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.
Artigo em Inglês | MEDLINE | ID: mdl-35081499

RESUMO

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.


Assuntos
Citoesqueleto/metabolismo , Exossomos , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Biomarcadores , Carcinoma de Células Renais/metabolismo , Transformação Celular Neoplásica , DNA/metabolismo , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Genoma Humano , Humanos , Técnicas In Vitro , Neoplasias Renais/metabolismo , Células-Tronco Mesenquimais/citologia , Microscopia de Força Atômica , Regiões Promotoras Genéticas , Proteínas Supressoras de Tumor/genética
2.
Int J Mol Sci ; 19(10)2018 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-30249017

RESUMO

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.


Assuntos
Metilação de DNA , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Células-Tronco Mesenquimais/patologia , Tubulina (Proteína)/metabolismo , Proteínas Supressoras de Tumor/genética , Animais , Apoptose , Biomarcadores Tumorais , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Neoplasias Hepáticas/genética , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Prognóstico , Regiões Promotoras Genéticas , Estresse Mecânico , Tubulina (Proteína)/genética , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Biochem Biophys Res Commun ; 494(3-4): 470-476, 2017 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-29066347

RESUMO

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.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética/genética , Neoplasias Hematológicas/genética , Janus Quinase 2/genética , Transtornos Mieloproliferativos/genética , Fator de Transcrição STAT3/genética , Animais , Linhagem Celular Tumoral , Epigenômica/métodos , Camundongos
4.
Ann Surg Oncol ; 24(2): 578-585, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26671036

RESUMO

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.


Assuntos
Neoplasias do Colo/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Proteínas do Grupo Polycomb/genética , Proteínas Supressoras de Tumor/genética , Movimento Celular , Neoplasias do Colo/patologia , Epigênese Genética , Genes Supressores de Tumor , Humanos , Células Tumorais Cultivadas
5.
Cancer Cell Int ; 15: 98, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26464562

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-25264105

RESUMO

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.


Assuntos
Epigênese Genética/efeitos dos fármacos , Etanol/toxicidade , Células-Tronco Mesenquimais/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Animais , Sequência de Bases , Primers do DNA , Modelos Animais de Doenças , Feminino , Transtornos do Espectro Alcoólico Fetal/etiologia , Transtornos do Espectro Alcoólico Fetal/genética , Masculino , Reação em Cadeia da Polimerase , Gravidez , Ratos
7.
J Pathol ; 230(3): 298-309, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23362108

RESUMO

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/)


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Bucais/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteínas Supressoras de Tumor/genética , Animais , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular , Estudos de Coortes , Metilação de DNA , Regulação para Baixo , Epigênese Genética , Inativação Gênica , Genes Ligados ao Cromossomo X , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Camundongos , Camundongos SCID , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas/genética , Distribuição Aleatória , Fatores Sexuais , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo
8.
Biochem Biophys Res Commun ; 442(1-2): 56-61, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24211627

RESUMO

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.


Assuntos
Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Peptídeos/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proliferação de Células , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Invasividade Neoplásica , Peptídeos/metabolismo , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/metabolismo
9.
Adv Exp Med Biol ; 754: 195-211, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22956503

RESUMO

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.


Assuntos
Epigênese Genética , Células-Tronco Mesenquimais/patologia , Neoplasias/genética , Neoplasias/patologia , Animais , Humanos
10.
Biochem Biophys Res Commun ; 422(4): 578-85, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22595458

RESUMO

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.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteínas de Ligação ao Cálcio/genética , Metilação de DNA , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Regiões Promotoras Genéticas
11.
Biochem Biophys Res Commun ; 425(2): 290-6, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22842566

RESUMO

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.


Assuntos
Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Neoplasias Hepáticas/genética , Proteínas do Grupo Polycomb/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Cromatina/metabolismo , Loci Gênicos/genética , Células Hep G2 , Humanos , Fatores de Transcrição Kruppel-Like/genética , Proteínas Associadas aos Microtúbulos/genética , Antígenos de Histocompatibilidade Menor
12.
J Biomed Sci ; 18: 3, 2011 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-21219604

RESUMO

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.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Metilação de DNA/efeitos dos fármacos , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/metabolismo , Procainamida/análogos & derivados , Procainamida/farmacologia , Antiarrítmicos/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Humanos
13.
J Biomed Sci ; 18: 12, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-21299869

RESUMO

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.


Assuntos
Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas de Neoplasias/biossíntese , Neoplasias/metabolismo , Animais , Morte Celular/genética , Sobrevivência Celular , Metilação de DNA/genética , Feminino , Células Hep G2 , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Antígenos de Histocompatibilidade Menor , Proteínas de Neoplasias/genética , Neoplasias/patologia
14.
Semin Cancer Biol ; 19(3): 198-208, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19429484

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/genética , Metilação de DNA , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Animais , Genes Supressores de Tumor/fisiologia , Histona Desacetilases/metabolismo , Humanos , Neoplasias/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
15.
Lab Invest ; 90(3): 414-25, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20065949

RESUMO

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.


Assuntos
Apoptose , Metilação de DNA , Proteínas Musculares/metabolismo , Neoplasias Ovarianas/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células , Cisplatino/farmacologia , Decitabina , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética/efeitos dos fármacos , Feminino , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Proteínas Musculares/genética , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/mortalidade , Prognóstico , Regiões Promotoras Genéticas , Modelos de Riscos Proporcionais , Proteínas Ligases SKP Culina F-Box/genética , Proteína Smad4/metabolismo , Taiwan/epidemiologia , Fator de Crescimento Transformador beta/metabolismo , Adulto Jovem
16.
Biochem Biophys Res Commun ; 402(2): 228-34, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20933502

RESUMO

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.


Assuntos
Proteínas E1A de Adenovirus/genética , Citomegalovirus/genética , Metilação de DNA , Regulação Viral da Expressão Gênica , Inativação Gênica , Linhagem Celular , Humanos , Regiões Promotoras Genéticas
17.
Biochem Biophys Res Commun ; 400(3): 305-12, 2010 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-20727853

RESUMO

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.


Assuntos
Linhagem da Célula/genética , Metilação de DNA , Inativação Gênica , Células-Tronco Mesenquimais/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Animais , Diferenciação Celular/genética , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/citologia , Proteínas Associadas aos Microtúbulos/genética , Antígenos de Histocompatibilidade Menor , Regiões Promotoras Genéticas , Ratos
18.
Life Sci ; 78(13): 1483-93, 2006 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-16324718

RESUMO

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.


Assuntos
Transtornos do Crescimento/induzido quimicamente , Nicotina/toxicidade , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Modelos Animais de Doenças , Feminino , Masculino , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Aumento de Peso/efeitos dos fármacos
19.
Brain Res ; 1008(1): 69-80, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15081384

RESUMO

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.


Assuntos
Depressores do Sistema Nervoso Central/administração & dosagem , Etanol/administração & dosagem , Neurônios/efeitos dos fármacos , Receptores de GABA-A/fisiologia , Septo do Cérebro/citologia , Análise de Variância , Animais , Animais Recém-Nascidos , Anticonvulsivantes/farmacologia , Bicuculina/farmacologia , Peso Corporal/efeitos dos fármacos , Células Cultivadas , Depressores do Sistema Nervoso Central/farmacologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas , Embrião de Mamíferos , Etanol/sangue , Etanol/farmacologia , Feminino , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Moduladores GABAérgicos/farmacologia , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp/métodos , Penicilinas/farmacologia , Picrotoxina/farmacologia , Gravidez , Inibidores da Síntese de Proteínas/farmacologia , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/efeitos dos fármacos , Estreptomicina/farmacologia , Fatores de Tempo , Triazóis/farmacologia , Regulação para Cima/efeitos dos fármacos , Zinco/farmacologia , Zolpidem , Ácido gama-Aminobutírico/farmacologia
20.
Brain Res Dev Brain Res ; 142(1): 89-99, 2003 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-12694947

RESUMO

The impact of binge-like, early postnatal ethanol treatment on AMPA or kainate whole cell currents was examined in acutely isolated medial septum/diagonal band (MS/DB) neurons. AMPA (10 or 100 microM) current was inhibited by GYKI 52466, a selective AMPA receptor (AMPAR) antagonist, in all neurons isolated on postnatal day (PD) 5-8, PD 12-15 or PD 32-35. Cyclothiazide, a selective inhibitor of AMPAR desensitization, also effectively potentiated AMPA currents. This suggests that non-NMDA, ionotropic glutamate receptors on immature MS/DB neuron are predominantly AMPARs. Concentration-dependent kainate (10-1000 microM) application evoked nondesensitizing currents that exhibited an increase in the maximum response by the end of first postnatal month, consistent with developmental regulation of AMPAR function. Acute 3 s ethanol application (100 mM) consistently blunted AMPA- and kainate currents approximately 20-30% across age groups. Inhibition was sustained during continuous ethanol superfusion lasting 10-12 min without evidence of acute tolerance. Repeated oral intubation of rat pups with ethanol (5.25 g/kg/day on PD 4-9), which models third trimester human binge drinking, resulted in peak blood ethanol levels of approximately 350 mg/dl (measured 90 min after PD 6 dosing). AMPA or kainate currents were upregulated in neurons isolated on PD 32-35 by earlier ethanol intubation suggesting that binge-like intoxication augments developing AMPAR function. Despite this augmentation of AMPAR function, no significant changes were found in the sensitivity of AMPA currents to GYKI 52466, cyclothiazide or acute ethanol (100 mM) sensitivity or in the levels of GluR1/GluR2 subunit proteins from MS/DB tissue. These results indicate that non-NMDA ionotrophic glutamate receptors on immature MS/DB neurons, which are largely of the AMPAR subtype, are moderately sensitive to immediate inhibition by ethanol. Repeating this inhibition during early postnatal binge-like intoxication can augment normal development of AMPAR function.


Assuntos
Transtornos do Sistema Nervoso Induzidos por Álcool/fisiopatologia , Benzodiazepinas , Depressores do Sistema Nervoso Central/farmacologia , Feixe Diagonal de Broca/crescimento & desenvolvimento , Etanol/farmacologia , Neurônios/efeitos dos fármacos , Receptores de AMPA/efeitos dos fármacos , Intoxicação Alcoólica/fisiopatologia , Animais , Ansiolíticos/farmacologia , Anti-Hipertensivos/farmacologia , Benzotiadiazinas/farmacologia , Western Blotting , Células Cultivadas , Feixe Diagonal de Broca/citologia , Feixe Diagonal de Broca/efeitos dos fármacos , Feixe Diagonal de Broca/fisiopatologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Ácido Caínico/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Modelos Animais , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/antagonistas & inibidores , Receptores de AMPA/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA