Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Biochem Biophys Res Commun ; 594: 109-116, 2022 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-35081499

RESUMEN

HIC1 and RassF1A methylation, which cause loss of gene function, are found in various cancers, including renal cell carcinoma (RCC), and could alter cell stiffness and the content of extracellular vesicles (EVs). These physiological changes may provide a tumoral survival advantage and thus could serve as cellular biomarkers for monitoring cell transformation, although direct associations between these changes and cell transformation remain to be established. As we found HIC1 and RassF1A methylation and expression changes in RCC samples, we examined the effects of gain and loss of HIC1 and RassF1A expression on cell DNA content, cytoskeletal structure, and Piwi-interacting RNA (piRNA) expression in EVs. We found HIC1 and RassF1A hypermethylation and abnormal expression in RCC patient samples was independent of the somatic mutations found in publicly available data. Cell stiffness was reduced in accordance with disrupted cytoskeleton conformation after knockdown of HIC1 or RassF1A. Gain or loss of HIC1 expression induced instability in genomic content, abnormal RassF1A expression disturbed cytoskeletal structure, and the abnormal expression of either gene altered piRNA content in EVs. These results suggest a causal relationship between abnormal tumor suppressor gene expression, cell stiffness, and piRNA expression.


Asunto(s)
Citoesqueleto/metabolismo , Exosomas , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Biomarcadores , Carcinoma de Células Renales/metabolismo , Transformación Celular Neoplásica , ADN/metabolismo , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Genoma Humano , Humanos , Técnicas In Vitro , Neoplasias Renales/metabolismo , Células Madre Mesenquimatosas/citología , Microscopía de Fuerza Atómica , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor/genética
2.
Int J Mol Sci ; 19(10)2018 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-30249017

RESUMEN

Cell stiffness is a potential biomarker for monitoring cellular transformation, metastasis, and drug resistance development. Environmental factors relayed into the cell may result in formation of inheritable markers (e.g., DNA methylation), which provide selectable advantages (e.g., tumor development-favoring changes in cell stiffness). We previously demonstrated that targeted methylation of two tumor suppressor genes, hypermethylated in cancer 1 (HIC1) and Ras-association domain family member 1A (RassF1A), transformed mesenchymal stem cells (MSCs). Here, transformation-associated cytoskeleton and cell stiffness changes were evaluated. Atomic force microscopy (AFM) was used to detect cell stiffness, and immunostaining was used to measure cytoskeleton expression and distribution in cultured cells as well as in vivo. HIC1 and RassF1A methylation (me_HR)-transformed MSCs developed into tumors that clonally expanded in vivo. In me_HR-transformed MSCs, cell stiffness was lost, tubulin expression decreased, and F-actin was disorganized; DNA methylation inhibitor treatment suppressed their tumor progression, but did not fully restore their F-actin organization and stiffness. Thus, me_HR-induced cell transformation was accompanied by the loss of cellular stiffness, suggesting that somatic epigenetic changes provide inheritable selection markers during tumor propagation, but inhibition of oncogenic aberrant DNA methylation cannot restore cellular stiffness fully. Therefore, cell stiffness is a candidate biomarker for cells' physiological status.


Asunto(s)
Metilación de ADN , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Células Madre Mesenquimatosas/patología , Tubulina (Proteína)/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Apoptosis , Biomarcadores de Tumor , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Neoplasias Hepáticas/genética , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Pronóstico , Regiones Promotoras Genéticas , Estrés Mecánico , Tubulina (Proteína)/genética , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Biochem Biophys Res Commun ; 494(3-4): 470-476, 2017 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-29066347

RESUMEN

Negative valine (V) to phenylalanine (F) switch at the Janus kinase (JAK2) 617 codon (V617F) is the dominant driver mutation in patients with myeloproliferative neoplasms (MPNs). JAK2V617F was proved to be sufficient for cell transformation; however, independent mutations might influence the following epigenomic modifications. To assess the JAK2V617F-induced downstream epigenomic changes without interferences, we profiled the epigenomic changes in ectopically expressed JAK2V617F in Ba/F3 cells. Antibodies against phosphorylated signal transducer and activator of transcription 3 (pSTAT3) and enhancer of zeste homolog 2 (EZH2) were used for chromatin-immunoprecipitation sequencing (ChIP-seq) to detect the downstream epigenomic targets in the JAK2-STAT3 signaling pathway. To confirm the JAK2V617F-induced epigenetic changes in vivo, DNA methylation changes in the target loci in patients with MPNs were detected through methylation-specific polymerase chain reaction and were clustered against the changes within controls. We found that ectopically expressed JAK2V617F in Ba/F3 cells reduced the binding specificity; it was associated with cis-regulatory elements and recognized DNA motifs in both pSTAT3-downstream and EZH2-associated targets. Overlapping target loci between the control and JAK2V617F were <3% and 0.4%, respectively, as identified through pSTAT3 and EZH2 ChIP-seq. Furthermore, the methylation changes in the direct target loci (FOXH1, HOXC9, and SRF) were clustered independently from the control locus (L1TD1) and other mutation genes (HMGA2 and Lin28A) in the analyzed MPN samples. Therefore, JAK2V617F influences target binding in both pSTAT3 and EZH2. Without mutations in epigenetic regulators, JAK2V617F can induce downstream epigenomic modifications. Thus, epigenetic changes in JAK2 downstream targets might be trackable in vivo.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , Epigénesis Genética/genética , Neoplasias Hematológicas/genética , Janus Quinasa 2/genética , Trastornos Mieloproliferativos/genética , Factor de Transcripción STAT3/genética , Animales , Línea Celular Tumoral , Epigenómica/métodos , Ratones
4.
Ann Surg Oncol ; 24(2): 578-585, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26671036

RESUMEN

BACKGROUND: Methylation changes within tumor suppressor (TS) genes or polycomb group target (PcG) genes alter cell fates. Chromatin associated with PcG targets is bivalent in stem cells, while TS genes are not normally bivalent. PcG target methylation changes have been identified in tumor stem cells, and abnormal methylation is found in TS genes in cancers. If the epigenetic states of genes influence DNA methylation, then methylation of PcG targets and TS genes may evolve differently during cancer development. More importantly, methylation changes may be part of a sequence in tumorigenesis. METHODS: Chromatin and methylation states of 4 PcG targets and 2 TS genes were determined in colon cancer cells. The methylation states were also detected in 100 pairs of colon cancer samples. Principle component analysis (PCA) was used to reveal whether TS methylation or PcG methylation was the main methylation change associated with colon cancers. RESULTS: Chromatin and methylation states differ in colon cancer cell lines. The methylation states within PcG targets clustered independently from the methylation states in TS genes, a finding we previously reported in liver cancers. PCA in colon cancers revealed the strongest association with methylation changes in 2 TS genes, HIC1 and RassF1A. Loss of HIC1 methylation correlated with decreased tumor migration. CONCLUSIONS: PcG and TS methylation states cluster independently from each other. The deduced principle component correlated better with TS methylation than PcG methylation in colon cancer. Abnormal methylation changes may represent a sequential biomarker profile to identify developing colon cancer.


Asunto(s)
Neoplasias del Colon/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Proteínas del Grupo Polycomb/genética , Proteínas Supresoras de Tumor/genética , Movimiento Celular , Neoplasias del Colon/patología , Epigénesis Genética , Genes Supresores de Tumor , Humanos , Células Tumorales Cultivadas
5.
Cancer Cell Int ; 15: 98, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26464562

RESUMEN

BACKGROUND AND PROPOSE: Changes in DNA methylation are associated with changes in somatic cell fate without the alteration of coding sequences. In addition to its use as a traceable biomarker, reversible DNA methylation could also serve as a therapeutic target. In particular, if the development of drug resistance is associated with changes in DNA methylation, then demethylation might reverse the resistance phenotype. The reversion of the drug-resistance might then be feasible if the association between abnormal DNA methylation and the development of drug-resistance could be identified. METHODS: Methylation differences between the drug-resistance cervical cancer cell, SiHa, and its derived oxaliplatin-resistant S3 cells were detected by methylation specific microarray. The drug-resistance cells were treated with demethylation agent to see if the resistance phenotype were reversed. Targeted methylation of one of the identified locus in normal cell is expected to recapitulate the development of resistance and a two-component reporter system is adopted to monitor the increase of DNA methylation in live cells. RESULTS: In this report, we identified methylation changes, both genome-wide and within individual loci, in the oxaliplatin-resistant cervical cancer cell S3 compared with its parental cell line SiHa. Treatment of S3 with a demethylation agent reversed increases in methylation and allowed the expression of methylation-silenced genes. Treatment with the demethylation agent also restored the sensitivity of S3 to cisplatin, taxol, and oxaliplatin to the same level as that of SiHa. Finally, we found that methylation of the target gene Casp8AP2 is sufficient to increase drug resistance in different cells. CONCLUSIONS: These results suggest that global methylation is associated with the development of drug resistance and could serve as a biomarker and therapeutic target for drug resistance in cervical cancer.

6.
Biochem Biophys Res Commun ; 453(3): 338-44, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25264105

RESUMEN

Fetal alcohol syndrome (FAS) is a birth defect due to maternal alcohol consumption during pregnancy. Because mesenchymal stem cells (MSCs) are the main somatic stem cells in adults and may contribute to tissue homeostasis and repair in adulthood, we investigated whether early life ethanol exposure affects MSCs and contributes to the propensity for disease onset in later life. Using a rodent model of FAS, we found that ethanol exposure (5.25g/kg/day) from postnatal days 4 to 9 in rat pups (mimic of human third trimester) caused long-term anomalies in bone marrow-derived MSCs. MSCs isolated from ethanol-exposed animals were prone to neural induction but resistant to osteogenic and adipogenic inductions compared to their age-matched controls. The altered differentiation may contribute to the severe trabecular bone loss seen in ethanol-exposed animals at 3months of age as well as overt growth retardation. Expression of alkaline phosphatase, osteocalcin, aP2, and PPARγ were substantially inhibited, but BDNF was up-regulated in MSCs isolated from ethanol-exposed 3month-old animals. Several signaling pathways were distorted in ethanol-exposed MSCs via altered trimethylation at histone 3 lysine 27. These results demonstrate that early life ethanol exposure can have long-term impacts in rat MSCs by both genetic and epigenetic mechanisms.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Etanol/toxicidad , Células Madre Mesenquimatosas/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Animales , Secuencia de Bases , Cartilla de ADN , Modelos Animales de Enfermedad , Femenino , Trastornos del Espectro Alcohólico Fetal/etiología , Trastornos del Espectro Alcohólico Fetal/genética , Masculino , Reacción en Cadena de la Polimerasa , Embarazo , Ratas
7.
Biochem Biophys Res Commun ; 442(1-2): 56-61, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24211627

RESUMEN

Endosulfine alpha (ENSA) is an endogenous ligand of sulfonylurea receptor that was reported to be associated with an ATP-dependent potassium channel that controls insulin release and the onset of type 2 diabetes. ENSA also interacts with microtubule-associated serine/threonine-protein kinase-like (MASTL) to regulate the cell cycle. Previously, we identified ENSA as a possible bivalent gene in mesenchymal stem cells (MSCs) and hypothesized its methylation might determine cellular differentiation and transformation. Because there was no link between aberrant ENSA expression and tumorigenesis, we aimed to determine if ENSA is abnormally regulated in liver cancer and plays a role in liver cancer propagation. The epigenetic states of the ENSA promoter were evaluated in different cancer cell lines and patient samples. ENSA was overexpressed in a liver cancer cell line, and its interaction with MASTL and possible tumor suppression capabilities were also determined in cultured cells and mice. Distinct ENSA promoter methylation was observed in liver cancer (n=100 pairs) and breast cancer (n=100 pairs). ENSA was predominantly hypomethylated in liver cancer but was hypermethylated in breast cancer. Overexpressed ENSA interacts with MASTL and suppresses hepatic tumor growth. We also found that ENSA is hypermethylated in CD90-expressing (CD90(+)) cells compared to CD90 non-expressing (CD90(-)) liver cancer cells. These data reveal ENSA methylation changes during hepatic tumor evolution. Overexpressed ENSA suppresses tumor growth in an established hepatic cell line whereas hypermethylated ENSA might help maintain liver cancer initiating cells.


Asunto(s)
Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Péptidos/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Invasividad Neoplásica , Péptidos/metabolismo , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo
8.
J Pathol ; 230(3): 298-309, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23362108

RESUMEN

The strong associations between oral squamous cell carcinoma (OSCC) and dietary habits such as alcohol consumption (A), betel quid chewing (B) and cigarette smoking (C) and its predominance in men have been well documented; however, systemic analysis of OSCC is limited. Our study applied high-throughput screening methods to identify causative epigenetic targets in a cohort of men with ABC-associated OSCC. We identified BEX1 and LDOC1 as two epigenetically silenced X-linked tumour suppressors and demonstrated a functional link between the transcription of BEX1 and LDOC1 and promoter hypermethylation. Methylation of the BEX1 and LDOC1 promoters was associated significantly (p < 0.0001) with OSCC and were detected in 75% (42/56) and 89% (50/56) of the samples, respectively. We observed concordant increases in the methylation of both genes in 71% (40/56) of the tumours, and potent in vitro and in vivo growth inhibitory effects in OSCC cells ectopically expressing BEX1 and/or LDOC1. Restored expression of BEX1 and LDOC1 suppressed the nuclear factor-κB (NF-κB) signalling pathway, which is the most frequently hyperactivated signalling pathway in OSCC. This suppression might result from decreased p50 and p65 expression. These findings suggest that silencing of BEX1 and LDOC1 by promoter hypermethylation might represent a critical event in the molecular pathogenesis of OSCC and account for the oncogenic effects of ABC exposure and the male predominance of OSCC occurrence. Microarray data are available in the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo/)


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Boca/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética , Animales , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular , Estudios de Cohortes , Metilación de ADN , Regulación hacia Abajo , Epigénesis Genética , Silenciador del Gen , Genes Ligados a X , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones SCID , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Distribución Aleatoria , Factores Sexuales , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo
9.
Adv Exp Med Biol ; 754: 195-211, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22956503

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent stem cells of mesodermal origin that can be isolated from various sources and induced into different cell types. Although MSCs possess immune privilege and are more easily obtained than embryonic stem cells, their propensity to tumorigenesis has not been fully explored. Epigenomic changes in DNA methylation and chromatin structure have been hypothesized to be critical in the determination of lineage-specific differentiation and tumorigenesis of MSCs, but this has not been formally proven. We applied a targeted DNA methylation method to methylate a Polycomb group protein-governed gene, Trip10, in MSCs, which accelerated the cell fate determination of MSCs. In addition, targeted methylation of HIC1 and RassF1A, both tumor suppressor genes, transformed MSCs into tumor stem cell-like cells. This new method will allow better control of the differentiation of MSCs and their use in downstream applications.


Asunto(s)
Epigénesis Genética , Células Madre Mesenquimatosas/patología , Neoplasias/genética , Neoplasias/patología , Animales , Humanos
10.
Biochem Biophys Res Commun ; 425(2): 290-6, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22842566

RESUMEN

Polycomb-group proteins mark specific chromatin conformations in embryonic and somatic stem cells that are critical for maintenance of their "stemness". These proteins also mark altered chromatin modifications identified in various cancers. In normal differentiated cells or advanced cancerous cells, these polycomb-associated loci are frequently associated with increased DNA methylation. It has thus been hypothesized that changes in DNA methylation status within polycomb-associated loci may dictate cell fate and that abnormal methylation within these loci may be associated with tumor development. To assess this, we examined the methylation states of four polycomb target loci -Trip10, Casp8AP2, ENSA, and ZNF484 - in liver cancer. These four targets were selected because their methylation levels are increased during mesenchymal stem cell-to-liver differentiation. We found that these four loci were hypomethylated in most early-stage liver cancer specimens. For comparison, two non-polycomb tumor suppressor genes, HIC1 and RassF1A, were also examined. Whereas the methylation level of HIC1 did not differ significantly between normal and tumor samples, RassF1A was significantly hypermethylated in liver tumor samples. Unsupervised clustering analysis classified the methylation changes within polycomb and non-polycomb targets to be independent, indicating independent epigenetic evolution. Thus, pre-deposited polycomb marks within somatic stem cells may contribute to the determination of methylation changes during hepatic tumorigenesis.


Asunto(s)
Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Neoplasias Hepáticas/genética , Proteínas del Grupo Polycomb/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Línea Celular Tumoral , Cromatina/metabolismo , Sitios Genéticos/genética , Células Hep G2 , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Asociadas a Microtúbulos/genética , Antígenos de Histocompatibilidad Menor
11.
Biochem Biophys Res Commun ; 422(4): 578-85, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22595458

RESUMEN

Casp8AP2 contains a FLASH functional domain and is critical for the formation of death complex and the relay of death signal into the cells. Genetic defects in Casp8AP2 are associated with several diseases. A CpG island within the Casp8AP2 promoter is differentially regulated during somatic stem cell differentiation, and aberrant DNA methylation within the Casp8AP2 promoter has been reported in cancers. We hypothesized that abnormal DNA methylation of Casp8AP2 promoter might contribute to prolonged cellular survival or drug resistance in cancer. The epigenetic state within the Casp8AP2 promoter was then determined in different cancer cell lines and patient samples by methylation-specific PCR. Targeted Casp8AP2 methylation within normal and tumor cells was performed to see whether methylation promoted drug resistance. We found differential Casp8AP2 methylation among the normal and tumoral samples. Global demethylation in a platinum drug-resistant human gastric cancer cell line reversed Casp8AP2 methylation and diminished drug resistance. Targeted methylation of the Casp8AP2 promoter in somatic stem cells and cancer cells increased their resistance to drugs including platinum drugs. These data demonstrate that methylation within the Casp8AP2 promoter correlates with the development of drug resistance and might serve as a biomarker and treatment target for drug resistance in cancer cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Metilación de ADN , Resistencia a Antineoplásicos/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Línea Celular , Línea Celular Tumoral , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Regiones Promotoras Genéticas
12.
Cancer Res ; 71(13): 4653-63, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21518779

RESUMEN

Although DNA hypermethylation within promoter CpG islands is highly correlated with tumorigenesis, it has not been established whether DNA hypermethylation within a specific tumor suppressor gene (TSG) is sufficient to fully transform a somatic stem cell. In this study, we addressed this question using a novel targeted DNA methylation technique to methylate the promoters of HIC1 and RassF1A, two well-established TSGs, along with a two-component reporter system to visualize successful targeting of human bone marrow-derived mesenchymal stem cells (MSC) as a model cell system. MSCs harboring targeted promoter methylations of HIC1/RassF1A displayed several features of cancer stem/initiating cells including loss of anchorage dependence, increased colony formation capability, drug resistance, and pluripotency. Notably, inoculation of immunodeficient mice with low numbers of targeted MSC resulted in tumor formation, and subsequent serial xenotransplantation and immunohistochemistry confirmed the presence of stem cell markers and MSC lineage in tumor xenografts. Consistent with the expected mechanism of TSG hypermethylation, treatment of the targeted MSC with a DNA methyltransferase inhibitor reversed their tumorigenic phenotype. To our knowledge, this is the first direct demonstration that aberrant TSG hypermethylation is sufficient to transform a somatic stem cell into a fully malignant cell with cancer stem/initiating properties.


Asunto(s)
Transformación Celular Neoplásica/genética , Metilación de ADN , Genes Supresores de Tumor , Células Madre Mesenquimatosas/fisiología , Células Madre Neoplásicas/fisiología , Animales , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/patología , Clonación Molecular , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Células Madre Mesenquimatosas/patología , Ratones , Ratones Desnudos , Células Madre Neoplásicas/patología , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Trasplante Heterólogo , Proteínas Supresoras de Tumor/genética
13.
J Biomed Sci ; 18: 12, 2011 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-21299869

RESUMEN

BACKGROUND: The Cdc42-interacting protein-4, Trip10 (also known as CIP4), is a multi-domain adaptor protein involved in diverse cellular processes, which functions in a tissue-specific and cell lineage-specific manner. We previously found that Trip10 is highly expressed in estrogen receptor-expressing (ER+) breast cancer cells. Estrogen receptor depletion reduced Trip10 expression by progressively increasing DNA methylation. We hypothesized that Trip10 functions as a tumor suppressor and may be involved in the malignancy of ER-negative (ER-) breast cancer. To test this hypothesis and evaluate whether Trip10 is epigenetically regulated by DNA methylation in other cancers, we evaluated DNA methylation of Trip10 in liver cancer, brain tumor, ovarian cancer, and breast cancer. METHODS: We applied methylation-specific polymerase chain reaction and bisulfite sequencing to determine the DNA methylation of Trip10 in various cancer cell lines and tumor specimens. We also overexpressed Trip10 to observe its effect on colony formation and in vivo tumorigenesis. RESULTS: We found that Trip10 is hypermethylated in brain tumor and breast cancer, but hypomethylated in liver cancer. Overexpressed Trip10 was associated with endogenous Cdc42 and huntingtin in IMR-32 brain tumor cells and CP70 ovarian cancer cells. However, overexpression of Trip10 promoted colony formation in IMR-32 cells and tumorigenesis in mice inoculated with IMR-32 cells, whereas overexpressed Trip10 substantially suppressed colony formation in CP70 cells and tumorigenesis in mice inoculated with CP70 cells. CONCLUSIONS: Trip10 regulates cancer cell growth and death in a cancer type-specific manner. Differential DNA methylation of Trip10 can either promote cell survival or cell death in a cell type-dependent manner.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas de Neoplasias/biosíntesis , Neoplasias/metabolismo , Animales , Muerte Celular/genética , Supervivencia Celular , Metilación de ADN/genética , Femenino , Células Hep G2 , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Antígenos de Histocompatibilidad Menor , Proteínas de Neoplasias/genética , Neoplasias/patología
14.
J Biomed Sci ; 18: 3, 2011 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-21219604

RESUMEN

BACKGROUND: Targeting abnormal DNA methylation represents a therapeutically relevant strategy for cancer treatment as demonstrated by the US Food and Drug Administration approval of the DNA methyltransferase inhibitors azacytidine and 5-aza-2'-deoxycytidine for the treatment of myelodysplastic syndromes. But their use is associated with increased incidences of bone marrow suppression. Alternatively, procainamide has emerged as a potential DNA demethylating agent for clinical translation. While procainamide is much safer than 5-aza-2'-deoxycytidine, it requires high concentrations to be effective in DNA demethylation in suppressing cancer cell growth. Thus, our laboratories have embarked on the pharmacological exploitation of procainamide to develop potent DNA methylation inhibitors through lead optimization. METHODS: We report the use of a DNA methylation two-component enhanced green fluorescent protein reporter system as a screening platform to identify novel DNA methylation inhibitors from a compound library containing procainamide derivatives. RESULTS: A lead agent IM25, which exhibits substantially higher potency in GSTp1 DNA demethylation with lower cytotoxicity in MCF7 cells relative to procainamide and 5-aza-2'-deoxycytidine, was identified by the screening platform. CONCLUSIONS: Our data provide a proof-of-concept that procainamide could be pharmacologically exploited to develop novel DNA methylation inhibitors, of which the translational potential in cancer therapy/prevention is currently under investigation.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Metilación de ADN/efectos de los fármacos , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/metabolismo , Procainamida/análogos & derivados , Procainamida/farmacología , Antiarrítmicos/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos
15.
Biochem Biophys Res Commun ; 402(2): 228-34, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20933502

RESUMEN

DNA methylation is a gene-silencing and host defense system that can down-regulate viral gene expression in mammalian cells. An established targeted DNA methylation method was used to demonstrate that genome-integrated CMV and adenovirus type 5 E1A promoters were hypermethylated after MCF7 and HEK293 cells were transfected with in vitro methylated viral promoter fragments. In both cases, the targeted methylation-induced gene silencing could be reversed by addition of 5-aza-2'-deoxycytidine, confirming that the CMV and E1A promoters are regulated by DNA methylation. The kinetics of the targeted DNA methylation was determined using a reporter system in live cells. In conclusion, targeted DNA methylation is able to efficiently silence susceptible viral promoters and provides an alternative strategy to study the impact of loci-specific DNA methylation in viral gene expression.


Asunto(s)
Proteínas E1A de Adenovirus/genética , Citomegalovirus/genética , Metilación de ADN , Regulación Viral de la Expresión Génica , Silenciador del Gen , Línea Celular , Humanos , Regiones Promotoras Genéticas
16.
Biochem Biophys Res Commun ; 400(3): 305-12, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20727853

RESUMEN

Epigenetic regulation of gene expression by DNA methylation and histone modification controls cell fate during development and homeostasis in adulthood. Aberrant epigenetic modifications may lead to abnormal development, even diseases. We have found that Trip10 (thyroid hormone receptor interactor 10), an adaptor protein involved in diverse functions, is epigenetically regulated during lineage-specific induction of human bone marrow-derived mesenchymal stem cells (MSCs). To determine whether DNA methylation-induced gene silencing is sufficient to restrict cell fate changes, we applied an invitro method to specifically methylate the promoter of Trip10. Our hypothesis was that the methylation status of the Trip10 promoter in MSCs alters the differentiation preference of MSCs. Transfection of in vitro-methylated Trip10 promoter DNA into MSCs resulted in progressive accumulation of cytosine methylation at the endogenous Trip10 promoter, reduced Trip10 expression, and accelerated MSC-to-neuron and MSC-to-osteocyte differentiation. A two-component EGFP reporter gene system was established to confirm the level of transcriptional silencing and visualize the targeted DNA methylation. EGFP expression induced in the reporter system by targeted Trip10 methylation was reversed by adding 5-aza-2'-deoxycytidine, a DNA methyltransferase inhibitor, confirming that the suppressed Trip10 expression and disrupted MSC differentiation resulted from the in vitro-introduced methylations in the Trip10 promoter. With this targeted DNA methylation and reporter system, we are able to monitor the progression of locus-specific DNA methylation in vivo and correlate such changes with potential functional changes. Using this approach, we have established a new role for Trip10, showing that the level of Trip10 expression is associated with the maintenance and differentiation of MSCs.


Asunto(s)
Linaje de la Célula/genética , Metilación de ADN , Silenciador del Gen , Células Madre Mesenquimatosas/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Proteínas Asociadas a Microtúbulos/genética , Antígenos de Histocompatibilidad Menor , Regiones Promotoras Genéticas , Ratas
17.
Lab Invest ; 90(3): 414-25, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20065949

RESUMEN

Resistance to TGF-beta is frequently observed in ovarian cancer, and disrupted TGF-beta/SMAD4 signaling results in the aberrant expression of downstream target genes in the disease. Our previous study showed that ADAM19, a SMAD4 target gene, is downregulated through epigenetic mechanisms in ovarian cancer with aberrant TGF-beta/SMAD4 signaling. In this study, we investigated the mechanism of downregulation of FBXO32, another SMAD4 target gene, and the clinical significance of the loss of FBXO32 expression in ovarian cancer. Expression of FBXO32 was observed in the normal ovarian surface epithelium, but not in ovarian cancer cell lines. FBXO32 methylation was observed in ovarian cancer cell lines displaying constitutive TGF-beta/SMAD4 signaling, and epigenetic drug treatment restored FBXO32 expression in ovarian cancer cell lines regardless of FBXO32 methylation status, suggesting that epigenetic regulation of this gene in ovarian cancer may be a common event. In advanced-stage ovarian tumors, a significant (29.3%; P<0.05) methylation frequency of FBXO32 was observed and the association between FBXO32 methylation and shorter progression-free survival was significant, as determined by both Kaplan-Meier analysis (P<0.05) and multivariate Cox regression analysis (hazard ratio: 1.003, P<0.05). Reexpression of FBXO32 markedly reduced proliferation of a platinum-resistant ovarian cancer cell line both in vitro and in vivo, due to increased apoptosis of the cells, and resensitized ovarian cancer cells to cisplatin. In conclusion, the novel tumor suppressor FBXO32 is epigenetically silenced in ovarian cancer cell lines with disrupted TGF-beta/SMAD4 signaling, and FBXO32 methylation status predicts survival in patients with ovarian cancer.


Asunto(s)
Apoptosis , Metilación de ADN , Proteínas Musculares/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Cisplatino/farmacología , Decitabina , Regulación hacia Abajo , Resistencia a Antineoplásicos , Epigénesis Genética/efectos de los fármacos , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Proteínas Musculares/genética , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/mortalidad , Pronóstico , Regiones Promotoras Genéticas , Modelos de Riesgos Proporcionales , Proteínas Ligasas SKP Cullina F-box/genética , Proteína Smad4/metabolismo , Taiwán/epidemiología , Factor de Crecimiento Transformador beta/metabolismo , Adulto Joven
18.
Semin Cancer Biol ; 19(3): 198-208, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19429484

RESUMEN

Epigenetic events like DNA methylation are known to regulate gene expression, and dysregulation of these events is associated with neoplastic proliferation. Here, we provide a step-by-step review of the approach that has gradually developed to identify critical DNA methylation during neoplasia. DNA methylation has first been tightly linked to the regulation of gene expression and functions. Next, the clinical importance of such DNA methylation has been probed by inducing loss of the maintenance of normal DNA methylation, which has been found to trigger onset of disease. Methylation changes can be signal-specific and lineage-specific, providing a record what cells have encountered and what they have become. Comparison of methylation associated with normal cellular differentiation and abnormal cell fate changes is expected to uncover critical methylation changes. We also propose a specific scheme that can be used to excavate critical DNA methylation associated with cell evolution.


Asunto(s)
Transformación Celular Neoplásica/genética , Metilación de ADN , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Animales , Genes Supresores de Tumor/fisiología , Histona Desacetilasas/metabolismo , Humanos , Neoplasias/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología
19.
Life Sci ; 78(13): 1483-93, 2006 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-16324718

RESUMEN

In the United State, 20% of pregnant women smoke. One of the most consistent adverse outcomes is reduced birth weight in the off-spring. Animal studies using chronic nicotine, the major psychoactive tobacco ingredient, have shown conflicting results, questioning the role of nicotine in growth retardation. To evaluate the direct effects of nicotine during a period equivalent to the human third trimester, we developed an oral gastric intubation model using neonatal rat pups. Nicotine (6 mg/kg/day) was dissolve in milk-formula and delivered during three feedings daily from postnatal day (P)1 to P7. Nicotine immediately and significantly [P<0.05] decreased weight gain per day (WGD) by 13.5% (+/-) 1 day after onset of treatment in both genders and throughout the treatment period. This resulted in significantly lower body weight at P4 and P5 in male and female pups, respectively. After nicotine withdrawal, WGD returned to control level within 1 day, whereas total body weight recovered by P18. There were no long-term consequences on body weight or growth pattern in either gender. The nicotinic acetylcholine receptor (nAChR) antagonist dihydro-beta-erythroidine (DHbetaE) reversed nicotine's effects on WGD suggesting an involvement of heteromeric alpha4beta2, whereas methyllycaconitine (MLA) an antagonist for the homomeric alpha7-type receptor was ineffective. The immediate decrease of growth in neonatal pups suggests that nicotine's effect on birth weight results from direct anorexic rather then indirect effects due to placental dysfunction or increased fetal hypoxia. The postnatal oral gastric intubation model seems to accurately reflect the direct effects of nicotine in neonates.


Asunto(s)
Trastornos del Crecimiento/inducido químicamente , Nicotina/toxicidad , Animales , Animales Recién Nacidos , Peso Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Modelos Animales de Enfermedad , Femenino , Masculino , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales , Aumento de Peso/efectos de los fármacos
20.
Brain Res ; 1008(1): 69-80, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15081384

RESUMEN

Six-day 'binge' ethanol intoxication postnatal days (PD) 4-9 delays up-regulation of gamma-aminobutyric acid type A receptors (GABAARs) in developing rat septal neurons [Dev. Brain Res. 130 (2001) 25]. This distortion occurs during synaptogenesis and could contribute to cognitive dysfunction in fetal alcohol syndrome (FAS). Here, we asked two questions concerning requirements for vulnerability to GABAAR blunting by ethanol. First, we asked whether receptor blunting required PD 4-9 ethanol exposure in rat pups and found that just a brief 2-day exposure (PD 8-9) was as effective as all 6 days. However, 2-day exposure on PD 4-5 was ineffective, showing that 'binge' timing was important. We also asked whether 'binge' exposure directly inhibited intrinsic processes of septal neurons and could blunt GABAARs on cells maturing outside the brain. Embryonic septal neurons grown in serum-free dispersed culture developed extensive dendritic arborizations, spontaneous synaptic activity and robust whole-cell GABAAR function, but surprisingly, did not show developmental up-regulation of GABAARs like septal neurons maturing in vivo [Brain Res. 810 (1998) 100]. Furthermore, age-matched 6-day 'binge' ethanol exposure did not blunt GABAAR function in septal neurons in vitro. These results suggest developmental mechanisms driving up-regulation of GABAAR function in septal neurons in vivo briefly becomes vulnerable to ethanol insult in early postnatal life. While septal neurons express comparable functional GABAARs whether maturing in vivo or in vitro, vulnerability to ethanol-induced receptor blunting requires elements of an intact brain environment not replicated in culture.


Asunto(s)
Depresores del Sistema Nervioso Central/administración & dosificación , Etanol/administración & dosificación , Neuronas/efectos de los fármacos , Receptores de GABA-A/fisiología , Tabique del Cerebro/citología , Análisis de Varianza , Animales , Animales Recién Nacidos , Anticonvulsivantes/farmacología , Bicuculina/farmacología , Peso Corporal/efectos de los fármacos , Células Cultivadas , Depresores del Sistema Nervioso Central/farmacología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Interacciones Farmacológicas , Embrión de Mamíferos , Etanol/sangre , Etanol/farmacología , Femenino , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Moduladores del GABA/farmacología , Técnicas In Vitro , Masculino , Potenciales de la Membrana/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp/métodos , Penicilinas/farmacología , Picrotoxina/farmacología , Embarazo , Inhibidores de la Síntesis de la Proteína/farmacología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/efectos de los fármacos , Estreptomicina/farmacología , Factores de Tiempo , Triazoles/farmacología , Regulación hacia Arriba/efectos de los fármacos , Zinc/farmacología , Zolpidem , Ácido gamma-Aminobutírico/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA