Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 12(10)2023 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-37408267

RESUMEN

Indoleamine 2,3-dioxygenase 2 (IDO2) is a tryptophan-catabolizing enzyme and a homolog of IDO1 with a distinct expression pattern compared with IDO1. In dendritic cells (DCs), IDO activity and the resulting changes in tryptophan level regulate T-cell differentiation and promote immune tolerance. Recent studies indicate that IDO2 exerts an additional, non-enzymatic function and pro-inflammatory activity, which may play an important role in diseases such as autoimmunity and cancer. Here, we investigated the impact of aryl hydrocarbon receptor (AhR) activation by endogenous compounds and environmental pollutants on the expression of IDO2. Treatment with AhR ligands induced IDO2 in MCF-7 wildtype cells but not in CRISPR-cas9 AhR-knockout MCF-7 cells. Promoter analysis with IDO2 reporter constructs revealed that the AhR-dependent induction of IDO2 involves a short-tandem repeat containing four core sequences of a xenobiotic response element (XRE) upstream of the start site of the human ido2 gene. The analysis of breast cancer datasets revealed that IDO2 expression increased in breast cancer compared with normal samples. Our findings suggest that the AhR-mediated expression of IDO2 in breast cancer could contribute to a pro-tumorigenic microenvironment in breast cancer.


Asunto(s)
Neoplasias de la Mama , Indolamina-Pirrol 2,3,-Dioxigenasa , Receptores de Hidrocarburo de Aril , Femenino , Humanos , Neoplasias de la Mama/genética , Diferenciación Celular , Tolerancia Inmunológica , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Triptófano/metabolismo , Microambiente Tumoral , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo
2.
Br J Cancer ; 127(3): 436-448, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35440669

RESUMEN

BACKGROUND: LRIG1, the founding member of the LRIG (leucine-rich repeat and immunoglobulin-like domain) family of transmembrane proteins, is a negative regulator of receptor tyrosine kinases and a tumour suppressor. Decreased LRIG1 expression is consistently observed in cancer, across diverse tumour types, and is linked to poor patient prognosis. However, mechanisms by which LRIG1 is repressed are not fully understood. Silencing of LRIG1 through promoter CpG island methylation has been reported in colorectal and cervical cancer but studies in breast cancer remain limited. METHODS: In silico analysis of human breast cancer patient data were used to demonstrate a correlation between DNA methylation and LRIG1 silencing in basal/triple-negative breast cancer, and its impact on patient survival. LRIG1 gene expression, protein abundance, and methylation enrichment were examined by quantitative reverse-transcription PCR, immunoblotting, and methylation immunoprecipitation, respectively, in breast cancer cell lines in vitro. We examined the impact of global demethylation on LRIG1 expression and methylation enrichment using 5-aza-2'-deoxycytidine. We also examined the effects of targeted demethylation of the LRIG1 CpG island, and transcriptional activation of LRIG1 expression, using the RNA guided deadCas9 transactivation system. RESULTS: Across breast cancer subtypes, LRIG1 expression is lowest in the basal/triple-negative subtype so we investigated whether differential methylation may contribute to this. Indeed, we find that LRIG1 CpG island methylation is most prominent in basal/triple-negative cell lines and patient samples. Use of the global demethylating agent 5-aza-2'-deoxycytidine decreases methylation leading to increased LRIG1 transcript expression in basal/triple-negative cell lines, while having no effect on LRIG1 expression in luminal/ER-positive cell lines. Using a CRISPR/deadCas9 (dCas9)-based targeting approach, we demonstrate that TET1-mediated demethylation (Tet1-dCas9) along with VP64-mediated transcriptional activation (VP64-dCas9) at the CpG island, increased endogenous LRIG1 expression in basal/triple-negative breast cancer cells, without transcriptional upregulation at predicted off-target sites. Activation of LRIG1 by the dCas9 transactivation system significantly increased LRIG1 protein abundance, reduced site-specific methylation, and reduced cancer cell viability. Our findings suggest that CRISPR-mediated targeted activation may be a feasible way to restore LRIG1 expression in cancer. CONCLUSIONS: Our study contributes novel insight into mechanisms which repress LRIG1 in triple-negative breast cancer and demonstrates for the first time that targeted de-repression of LRIG1 in cancer cells is possible. Understanding the epigenetic mechanisms associated with repression of tumour suppressor genes holds potential for the advancement of therapeutic approaches.


Asunto(s)
Metilación de ADN , Glicoproteínas de Membrana , Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , Islas de CpG/genética , Decitabina/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Neoplasias de la Mama Triple Negativas/genética
3.
BMC Complement Altern Med ; 16: 130, 2016 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-27193767

RESUMEN

BACKGROUND: Wilms' tumor 1 (WT1) is a biological marker for predicting leukemia progression. In this study, mammea E/BB, an active compound from Saraphi (Mammea siamensis) seed extract was examined for its effect on down-regulatory mechanism of WT1 gene expression, WT1 protein and mRNA stability, and cell proliferation in K562 cell line. METHODS: M. siamensis seeds were obtained from the region of Chiang Mai (North of Thailand). Mammea E/BB was extracted from seeds of M. siamensis. WT1 protein expression and stability were evaluated by Western blot analysis. WT1 mRNA stability was assessed by qRT-PCR. WT1-DNA binding and WT1 promoter activity were assayed by ChIP assay and luciferase-reporter assay, respectively. Cell cycle arrest was studied by flow cytometry. RESULTS: Treatment with mammea E/BB led to down-regulation of WT1 expression. The suppression of WT1 expression did not involve protein and mRNA degradation. Rather, WT1 protein was down-regulated through disruption of transcriptional auto-regulation of the WT1 gene. Mammea E/BB inhibited WT1-DNA binding at the WT1 promoter and decreased luciferase activity. It also disrupted c-Fos/AP-1 binding to the WT1 promoter via ERK1/2 signaling pathway and induced S phase cell cycle arrest in K562 cells. CONCLUSION: Mammea E/BB had pleotropic effects on kinase signaling pathways, resulting in inhibition of leukemia cell proliferation.


Asunto(s)
Cumarinas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Mammea/química , Extractos Vegetales/farmacología , Proteínas WT1/biosíntesis , Proliferación Celular/efectos de los fármacos , Cumarinas/química , Humanos , Células K562 , Estructura Molecular , Estabilidad del ARN/efectos de los fármacos , ARN Neoplásico , Proteínas WT1/genética
4.
BMC Genomics ; 15: 520, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24962896

RESUMEN

BACKGROUND: The ZNF217 gene, encoding a C2H2 zinc finger protein, is located at 20q13 and found amplified and overexpressed in greater than 20% of breast tumors. Current studies indicate ZNF217 drives tumorigenesis, yet the regulatory mechanisms of ZNF217 are largely unknown. Because ZNF217 associates with chromatin modifying enzymes, we postulate that ZNF217 functions to regulate specific gene signaling networks. Here, we present a large-scale functional genomic analysis of ZNF217, which provides insights into the regulatory role of ZNF217 in MCF7 breast cancer cells. RESULTS: ChIP-seq analysis reveals that the majority of ZNF217 binding sites are located at distal regulatory regions associated with the chromatin marks H3K27ac and H3K4me1. Analysis of ChIP-seq transcription factor binding sites shows clustering of ZNF217 with FOXA1, GATA3 and ERalpha binding sites, supported by the enrichment of corresponding motifs for the ERalpha-associated cis-regulatory sequences. ERalpha expression highly correlates with ZNF217 in lysates from breast tumors (n = 15), and ERalpha co-precipitates ZNF217 and its binding partner CtBP2 from nuclear extracts. Transcriptome profiling following ZNF217 depletion identifies differentially expressed genes co-bound by ZNF217 and ERalpha; gene ontology suggests a role for ZNF217-ERalpha in expression programs associated with ER+ breast cancer studies found in the Molecular Signature Database. Data-mining of expression data from breast cancer patients correlates ZNF217 with reduced overall survival. CONCLUSIONS: Our genome-wide ZNF217 data suggests a functional role for ZNF217 at ERalpha target genes. Future studies will investigate whether ZNF217 expression contributes to aberrant ERalpha regulatory events in ER+ breast cancer and hormone resistance.


Asunto(s)
Neoplasias de la Mama/metabolismo , Cromatina/metabolismo , Receptor alfa de Estrógeno/genética , Transactivadores/fisiología , Sitios de Unión , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Análisis por Conglomerados , Femenino , Factor de Transcripción GATA3/fisiología , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Genoma Humano , Factor Nuclear 3-alfa del Hepatocito/fisiología , Humanos , Estimación de Kaplan-Meier , Células MCF-7 , Unión Proteica , Transcriptoma
5.
Cancer Discov ; 2(7): 638-51, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22728437

RESUMEN

UNLABELLED: The transcription factor ZNF217 is a candidate oncogene in the amplicon on chromosome 20q13 that occurs in 20% to 30% of primary human breast cancers and that correlates with poor prognosis. We show that Znf217 overexpression drives aberrant differentiation and signaling events, promotes increased self-renewal capacity, mesenchymal marker expression, motility, and metastasis, and represses an adult tissue stem cell gene signature downregulated in cancers. By in silico screening, we identified candidate therapeutics that at low concentrations inhibit growth of cancer cells expressing high ZNF217. We show that the nucleoside analogue triciribine inhibits ZNF217-induced tumor growth and chemotherapy resistance and inhibits signaling events [e.g., phospho-AKT, phospho-mitogen-activated protein kinase (MAPK)] in vivo. Our data suggest that ZNF217 is a biomarker of poor prognosis and a therapeutic target in patients with breast cancer and that triciribine may be part of a personalized treatment strategy in patients overexpressing ZNF217. Because ZNF217 is amplified in numerous cancers, these results have implications for other cancers. SIGNIFICANCE: This study finds that ZNF217 is a poor prognostic indicator and therapeutic target in patients with breast cancer and may be a strong biomarker of triciribine treatment efficacy in patients. Because previous clinical trials for triciribine did not include biomarkers of treatment efficacy, this study provides a rationale for revisiting triciribine in the clinical setting as a therapy for patients with breast cancer who overexpress ZNF217.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Transactivadores/genética , Animales , Antibióticos Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Western Blotting , Neoplasias de la Mama/patología , Neoplasias de la Mama/prevención & control , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Células MCF-7 , Ratones , Células 3T3 NIH , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleósidos/farmacología , Análisis de Supervivencia , Transactivadores/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
FEBS Lett ; 585(14): 2235-42, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21658388

RESUMEN

The aim of this study was to investigate the inhibitory mechanism of pure curcumin on WT1 expression in leukemic K562 cells. Pure curcumin suppressed WT1 expression, independent of effects on protein degradation or WT1 mRNA stability. Chromatin immunoprecipitation and reporter gene assays indicate that pure curcumin treatment attenuates WT1 auto-regulation. Interestingly, PKCα inhibition mimicks the repressive effects of pure curcumin in K562 cells. Conversely, myristoylated PKCα over-expression increased WT1 expression and reversed the inhibitory effect of pure curcumin. Our study indicates that pure curcumin attenuates WT1 auto-regulatory function through inhibition of PKCα signaling in K562 cells.


Asunto(s)
Antineoplásicos/farmacología , Curcumina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células K562 , Proteína Quinasa C-alfa/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas WT1/genética , Genes Reporteros , Humanos , Células K562/efectos de los fármacos , Células K562/fisiología , Regiones Promotoras Genéticas , Proteínas WT1/metabolismo
7.
Spine (Phila Pa 1976) ; 28(10): 1027-35; discussion 1035, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12768144

RESUMEN

STUDY DESIGN: A prospective study of spondylolysis and spondylolisthesis was initiated in 1955 with a radiographic and clinical study of 500 first-grade children. OBJECTIVE: To determine the natural history of spondylolysis and spondylolisthesis. SUMMARY OF BACKGROUND DATA: Most studies on the natural history of spondylolysis and spondylolisthesis are based on patient populations presenting with pain. Critical to any natural history investigation is the study of a population of affected individuals, whether symptomatic or not, from onset of the condition through their lives. METHODS: By study of a population from the age of 6 years to adulthood, 30 individuals were identified to have pars lesions. Data collection at a 45-year follow-up assessment included magnetic resonance imaging, a back pain questionnaire, and the SF-36 Survey. RESULTS: No subject with a pars defect was lost to follow-up evaluation once a lesion was identified. Subjects with unilateral defects never experienced slippage over the course of the study. Progression of spondylolisthesis slowed with each decade. There was no association of slip progression and low back pain. There was no statistically significant difference between the study population SF-36 scores and those of the general population the same age. CONCLUSIONS: This report is the only prospective study to document the natural history of spondylolysis and spondylolisthesis from onset through more than 45 years of life in a population unselected for pain. Subjects with pars defects follow a clinical course similar to that of the general population. There appears to be a marked slowing of slip progression with each decade, and no subject has reached a 40% slip.


Asunto(s)
Espondilolistesis/patología , Espondilólisis/patología , Adolescente , Adulto , Niño , Progresión de la Enfermedad , Estudios de Seguimiento , Humanos , Disco Intervertebral/patología , Vértebras Lumbares/patología , Imagen por Resonancia Magnética , Encuestas y Cuestionarios , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA