trypanothione as a novel radioprotective agent.


Asunto(s)
Escherichia coli/metabolismo , Glutatión/análogos & derivados , Espermidina/análogos & derivados , Transgenes , Animales , Secuencia de Bases , Cromatografía Líquida de Alta Presión , Daño del ADN , Cartilla de ADN , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Escherichia coli/efectos de la radiación , Glutatión/biosíntesis , Estrés Oxidativo , Reacción en Cadena de la Polimerasa , Espermidina/biosíntesis , Trypanosoma cruzi/genética
9.
Cancer Res ; 69(15): 6355-63, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19602586

RESUMEN

Increased expression of heparanase stimulates the progression of various human cancers, including breast cancer. Therefore, a deeper understanding of the mechanisms involved in regulating heparanase is critical in developing effective treatments for heparanase-overexpressing cancers. In this study, we investigated the potential use of extracellular superoxide dismutase (EcSOD) to enhance the inhibitory effects of heparin/low molecular weight heparin (LMWH) in breast cancer cells. EcSOD binds to cell surfaces and the extracellular matrix through heparin-binding domain (HBD). Deleting this HBD rendered the protein a more potent inhibitor of breast cancer growth, survival, and invasion. Among the treatment combinations examined, EcSODDeltaHBD plus LMWH provided the best tumor suppressive effects in inhibiting breast cancer growth and invasion in vitro. We have further shown that overexpression of EcSOD decreased accumulation of vascular endothelial growth factor in the culture medium and increased the level of intact cell surface-associated heparan sulfate, thus implicating inhibition of heparanase expression as a potential mechanism. Overexpression of EcSOD inhibited steady-state heparanase mRNA levels by >50% as determined by quantitative reverse transcription-PCR. Moreover, heparanase promoter activation was suppressed by EcSOD as indicated by a luciferase reporter assay. These findings reveal a previously unrecognized molecular pathway showing that regulation of heparanase transcription can be mediated by oxidative stress. Our study implies that overexpression of EcSOD is a promising strategy to enhance the efficacy of heparin/LMWH by inhibiting heparanase as a novel treatment for breast cancer.


Asunto(s)
Neoplasias de la Mama/enzimología , Glucuronidasa/biosíntesis , Superóxido Dismutasa/biosíntesis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glucuronidasa/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina de Bajo-Peso-Molecular/farmacología , Humanos , Invasividad Neoplásica , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Transcripción Genética , Transfección , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Exp Mol Pathol ; 83(2): 277-82, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17651731

RESUMEN

Amyloid precursor protein (APP) has been implicated in squamous cell carcinoma. In this study we show that forced expression of the transcription factor activating protein 2alpha (AP-2alpha) results in significantly increased steady state levels of APP mRNA in human keratinocytes. Sequence analysis of the 5' end of the human APP gene revealed five putative binding sites for AP-2, suggesting that APP is a direct target for transactivation by AP-2. AP-2 protein bound at least 3 of these putative promoter elements in vitro as determined by electrophoretic mobility shift assay. Chromatin immunoprecipitation (ChIP) analysis showed that these binding sites were occupied by AP-2 in cells, thus indicating the relevance to AP-2 binding in vivo. We then analyzed APP and AP-2 mRNA and protein expression in squamous cell carcinoma tumor samples. Analysis of RNA extracted from human tissue showed a significant positive correlation between AP-2alpha and APP mRNA expression. Immunohistochemical staining of tumor samples also demonstrated a positive correlation which was substantiated through western blot studies. Taken together, these findings demonstrate a role for the transcription factor AP-2alpha in the regulation of APP gene expression in human keratinocytes.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Boca/genética , Receptores de Superficie Celular/genética , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/fisiología , Transcripción Genética , Carcinoma de Células Escamosas/patología , Línea Celular , Cromatina/genética , Cartilla de ADN , Humanos , Queratinocitos/fisiología , Neoplasias de la Boca/patología , Nexinas de Proteasas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
11.
J Biol Chem ; 282(2): 1322-33, 2007 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-17110379

RESUMEN

The ARF tumor suppressor signals through p53 and other poorly defined anti-proliferative pathways to block carcinogenesis. In a search for new regulators of ARF signaling, we discovered a novel nuclear protein that we named NIAM (nuclear interactor of ARF and MDM2) for its ability to bind both ARF and the p53 antagonist MDM2. NIAM protein is normally expressed at low to undetectable levels in cells because of, at least in part, MDM2-mediated ubiquitination and proteasomal degradation. When reintroduced into cells, NIAM activated p53, caused a G1 phase cell cycle arrest, and collaborated with ARF in an additive fashion to suppress proliferation. Notably, NIAM retains growth inhibitory activity in cells lacking ARF and/or p53, and knockdown experiments revealed that it is not essential for ARF-mediated growth inhibition. Thus, NIAM and ARF act in separate anti-proliferative pathways that intersect mechanistically and suppress growth more effectively when jointly activated. Intriguingly, silencing of NIAM accelerated chromosomal instability, and microarray analyses showed reduced NIAM mRNA expression in numerous primary human tumors. This study identifies a novel protein with tumor suppressor-like behaviors and functional links to ARF-MDM2-p53 signaling.


Asunto(s)
Cromosomas/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Adenocarcinoma , Ancrod , Animales , Neoplasias de la Mama , División Celular/fisiología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Fibroblastos/citología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares , Osteosarcoma , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor , Ubiquitina/metabolismo
12.
Gynecol Oncol ; 102(2): 319-24, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16457875

RESUMEN

OBJECTIVE: Maspin expression is often deregulated in human cancer cells compared to their normal cells due to loss of epigenetic control. In contrast to normal human ovarian surface epithelial (HOSE) cells, ovarian carcinoma cells display a gain of maspin mRNA expression. The objective of this study was to determine whether gain of maspin expression in ovarian cancer is governed by epigenetic mechanisms. METHODS: We examined the cytosine methylation and chromatin accessibility status of the maspin promoter in normal HOSE cells and ovarian carcinoma cells with real-time RT-PCR, sodium bisulfite genomic sequencing, and chromatin accessibility assays. 5-Aza-2'-deoxycytidine (5-aza-dC) was used to induce demethylation of the maspin promoter. Ad p53 was used to induce transient overexpression of wild-type p53. RESULTS: Normal HOSE cells were maspin-negative in association with methylation of the maspin promoter. In the maspin-positive ovarian cancer cell lines, the maspin promoter was unmethylated. Increased maspin expression in ovarian carcinoma cells was accompanied by a more accessible chromatin structure in the maspin promoter. In the maspin-negative ovarian cancer cell line A222, maspin could be induced following 5-aza-dC treatment or by forced overexpression of p53. CONCLUSIONS: These results suggest that changes in cytosine methylation and chromatin accessibility play an important role in maspin expression in human ovarian carcinoma. Deregulation of maspin expression in ovarian cancer is due to loss of epigenetic control as has been shown in other cancers. This observation provides further evidence of the strict epigenetic control of the maspin gene.


Asunto(s)
Neoplasias Ováricas/genética , Serpinas/genética , Cromatina/genética , Metilación de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Neoplasias Ováricas/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Serpinas/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA