Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(17)2021 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-34502300

RESUMO

Folate depletion causes chromosomal instability by increasing DNA strand breakage, uracil misincorporation, and defective repair. Folate mediated one-carbon metabolism has been suggested to play a key role in the carcinogenesis and progression of hepatocellular carcinoma (HCC) through influencing DNA integrity. Methylenetetrahydrofolate reductase (MTHFR) is the enzyme catalyzing the irreversible conversion of 5,10-methylenetetrahydrofolate to 5-methyltetrahydrofolate that can control folate cofactor distributions and modulate the partitioning of intracellular one-carbon moieties. The association between MTHFR polymorphisms and HCC risk is inconsistent and remains controversial in populational studies. We aimed to establish an in vitro cell model of liver origin to elucidate the interactions between MTHFR function, folate status, and chromosome stability. In the present study, we (1) examined MTHFR expression in HCC patients; (2) established cell models of liver origin with stabilized inhibition of MTHFR using small hairpin RNA delivered by a lentiviral vector, and (3) investigated the impacts of reduced MTHFR and folate status on cell cycle, methyl group homeostasis, nucleotide biosynthesis, and DNA stability, all of which are pathways involved in DNA integrity and repair and are critical in human tumorigenesis. By analyzing the TCGA/GTEx datasets available within GEPIA2, we discovered that HCC cancer patients with higher MTHFR had a worse survival rate. The shRNA of MTHFR (shMTHFR) resulted in decreased MTHFR gene expression, MTHFR protein, and enzymatic activity in human hepatoma cell HepG2. shMTHFR tended to decrease intracellular S-adenosylmethionine (SAM) contents but folate depletion similarly decreased SAM in wildtype (WT), negative control (Neg), and shMTHFR cells, indicating that in cells of liver origin, shMTHFR does not exacerbate the methyl group supply in folate depletion. shMTHFR caused cell accumulations in the G2/M, and cell population in the G2/M was inversely correlated with MTHFR gene level (r = -0.81, p < 0.0001), MTHFR protein expression (r = -0.8; p = 0.01), and MTHFR enzyme activity (r = -0.842; p = 0.005). Folate depletion resulted in G2/M cell cycle arrest in WT and Neg but not in shMTHFR cells, indicating that shMTHFR does not exacerbate folate depletion-induced G2/M cell cycle arrest. In addition, shMTHFR promoted the expression and translocation of nuclei thymidine synthetic enzyme complex SHMT1/DHFR/TYMS and assisted folate-dependent de novo nucleotide biosynthesis under folate restriction. Finally, shMTHFR promoted nuclear MLH1/p53 expression under folate deficiency and further reduced micronuclei formation and DNA uracil misincorporation under folate deficiency. In conclusion, shMTHFR in HepG2 induces cell cycle arrest in G2/M that may promote nucleotide supply and assist cell defense against folate depletion-induced chromosome segregation and uracil misincorporation in the DNA. This study provided insight into the significant impact of MTHFR function on chromosome stability of hepatic tissues. Data from the present study may shed light on the potential regulatory mechanism by which MTHFR modulates the risk for hepatic malignancies.


Assuntos
Carcinoma Hepatocelular/patologia , Segregação de Cromossomos , DNA de Neoplasias/genética , Ácido Fólico/metabolismo , Metilenotetra-Hidrofolato Redutase (NADPH2)/antagonistas & inibidores , Uracila/metabolismo , Apoptose , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Instabilidade Cromossômica , DNA de Neoplasias/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Polimorfismo Genético , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas
2.
Int J Mol Sci ; 21(4)2020 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-32093290

RESUMO

Prodrug activator gene therapy mediated by murine leukemia virus (MLV)-based retroviral replicating vectors (RRV) was previously shown to be highly effective in killing glioma cells both in culture and in vivo. To avoid receptor interference and enable dual vector co-infection with MLV-RRV, we have developed another RRV based on gibbon ape leukemia virus (GALV) that also shows robust replicative spread in a wide variety of tumor cells. We evaluated the potential of GALV-based RRV as a cancer therapeutic agent by incorporating yeast cytosine deaminase (CD) and E. coli nitroreductase (NTR) prodrug activator genes into the vector. The expression of CD and NTR genes from GALV-RRV achieved highly efficient delivery of these prodrug activator genes to RG-2 glioma cells, resulting in enhanced cytotoxicity after administering their respective prodrugs 5-fluorocytosine and CB1954 in vitro. In an immune-competent intracerebral RG-2 glioma model, GALV-mediated CD and NTR gene therapy both significantly suppressed tumor growth with CB1954 administration after a single injection of vector supernatant. However, NTR showed greater potency than CD, with control animals receiving GALV-NTR vector alone (i.e., without CB1954 prodrug) showing extensive tumor growth with a median survival time of 17.5 days, while animals receiving GALV-NTR and CB1954 showed significantly prolonged survival with a median survival time of 30 days. In conclusion, GALV-RRV enabled high-efficiency gene transfer and persistent expression of NTR, resulting in efficient cell killing, suppression of tumor growth, and prolonged survival upon CB1954 administration. This validates the use of therapeutic strategies employing this prodrug activator gene to arm GALV-RRV, and opens the door to the possibility of future combination gene therapy with CD-armed MLV-RRV, as the latter vector is currently being evaluated in clinical trials.


Assuntos
Aziridinas/farmacologia , Neoplasias Encefálicas/terapia , Flucitosina/farmacologia , Terapia Genética , Vetores Genéticos , Glioma/terapia , Neoplasias Experimentais/terapia , Pró-Fármacos/farmacologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Citosina Desaminase/biossíntese , Citosina Desaminase/genética , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/genética , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Vírus da Leucemia do Macaco Gibão , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Nitrorredutases/biossíntese , Nitrorredutases/genética , Ratos Endogâmicos F344 , Proteínas de Saccharomyces cerevisiae/biossíntese , Proteínas de Saccharomyces cerevisiae/genética
3.
J Transl Med ; 12: 237, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25179542

RESUMO

BACKGROUND: The treatment of oral squamous cell carcinoma (OSCC) following early detection is associated with good outcomes. Therefore, the survival and prognosis of OSCC patients could be hugely improved by identifying reliable biomarkers for the early diagnosis of the disease. Our previous methylation microarray analysis results have suggested that the gene encoding tissue factor pathway inhibitor-2 (TFPI-2) is a potential clinical predictor as well as a key regulator involved in OSCC malignancy. METHODS: Methylation of the TFPI-2 promoter in oral tissue specimens was evaluated by bisulfite sequencing assay, quantitative methylation-specific PCR, and pyrosequencing assay. The differences in methylation levels among the groups were compared using the Mann-Whitney U test. The area under the receiver operating characteristic curve (AUROC) was used to evaluate the discrimination ability for detecting OSCC. Cellular TFPI-2 expression was analyzed by quantitative reverse-transcription PCR before and after treatment with 5'-aza-2'-deoxycytidine and trichostatin A, to confirm whether TFPI-2 was epigenetically silenced in OSCC cells. We investigated whether TFPI-2 plays a role as a tumor suppressor by establishing TFPI-2-overexpressing OSCC cells and subjecting them to in vitro cellular proliferation, migration, and invasion assays, as well as an in vivo metastasis assay. RESULTS: TFPI-2 was hypermethylated in OSCC tissues versus normal oral tissues (P < 0.0001), with AUROC = 0.91, when using a pyrosequencing assay to quantify the methylation level. TFPI-2 silencing in OSCC was regulated by both DNA methylation and chromatin histone modification. Restoration of TFPI-2 counteracted the invasiveness of OSCC by inhibiting the enzymatic activity of matrix metalloproteinase-2, and consequently interfered with OSCC metastasis in vivo. CONCLUSIONS: Our data suggest strongly that TFPI-2 is a down-regulated tumor suppressor gene in OSCC, probably involving epigenetic silencing mechanisms. The loss of TFPI-2 expression is a key event for oral tumorigenesis, especially in the process of tumor metastasis.


Assuntos
Carcinoma de Células Escamosas/genética , Metilação de DNA , Inativação Gênica , Glicoproteínas/genética , Neoplasias Bucais/genética , Regiões Promotoras Genéticas , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Bucais/diagnóstico , Neoplasias Bucais/patologia , Prognóstico
4.
Biomater Adv ; 154: 213639, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37793310

RESUMO

Inhibition to glioblastoma multiforme (GBM) propagation is a critical challenge in clinical practice because binding of inhibitors of apoptosis proteins (IAPs) to caspase prevents cancer cells from death. In this study, folic acid (FA), lactoferrin (Lf) and rabies virus glycoprotein (RVG) were grafted on lipopolymers (LPs) composed of poly(ε-caprolactone) and Compritol 888 ATO to encapsulate AZD5582 (AZD), GDC0152 (GDC) and curcumin (CURC). The standard deviations of initial particle diameter and particle diameter after storage for 30 days were involved in LP composition optimization. The functionalized LPs were used to permeate the blood-brain barrier (BBB) and constrain IAP quantity in GBM cells. Experimental results revealed that an increase in Span 20 (emulsifier) concentration enlarged the size of LPs, and enhanced the entrapment and releasing efficiency of AZD, DGC and CURC. 1H nuclear magnetic resonance spectra showed that the hydrogen bonds between the LPs and drugs supported the sustained release of AZD, DGC and CURC from the LPs. The LPs modified with the three targeting biomolecules facilitated the penetration of AZD, GDC and CURC across the BBB, and could recognize U87MG cells and human brain cancer stem cells. Immunofluorescence staining, flow cytometry and western blot demonstrated that CURC-incorporated LPs enhanced AZD and GDC activity in suppressing cellular IAP 1 (cIAP1) and X-linked IAP (XIAP) levels, and raising caspase-3 level in GBM. Surface FA, Lf and RVG also promoted the ability of the drug-loaded LPs to avoid carcinoma growth. The current FA-, Lf- and RVG-crosslinked LPs carrying AZD, DGC and CURC can be promising in hindering IAP expressions for GBM management.


Assuntos
Neoplasias Encefálicas , Curcumina , Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Curcumina/farmacologia , Curcumina/uso terapêutico , Curcumina/química , Lipopolissacarídeos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Apoptose
5.
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
6.
Mutat Res ; 678(1): 53-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19591959

RESUMO

Environmental tobacco smoke (ETS) exposure might increase the risk for childhood asthma, and we hypothesized the effect may be modified by the phase II genes NAD(P)H: quinone oxidoreductase 1 (NQO1) and glutathione S-transferase (GST) M1. To investigate the genetic and environmental associations with asthma, GSTM1 and NQO1 functional polymorphisms and ETS were analyzed in a two-staged cross-sectional study among elementary schoolchildren in Taiwan. Multiple logistic regression analysis revealed a significant association between the Ser allele of the NQO1 Pro187Ser polymorphism and asthma (OR=1.6, 95% CI 1.3-1.8). Although GSTM1 genotype itself was not significantly associated with asthma (OR=1.0, 95% CI 0.8-1.1), the GSTM1 genotype modified the association between the NQO1 polymorphism and asthma in children exposed to ETS (p=0.0002). The NQO1 gene might be involved in the development of asthma, especially in children carrying the GSTM1 null genotype who are exposed to ETS.


Assuntos
Asma/induzido quimicamente , Predisposição Genética para Doença , Glutationa Transferase/genética , NAD(P)H Desidrogenase (Quinona)/genética , Poluição por Fumaça de Tabaco , Asma/genética , Criança , Feminino , Humanos , Masculino , Polimorfismo Genético , Fatores de Risco
7.
Cancer Res ; 67(11): 5345-53, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545615

RESUMO

Replication-competent retrovirus (RCR) vectors are intrinsically incapable of infecting quiescent cells and have been shown to achieve highly efficient and tumor-restricted replicative spread and gene transfer in vivo after direct intratumoral injection in a variety of primary cancer models. However, i.v. delivery of RCR vectors expressing therapeutic genes has never previously been tested, particularly in an immunocompetent tumor model. Therefore, in the present study, we sought to test the therapeutic effect of an RCR vector (ACE-CD) carrying the yeast cytosine deaminase (CD) gene, which converts the nontoxic prodrug 5-fluorocytosine (5FC) into the chemotoxin 5-fluorouracil, after delivery by infusion into the locoregional circulation in a multifocal hepatic metastasis model of colon cancer. After confirmation of suicide gene cytotoxicity in vitro, multifocal hepatic tumors were established in syngeneic mice with murine CT26 colorectal cancer cells expressing firefly luciferase (CT26-Luc), and the ACE-CD vector was infused via intrasplenic injection into the portal circulation. Fourteen days after locoregional infusion, systemic administration of 5FC resulted in significant inhibition of bioluminescent signals in mice whose tumors had been infected with RCR but not in control mice. Notably, there was no detectable RCR vector spread to normal liver or bone marrow by quantitative PCR analysis. Our results thus show that locoregional delivery of a suicide gene by RCR vectors infused into the portal circulation results in progressive transduction of multiple tumor foci in the liver, without evidence of spread to adjacent normal parenchyma or extrahepatic tissues, and can achieve significant tumor growth inhibition.


Assuntos
Neoplasias Colorretais/terapia , Genes Transgênicos Suicidas , Terapia Genética/métodos , Retroviridae/fisiologia , Animais , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/virologia , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Modelos Animais de Doenças , Feminino , Flucitosina/farmacocinética , Fluoruracila/farmacocinética , Fluoruracila/farmacologia , Vetores Genéticos/genética , Humanos , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/secundário , Neoplasias Hepáticas Experimentais/terapia , Neoplasias Hepáticas Experimentais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Retroviridae/genética , Replicação Viral
8.
Front Biosci ; 13: 3083-95, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17981778

RESUMO

Oncolytic virotherapy represents an emerging field with tremendous promise for harnessing the replicative capabilities of viruses against rapidly proliferating cancer cells. Among the different replicating virus technologies being tested, replication-competent retrovirus (RCR) vectors based on murine leukemia virus (MLV) exhibit unique characteristics. MLV exhibits intrinsic tumor selectivity due to its inability to infect quiescent cells, and can achieve highly selective and stable gene transfer throughout entire solid tumors in vivo at efficiencies of up to >99%, even after initial inoculation at MOIs as low as 0.01. RCR vectors with suicide genes mediate synchronized cell killing after prodrug administration, and due to their ability to undergo stable integration, residual cancer cells serve as a reservoir for long-term viral persistence even as they migrate to new sites, enabling multiple cycles of prodrug to achieve prolonged survival benefit. Further testing in various tumor models, new vector targeting and delivery strategies, and development of GMP manufacturing, are being pursued through a multi-national consortium, and preparations are now being undertaken for clinical trials using RCR vectors in glioblastoma.


Assuntos
Terapia Genética/métodos , Neoplasias/genética , Neoplasias/terapia , Retroviridae/genética , Adenoviridae/genética , Vetores Genéticos , Humanos , Vírus da Leucemia Murina/genética , Masculino , Mutagênese , Neoplasias/patologia , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , Transcrição Gênica , Transfecção
9.
Sci Rep ; 8(1): 2213, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29396437

RESUMO

Glioblastoma multiforme (GBM), the most common malignant brain tumor, has a short period of survival even with recent multimodality treatment. The neurotropic JC polyomavirus (JCPyV) infects glial cells and oligodendrocytes and causes fatal progressive multifocal leukoencephalopathy in patients with AIDS. In this study, a possible gene therapy strategy for GBM using JCPyV virus-like particles (VLPs) as a gene delivery vector was investigated. We found that JCPyV VLPs were able to deliver the GFP reporter gene into tumor cells (U87-MG) for expression. In an orthotopic xenograft model, nude mice implanted with U87 cells expressing the near-infrared fluorescent protein and then treated by intratumoral injection of JCPyV VLPs carrying the thymidine kinase suicide gene, combined with ganciclovir administration, exhibited significantly prolonged survival and less tumor fluorescence during the experiment compared with controls. Furthermore, JCPyV VLPs were able to protect and deliver a suicide gene to distal subcutaneously implanted U87 cells in nude mice via blood circulation and inhibit tumor growth. These findings show that metastatic brain tumors can be targeted by JCPyV VLPs carrying a therapeutic gene, thus demonstrating the potential of JCPyV VLPs to serve as a gene therapy vector for the far highly treatment-refractory GBM.


Assuntos
Neoplasias Encefálicas/terapia , Portadores de Fármacos , Terapia Genética/métodos , Vetores Genéticos , Glioblastoma/terapia , Vírus JC/genética , Virossomos/genética , Animais , Linhagem Celular Tumoral , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Xenoenxertos , Humanos , Camundongos Nus , Transplante de Neoplasias , Transdução Genética , Resultado do Tratamento
10.
Neurosurg Focus ; 20(4): E25, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16709031

RESUMO

OBJECT: The authors had previously reported on a replication-competent retrovirus (RCR) that has been demonstrated to be stable, capable of effective transduction, and able to prolong survival in an intracranial tumor model in nude mice. The purpose of this study was further investigation of this gene therapy option. METHODS: The transduction efficiency of RCR in RG2, an immunocompetent intracranial tumor model, was tested in Fischer 344 rats. The immune response to the RCR vector was expressed by the quantification of CD4, CD8, and CD11/b in tumors. The pharmaceutical efficacy of the suicide gene CD in converting prodrug 5-fluorocytosine (5-FC) to 5-fluorouracil (5-FU) was measured using fluorine-19 nuclear magnetic resonance (19F-NMR) spectroscopy. Animal survival data were plotted on Kaplan-Meier survival curves. Finally, the biodistribution of RCR was determined using quantitative real-time polymerase chain reaction (RT-PCR) for the detection of retroviral env gene. There was no evidence of viral transduction in normal brain cells. Neither severe inflammation nor immunoreaction occurred after intracranial injection of RCR-green fluorescent protein compared with phosphate-buffered saline (PBS). The 19F-NMR spectroscopy studies demonstrated that RCR-CD was able to convert 5-FC to 5-FU effectively in vitro. The infection of RG2 brain tumors with RCR-CD and their subsequent treatment with 5-FC significantly prolonged survival compared with that in animals with RG2 transduced tumors treated with PBS. In contrast to the nude mouse model, evidence of virus dissemination to the systemic organs after intracranial injection was not detected using RT-PCR. CONCLUSIONS: The RCR-mediated suicide gene therapy described in this paper effectively transduced malignant gliomas in an immunocompetent in vivo rodent model, prolonging survival, without evidence of severe intracranial inflammation, and without local transduction of normal brain cells or systemic organs.


Assuntos
Neoplasias Encefálicas/terapia , Genes Transgênicos Suicidas/genética , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Glioma/terapia , Terapia Viral Oncolítica/métodos , Retroviridae/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , DNA Viral/análise , DNA Viral/genética , Modelos Animais de Doenças , Encefalite/fisiopatologia , Encefalite/prevenção & controle , Flucitosina/metabolismo , Terapia Genética/tendências , Vetores Genéticos/genética , Glioma/genética , Glioma/metabolismo , Humanos , Hospedeiro Imunocomprometido/genética , Masculino , Terapia Viral Oncolítica/tendências , Ratos , Ratos Endogâmicos F344 , Taxa de Sobrevida , Transdução Genética/métodos , Transdução Genética/tendências , Resultado do Tratamento , Replicação Viral/genética
11.
Oncol Lett ; 12(5): 4224-4230, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27895796

RESUMO

Tumor-node-metastasis is one of the leading causes of morbidity and mortality in thyroid cancer patients. Upregulation of vascular endothelial growth factor-C (VEGF-C) increases the migratory ability of thyroid cancer cells to lymph nodes. Expression of neuropilin-2 (NRP-2), the co-receptor of VEGF-C, has been reported to be correlated with lymph node metastasis in human thyroid cancer. The present study investigated the role of VEGF-C/NRP-2 signaling in the regulation of metastasis of two different types of human thyroid cancer cells. The results indicated that the VEGF-C/NRP-2 axis significantly promoted the metastatic activities of papillary thyroid carcinoma cells through the activation of the mitogen-activated protein kinase (MAPK) kinase (MEK)/extracellular signal-regulated kinase and p38 MAPK signaling cascades. However, neither MEK or p38 MAPK inhibitors produced significant inhibition of the migratory activity and invasiveness regulated by the VEGF-C/NRP-2 axis in follicular thyroid carcinoma cells. Finally, VEGF-C/NRP-2-mediated invasion and migration of thyroid cancer cells required the expression of NRP-2. The present results demonstrate that the promotion of metastasis by VEGF-C is mainly due to the upregulation of NRP-2 in thyroid cancer cells, and this metastatic activity regulated by the VEGF-C/NRP-2 axis provides further insight into the process of tumor metastasis.

12.
Oncol Lett ; 12(3): 2107-2114, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27602148

RESUMO

Oral cancer is the eighth most common type of cancer among men worldwide, with an age-standardized rate of 6.3 per 100,000, and is the fourth leading cause of cancer-associated mortality among men in Taiwan. Cisplatin and 5-fluorouracil (5-FU) are two of the most frequently utilized chemotherapy drugs for the treatment of oral cancer. Although oral cancer patients initially benefit from chemotherapy with these drugs, they may develop resistance to them, which worsens their prognosis and reduces survival rates. It has been reported that increased levels of epidermal growth factor receptor (EGFR) and multidrug resistance-associated protein 2 (MRP2) induce drug resistance in numerous types of human cancer. Therefore, the present study employed lentivirus vector-mediated RNA interference (RNAi) in order to target the genes encoding EGFR and MRP2 in the oral squamous cell carcinoma cell line OC2. It was observed that RNAi-mediated downregulation of EGFR or MRP2 increased the sensitivity to 5-FU and cisplatin in OC2 cells. Downregulation of EGFR resulted in significant suppression of OC2 tumor growth following 5-FU administration. However, simultaneous downregulation of the two genes did not further suppress the tumor growth, indicating that MRP2 does not have a significant role in the chemosensitivity of EGFR-downregulated cells to 5-FU. In contrast, downregulation of MRP2 was demonstrated to significantly enhance the therapeutic effects of cisplatin in EGFR-downregulated OC2 tumors. The observation that the expression of MRP2 was positively correlated with the level of cisplatin resistance in cells suggests that RNAi-mediated downregulation of MRP2 may be applicable as a therapeutic approach toward reversing MRP2-dependent cisplatin resistance in oral cancer.

13.
Epigenetics ; 10(3): 229-36, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25612142

RESUMO

Oral squamous cell carcinoma (OSCC) constitutes >90% of oral cancers and is the sixth most common malignancy among males worldwide and the fourth leading cause of death due to cancer among males in Taiwan. However, most patients do not receive a diagnosis of OSCC until the late stages, which have a lower survival rate. The use of molecular marker analysis to identify early-stage OSCC would permit optimal timing for treatments and consequently prolong survival. The aim of this study was to identify biomarkers of OSCC using the Illumina GoldenGate Methylation Cancer Panel, which comprised a total of 1,505 CpG sites covering 807 genes. Samples of buccal mucosa resected from 40 OSCC patients and normal tissue samples obtained from 15 patients (normal mucosa from OSCC patients or from patients undergoing surgery unrelated to OSCC) were analyzed. Fms-related tyrosine kinase 4 (FLT4) methylation exhibited a perfect specificity for detecting OSCC, with an area under the receiver operating characteristic curve of 0.91 for both all-stage and early-stage OSCC. Methylation of 7 genes (ASCL1, FGF3, FLT4, GAS7, KDR, TERT, and TFPI2) constitutes the top-20 panels for detecting OSCC. The top-20 panels for detecting early-stage OSCC contain 8 genes: ADCYAP1, EPHA7, FLT4, GSTM2, KDR, MT1A, NPY, and TFPI2. FLT4 RNA expression and methylation level were validated using RT-PCR and a pyrosequencing methylation assay. The median level of FLT4 expression was 2.14-fold for normal relative to OSCC tissue samples (P < 0.0001). Among the 8 pyrosequenced FLT4 CpG sites, methylation level was much higher in the OSCC samples. In conclusion, methylation statuses of selected genes, and especially FLT4, KDR, and TFPI2, might be of great potential as biomarkers for early detection of buccal OSCC.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/metabolismo , Metilação de DNA , Neoplasias Bucais/metabolismo , Adulto , Ilhas de CpG , Glicoproteínas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
14.
Clin Epigenetics ; 7: 1, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25628764

RESUMO

BACKGROUND: The dysregulation of transforming growth factor-ß (TGF-ß) signaling plays a crucial role in ovarian carcinogenesis and in maintaining cancer stem cell properties. Classified as a member of the ATP-binding cassette (ABC) family, ABCA1 was previously identified by methylated DNA immunoprecipitation microarray (mDIP-Chip) to be methylated in ovarian cancer cell lines, A2780 and CP70. By microarray, it was also found to be upregulated in immortalized ovarian surface epithelial (IOSE) cells following TGF-ß treatment. Thus, we hypothesized that ABCA1 may be involved in ovarian cancer and its initiation. RESULTS: We first compared the expression level of ABCA1 in IOSE cells and a panel of ovarian cancer cell lines and found that ABCA1 was expressed in HeyC2, SKOV3, MCP3, and MCP2 ovarian cancer cell lines but downregulated in A2780 and CP70 ovarian cancer cell lines. The reduced expression of ABCA1 in A2780 and CP70 cells was associated with promoter hypermethylation, as demonstrated by bisulfite pyro-sequencing. We also found that knockdown of ABCA1 increased the cholesterol level and promoted cell growth in vitro and in vivo. Further analysis of ABCA1 methylation in 76 ovarian cancer patient samples demonstrated that patients with higher ABCA1 methylation are associated with high stage (P = 0.0131) and grade (P = 0.0137). Kaplan-Meier analysis also found that patients with higher levels of methylation of ABCA1 have shorter overall survival (P = 0.019). Furthermore, tissue microarray using 55 ovarian cancer patient samples revealed that patients with a lower level of ABCA1 expression are associated with shorter progress-free survival (P = 0.038). CONCLUSIONS: ABCA1 may be a tumor suppressor and is hypermethylated in a subset of ovarian cancer patients. Hypermethylation of ABCA1 is associated with poor prognosis in these patients.

15.
Oncotarget ; 6(2): 915-34, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25460508

RESUMO

Radioresistance is still an emerging problem for radiotherapy of oral cancer. Aberrant epigenetic alterations play an important role in cancer development, yet the role of such alterations in radioresistance of oral cancer is not fully explored. Using a methylation microarray, we identified promoter hypermethylation of FHIT (fragile histidine triad) in radioresistant OML1-R cells, established from hypo-fractionated irradiation of parental OML1 radiosensitive oral cancer cells. Further analysis confirmed that transcriptional repression of FHIT was due to promoter hypermethylation, H3K27me3 and overexpression of methyltransferase EZH2 in OML1-R cells. Epigenetic interventions or depletion of EZH2 restored FHIT expression. Ectopic expression of FHIT inhibited tumor growth in both in vitro and in vivo models, while also resensitizing radioresistant cancer cells to irradiation, by restoring Chk2 phosphorylation and G2/M arrest. Clinically, promoter hypermethylation of FHIT inversely correlated with its expression and independently predicted both locoregional control and overall survival in 40 match-paired oral cancer patient samples. Further in vivo therapeutic experiments confirmed that inhibition of DNA methylation significantly resensitized radioresistant oral cancer cell xenograft tumors. These results show that epigenetic silencing of FHIT contributes partially to radioresistance and predicts clinical outcomes in irradiated oral cancer. The radiosensitizing effect of epigenetic interventions warrants further clinical investigation.


Assuntos
Hidrolases Anidrido Ácido/genética , Metilação de DNA , Neoplasias Bucais/radioterapia , Proteínas de Neoplasias/genética , Tolerância a Radiação/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Hidrolases Anidrido Ácido/metabolismo , Animais , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste , Epigênese Genética , Feminino , Inativação Gênica , Células HEK293 , Histonas/metabolismo , Humanos , Estimativa de Kaplan-Meier , Masculino , Metilação , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Proteínas de Neoplasias/metabolismo , Avaliação de Resultados em Cuidados de Saúde/métodos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Prognóstico , Carga Tumoral/genética , Carga Tumoral/efeitos da radiação
16.
Hum Gene Ther ; 14(2): 117-27, 2003 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-12614563

RESUMO

The first large randomized phase III trial in gene therapy demonstrated no improvement in the survival of patients injected with packaging cells that produced conventional replication-defective retroviral vectors carrying the herpes simplex virus thymidine kinase gene, a disappointing result that was attributed to extremely poor levels of transduction efficiency. To circumvent this problem, we have developed a modified replication-competent retrovirus (RCR) that is capable of transducing human glioma cell lines A-172, U-87, T-98G, U-373, and U-138 and rat glioma cell lines C6 and 9L, over multiple infection cycles in vitro, resulting in a tremendous enhancement in transduction efficiency over conventional replication-defective retroviral vectors at the same dose. Whereas the transduction efficiency of conventional retroviral vectors injected into preestablished subcutaneous U-87 tumors at a dose of 1.0 x 10(5) transducing units (TU) was only 0.2% at 6 weeks postinjection, the same dose of RCR vector resulted in up to 97.2% transduction. When RCR vectors at a dose of 1.0 x 10(4) TU were injected into preestablished intracranial U-87 tumors, transduction efficiency at 2 and 3 weeks was 74 and 98.1%, respectively. Notably, however, intracranial injection of RCR vectors did not result in detectable infection of normal brain cells. Furthermore, using a sensitive polymerase chain reaction assay, no detectable RCR signal could be observed in any extracerebral tissues, including lung, liver, kidney, upper gastrointestinal tract (esophagus and stomach), lower gastrointestinal tract (colon and small intestine), skin, spleen, and bone marrow. Treatment of U-87 intracranial gliomas with RCR vectors carrying the yeast cytosine deaminase suicide gene followed by 5-fluorocytosine prodrug administration resulted in 100% survival over a 60-day follow-up period, compared with 0% survival of control groups receiving vector alone or prodrug alone. Our results demonstrate that RCR vectors can achieve therapeutically significant levels of transduction in malignant human gliomas, and that RCR vector spread after intratumoral injection is restricted to the tumor itself.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Glioma/genética , Glioma/terapia , Vírus da Leucemia Murina , Animais , Genes Reporter/genética , Genes Reporter/fisiologia , Humanos , Camundongos , Ratos , Transdução Genética , Células Tumorais Cultivadas
17.
Hum Gene Ther ; 14(8): 789-802, 2003 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-12804141

RESUMO

Replication-competent murine leukemia virus (MLV) vectors can be engineered to achieve high efficiency gene transfer to solid tumors in vivo and tumor-restricted replication, however their safety can be further enhanced by redirecting tropism of the virus envelope. We have therefore tested the targeting capability and replicative stability of ecotropic and amphotropic replication-competent retrovirus (RCR) vectors containing two tandem repeats from the immunoglobulin G-binding domain of Staphylococcal protein A inserted into the proline-rich "hinge" region of the envelope, which enables modular use of antibodies of various specificities for vector targeting. The modified envelopes were efficiently expressed and incorporated into virions, were capable of capturing monoclonal anti-HER2 antibodies, and mediated efficient binding of the virus-antibody complex to HER2-positive target cells. While infectivity was markedly reduced by pseudotyping with targeted envelopes alone, coexpression of wild-type envelope rescued efficient cellular entry. Both ecotropic and amphotropic RCR vector/anti-HER2 antibody complexes achieved significant enhancement of transduction on murine target cells overexpressing HER2, which could be competed by preincubation with excess free antibodies. Interestingly, HER2-expressing human breast cancer cells did not show enhancement of transduction despite efficient antibody-mediated cell surface binding, suggesting that target cell-specific parameters markedly affect the efficiency of post-binding entry processes. Serial replication of targeted vectors resulted in selection of Z domain deletion variants, but reduction of the overall size of the vector genome enhanced its stability. Application of antibody-mediated targeting to the initial localization of replication-competent virus vectors to tumor sites will thus require optimized target selection and vector design.


Assuntos
Anticorpos Monoclonais/imunologia , Neoplasias da Mama/metabolismo , Vetores Genéticos , Vírus da Leucemia Murina/genética , Animais , Anticorpos Monoclonais/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , DNA Viral/análise , Feminino , Citometria de Fluxo , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde , Humanos , Cinética , Vírus da Leucemia Murina/fisiologia , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Camundongos , Células NIH 3T3 , Plasmídeos , Estrutura Terciária de Proteína , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Proteínas Recombinantes de Fusão/genética , Proteína Estafilocócica A/química , Transdução Genética , Proteínas do Envelope Viral/genética , Vírion/química , Replicação Viral
18.
Int J Environ Res Public Health ; 11(6): 6504-16, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25003170

RESUMO

Asthma is a chronic disease that is particularly common in children. The association between polymorphisms of the gene encoding intercellular adhesion molecule 1 (ICAM1) and gene-environment interactions with childhood asthma has not been fully investigated. A cross-sectional study was undertaken to investigate these associations among children in Taiwan. The effects of two functional single-nucleotide polymorphisms (SNPs) of ICAM1, rs5491 (K56M) and rs5498 (K469E), and exposure to environmental tobacco smoke (ETS) were studied. Two hundred and eighteen asthmatic and 877 nonasthmatic children were recruited from elementary schools. It was found that the genetic effect of each SNP was modified by the other SNP and by exposure to ETS. The risk of asthma was higher for children carrying the rs5491 AT or TT genotypes and the rs5498 GG genotype (odds ratio = 1.68, 95% confidence interval 1.09­2.59) than for those with the rs5491 AA and rs5498 AA or AG genotypes (the reference group). The risk for the other two combinations of genotypes did not differ significantly from that of the reference group (p of interaction = 0.0063). The two studied ICAM1 SNPs were associated with childhood asthma among children exposed to ETS, but not among those without ETS exposure (p of interaction = 0.05 and 0.01 for rs5491 and rs5498, respectively). Both ICAM1 and ETS, and interactions between these two factors are likely to be involved in the development of asthma in childhood.


Assuntos
Asma/genética , Molécula 1 de Adesão Intercelular/genética , Polimorfismo de Nucleotídeo Único , Poluição por Fumaça de Tabaco/efeitos adversos , Asma/etiologia , Criança , Feminino , Genótipo , Humanos , Masculino , Razão de Chances , Inquéritos e Questionários , Taiwan
19.
Oncol Rep ; 28(1): 21-6, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22552490

RESUMO

Gene therapy mediated by murine leukemia virus (MLV)-based replicating retrovirus vector (RRV) was previously proven to be highly effective in tumor cell killing, resulting in significant suppression of tumor growth in vivo. Recently, we developed a different form of RRV which is derived from another retrovirus, gibbon ape leukemia virus (GALV), as a cancer therapeutic agent. We compared the gene delivery efficiency and antitumor effects in the two types of RRV in experimental hepatocellular carcinoma (HCC). Our results show that both RRVs can efficiently spread throughout entire HCC cell populations in vitro and achieve high transduction efficiency in HCC xenografts in vivo, while GALV RRV, in general, exhibited more rapid replication kinetics in the tumors. In vitro, substantial HCC cell killing was achieved even when initially only 1% of the HCC cells were producing RRVs that express the yeast cytosine deaminase suicide gene, indicating that the high efficiency of gene transfer by replicative spread of RRVs greatly increased suicide gene toxicity. In vivo, GALV RRV-mediated suicide gene therapy efficiently suppressed HCC tumor growth and no detectable RRV signals were observed in extratumoral tissues, showing promise in using GALV RRV as a cancer therapeutic agent.


Assuntos
Carcinoma Hepatocelular/terapia , Vírus da Leucemia do Macaco Gibão/genética , Neoplasias Hepáticas Experimentais/terapia , Vírus Oncolíticos/genética , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biotransformação , Carcinoma Hepatocelular/patologia , Sobrevivência Celular/efeitos dos fármacos , Citosina Desaminase/biossíntese , Citosina Desaminase/genética , Flucitosina/metabolismo , Flucitosina/farmacologia , Flucitosina/uso terapêutico , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/genética , Terapia Genética , Células Hep G2 , Humanos , Vírus da Leucemia do Macaco Gibão/enzimologia , Vírus da Leucemia do Macaco Gibão/fisiologia , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Camundongos Nus , Terapia Viral Oncolítica , Vírus Oncolíticos/enzimologia , Vírus Oncolíticos/fisiologia , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Transdução Genética , Carga Tumoral/efeitos dos fármacos , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Int J Mol Med ; 25(5): 769-75, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20372821

RESUMO

Tumor-selective replicating viruses are attractive tools for cancer gene therapy, but generally achieve only transitory tumor suppression. However, replicating retrovirus vectors (RRVs) can achieve highly efficient and tumor-selective transduction, as well as persistent expression of transgenes. We therefore developed RRVs that express the yeast cytosine deaminase (yCD) and herpes simplex virus thymidine kinase (TK), which exhibit remarkably enhanced cytotoxicity after administration of the prodrugs 5-fluorocytosine (5-FC) and ganciclovir (GCV) concomitant with the efficiency of their replicative spread, and tested their therapeutic effect in vitro and in vivo. In subcutaneous MDA-MB-435 human breast cancer xenograft models, RRV-mediated yCD and TK suicide gene therapy significantly suppressed tumor growth after prodrug administration. Notably, no systemic spread of the vector to extratumoral tissues was detected. Our results thus demonstrate that efficient, tumor-selective, and stable integration achieved by RRVs causes efficient cell killing upon prodrug administration, resulting in significant suppression of tumor growth.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Neoplasias/genética , Retroviridae , Animais , Antimetabólitos/uso terapêutico , Antivirais/uso terapêutico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Feminino , Flucitosina/uso terapêutico , Ganciclovir/uso terapêutico , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Retroviridae/genética , Retroviridae/metabolismo , Timidina Quinase/genética , Timidina Quinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA