Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmacol Res ; 199: 106990, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37984506

RESUMO

Resistance to temozolomide (TMZ), the frontline chemotherapeutic agent for glioblastoma (GBM), has emerged as a formidable obstacle, underscoring the imperative to identify alternative therapeutic strategies to improve patient outcomes. In this study, we comprehensively evaluated a novel agent, O6-methyl-2'-deoxyguanosine-5'-triphosphate (O6-methyl-dGTP) for its anti-GBM activity both in vitro and in vivo. Notably, O6-methyl-dGTP exhibited pronounced cytotoxicity against GBM cells, including those resistant to TMZ and overexpressing O6-methylguanine-DNA methyltransferase (MGMT). Mechanistic investigations revealed that O6-methyl-dGTP could be incorporated into genomic DNA, disrupting nucleotide pools balance, and inducing replication stress, resulting in S-phase arrest and DNA damage. The compound exerted its anti-tumor properties through the activation of AIF-mediated apoptosis and the parthanatos pathway. In vivo studies using U251 and Ln229 cell xenografts supported the robust tumor-inhibitory capacity of O6-methyl-dGTP. In an orthotopic transplantation model with U87MG cells, O6-methyl-dGTP showcased marginally superior tumor-suppressive activity compared to TMZ. In summary, our research, for the first time, underscores the potential of O6-methyl-dGTP as an effective candidate against GBM, laying a robust scientific groundwork for its potential clinical adoption in GBM treatment regimens.


Assuntos
Glioblastoma , Polifosfatos , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Nucleosídeos/farmacologia , Nucleosídeos/uso terapêutico , Caspases , Linhagem Celular Tumoral , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Nucleotídeos , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/uso terapêutico , Desoxiguanosina/farmacologia , Desoxiguanosina/uso terapêutico , DNA , Resistencia a Medicamentos Antineoplásicos
2.
Neuro Oncol ; 25(11): 1976-1988, 2023 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-37279651

RESUMO

BACKGROUND: Temozolomide (TMZ) treatment efficacy in glioblastoma (GBM) has been limited by resistance. The level of O-6-methylguanine-DNA methyltransferase (MGMT) and intrinsic DNA damage repair factors are important for the TMZ response in patients. Here, we reported a novel compound, called EPIC-0307, that increased TMZ sensitivity by inhibiting specific DNA damage repair proteins and MGMT expression. METHODS: EPIC-0307 was derived by molecular docking screening. RNA immunoprecipitation (RIP), and chromatin immunoprecipitation by RNA (ChIRP) assays were used to verify the blocking effect. Chromatin immunoprecipitation (ChIP) and co-immunoprecipitation (Co-IP) assays were performed to explore the mechanism of EPIC-0307. A series of in vivo and in vitro experiments were designed to evaluate the efficacy of EPIC-0307 in sensitizing GBM cells to TMZ. RESULTS: EPIC-0307 selectively disrupted the binding of PRADX to EZH2 and upregulated the expression of P21 and PUMA, leading to cell cycle arrest and apoptosis in GBM cells. EPIC-0307 exhibited a synergistic inhibitory effect on GBM when combined with TMZ by downregulating TMZ-induced DNA damage repair responses and epigenetically silencing MGMT expression through modulating the recruitment of ATF3-pSTAT3-HDAC1 regulatory complex to the MGMT promoter. EPIC-0307 demonstrated significant efficacy in suppressing the tumorigenesis of GBM cells, restoring TMZ sensitivity. CONCLUSION: This study identified a potential small-molecule inhibitor (SMI) EPIC-0307 that selectively disrupted the PRADX-EZH2 interaction to upregulate expressions of tumor suppressor genes, thereby exerting its antitumor effects on GBM cells. EPIC-0307 treatment also increased the chemotherapeutic efficacy of TMZ by epigenetically downregulating DNA repair-associate genes and MGMT expression in GBM cells.


Assuntos
Glioblastoma , Humanos , Temozolomida/uso terapêutico , Glioblastoma/patologia , Antineoplásicos Alquilantes/uso terapêutico , Simulação de Acoplamento Molecular , Reparo do DNA , Enzimas Reparadoras do DNA/genética , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Metilases de Modificação do DNA/genética , RNA/farmacologia , RNA/uso terapêutico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteínas Supressoras de Tumor/genética
3.
BMC Complement Med Ther ; 23(1): 111, 2023 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-37024907

RESUMO

BACKGROUND: Despite rapid developments in immunotherapy and targeted therapy, dacarbazine (DTIC)-based chemotherapy still has been placed at the first-line for advanced melanoma patients who are after failure of immunotherapy or targeted therapy. However, the limited response rate and survival benefit challenge the DTIC-based chemotherapy for advanced melanoma patients. METHODS: Two melanoma cell lines, A375 and SK-MEL-28 were cultured with PA and DTIC over a range of concentrations for 72 h and the cell viabilities were detected by CCK8 assay. The Bliss model and ZIP model were used for calculating the synergistic effect of PA and DTIC. DNA double-strand breaks in the two cell lines were examined by the Comet assay, and cell apoptosis was analyzed by flow cytometry. The short hairpin RNA (shRNA)-mediated knockdown, Real-time polymerase chain reaction (RT-PCR) and Western blot were performed for molecular analysis. RESULTS: In the present study, we report that Protocatechuic aldehyde (PA) synergistically enhances the cytotoxicity of DTIC to two melanoma cell lines, A375 and SK-MEL-28. The combination of PA and DTIC augments DNA double-strand breaks and increases cell apoptosis. Further mechanism study reveals that PA destabilizes MGMT protein (O-6-Methylguanine-DNA Methyltransferase) through the ubiquitin-proteasome process and directly repairs DTIC-induced genetic lesions. Knockdown of MGMT compromises the synergistic effect between PA and DTIC. CONCLUSION: Our study demonstrates that the bioactive compound, Protocatechuic aldehyde, synergistically promotes the cytotoxicity of DTIC to melanoma cells through destabilization of MGMT protein. It could be a potential candidate for melanoma chemotherapy.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Apoptose , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/uso terapêutico , DNA/farmacologia , DNA/uso terapêutico
4.
ACS Appl Mater Interfaces ; 15(1): 106-119, 2023 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-36442077

RESUMO

Glioma is a deadly form of brain cancer, and the difficulty of treating glioma is exacerbated by the chemotherapeutic resistance developed in the tumor cells over the time of treatment. siRNA can be used to silence the gene responsible for the increased resistance, and sensitize the glioma cells to drugs. Here, iron oxide nanoparticles functionalized with peptides (NP-CTX-R10) were used to deliver siRNA to silence O6-methylguanine-DNA methyltransferase (MGMT) to sensitize tumor cells to alkylating drug, Temozolomide (TMZ). The NP-CTX-R10 could complex with siRNA through electrostatic interactions and was able to deliver the siRNA to different glioma cells. The targeting ligand chlorotoxin and cell penetrating peptide polyarginine (R10) enhanced the transfection capability of siRNA to a level comparable to commercially available Lipofectamine. The NP-siRNA was able to achieve up to 90% gene silencing. Glioma cells transfected with NP-siRNA targeting MGMT showed significantly elevated sensitivity to TMZ treatment. This nanoparticle formulation demonstrates the ability to protect siRNA from degradation and to efficiently deliver the siRNA to induce therapeutic gene knockdown.


Assuntos
Neoplasias Encefálicas , Glioma , Humanos , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , RNA Interferente Pequeno/farmacologia , Linhagem Celular Tumoral , Glioma/tratamento farmacológico , Glioma/genética , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Peptídeos/farmacologia , Nanopartículas Magnéticas de Óxido de Ferro , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Resistencia a Medicamentos Antineoplásicos
5.
Drug Deliv ; 30(1): 1-13, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36579448

RESUMO

Temozolomide (TMZ) is a conventional chemotherapeutic drug for glioma, however, its clinical application and efficacy is severely restricted by its drug resistance properties. O6-methylguanine-DNA methyltransferase (MGMT) is a DNA repair enzyme, which can repair the DNA damage caused by TMZ. A large number of clinical data show that reducing the expression of MGMT can enhance the chemotherapeutic efficacy of TMZ. Therefore, in order to improve the resistance of glioma to TMZ, an angiopep-2 (A2) modified nanoprodrug of polytemozolomide (P(TMZ)n) that combines with MGMT siRNA (siMGMT) targeting MGMT was developed (A2/T/D/siMGMT). It not only increased the amount of TMZ within tumor lesion site, but also reduced MGMT expression in glioma. The in vitro experiments indicated that the A2/T/D/siMGMT effectively enhanced the cellular uptake of TMZ and siMGMT, and resulted in a significant cell apoptosis and cytotoxicity in the glioma cells. The in vivo experiments showed that glioma growth was inhibited and the survival time of animals were prolonged remarkably after A2/T/D/siMGMT was injected via tail vein. The results showed that the therapeutic effect of A2/T/D/siMGMT in the treatment of glioma was significantly improved.


Assuntos
Neoplasias Encefálicas , Glioma , Animais , Temozolomida/farmacologia , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , RNA Interferente Pequeno/farmacologia , Linhagem Celular Tumoral , Glioma/tratamento farmacológico , Glioma/genética , Glioma/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Antineoplásicos Alquilantes/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética
6.
ACS Appl Mater Interfaces ; 14(34): 38617-38630, 2022 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-35974468

RESUMO

Chemo-/radioresistance is the most important reason for the failure of glioblastoma (GBM) treatment. Reversing the chemo-/radioresistance of GBM for boosting therapeutic efficacy is very challenging. Herein, we report a significant decrease in the chemo-/radioresistance of GBM by the in situ generation of SO2 within a tumor, which was released on demand from the prodrug 5-amino-1,3-dihydrobenzo[c]thiophene 2,2-dioxide (ATD) loaded on rare-earth-based scintillator nanoparticles (i.e., NaYF4:Ce@NaLuF4:Nd@ATD@DSPE-PEG5000, ScNPs) under X-ray irradiation. Our novel X-ray-responsive ScNPs efficiently converted highly penetrating X-rays into ultraviolet rays for controlling the decomposition of ATD to generate SO2, which effectively damaged the mitochondria of temozolomide-resistant U87 cells to lower the production of ATP and inhibit P-glycoprotein (P-gp) expression to reduce drug efflux. Meanwhile, the O6-methylguanine-DNA methyltransferase (MGMT) of drug-resistant tumor cells was also reduced to prevent the repair of damaged DNA and enhance cell apoptosis and the efficacy of chemo-/radiotherapy. The tumor growth was obviously suppressed, and the mice survived significantly longer than untreated temozolomide-resistant GBM-bearing mice. Our work demonstrates the potential of SO2 in reducing chemo-/radioresistance to improve the therapeutic effect against resistant tumors if it can be well controlled and in situ generated in tumor cells. It also provides insights into the rational design of stimuli-responsive drug delivery systems for the controlled release of drugs.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Animais , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/metabolismo , Camundongos , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico
7.
ACS Nano ; 16(5): 7409-7427, 2022 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-35549164

RESUMO

Glioblastoma (GBM) is an invasive cancer with high mortality in central nervous system. Resistance to temozolomide (TMZ) and immunosuppressive microenvironment lead to low outcome of the standardized treatment for GBM. In this study, a 2-deoxy-d-glucose modified lipid polymer nanoparticle loaded with TMZ and siPD-L1 (TMZ/siPD-L1@GLPN/dsb) was prepared to reprogram the TMZ-resistant and immunosuppressive microenvironment in orthotopic GBM. TMZ/siPD-L1@GLPN/dsb simultaneously delivered a large amount of TMZ and siPD-L1 to the deep area of the orthotopic TMZ-resistant GBM tissue. By inhibiting PD-L1 protein expression, TMZ/siPD-L1@GLPN/dsb markedly augmented the percentage of CD3+CD8+IFN-γ+ cells (Teff cells) and reduced the percentage of CD4+CD25+FoxP3+ cells (Treg cells) in orthotopic TMZ-resistant GBM tissue, which enhanced T-cell mediated cytotoxicity on orthotopic TMZ-resistant GBM. Moreover, TMZ/siPD-L1@GLPN/dsb obviously augmented the sensitivity of orthotopic TMZ-resistant GBM to TMZ through decreasing the protein expression of O6-methyl-guanine-DNA methyltransferase (MGMT) in TMZ-resistant GBM cells. Thus, TMZ/siPD-L1@GLPN/dsb markedly restrained the growth of orthotopic TMZ-resistant GBM and extended the survival time of orthotopic GBM rats through reversing a TMZ-resistant and immunosuppressive microenvironment. TMZ/siPD-L1@GLPN/dsb shows potential application to treat orthotopic TMZ-resistant GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Animais , Ratos , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/genética , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Sistemas de Liberação de Fármacos por Nanopartículas
8.
Neurochem Int ; 157: 105348, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35490896

RESUMO

BACKGROUND: Temozolomide (TMZ) is generally applied for glioma treatment, while drug resistance of TMZ limits its therapeutic efficacy. Mannose exerts evident anti-tumor effect. We intended to investigate whether mannose enhanced TMZ sensitivity to glioma and examined the underlying mechanism. METHODS: MTT and clone formation assays were performed to detect cell viability and proliferation. Cell apoptosis was measured by flow cytometry. The protein and gene expression levels were detected by Western blot and qRT-PCR assays. Xenograft glioma model was established to explore the influence of mannose in vivo. RESULTS: Mannose inhibited glioma cell growth, which was facilitated by knockdown of phosphomannose isomerase (PMI) while reversed by overexpression of PMI. Mannose enhanced the sensitivity of glioma cells to TMZ, indicated by the further inhibited cell viability and colony formation and the aggravated cell apoptosis, which was reversed by overexpression of O6-methylguanine DNA methyltransferase (MGMT). Furthermore, mannose and TMZ inhibited MGMT expression and Wnt/ß-catenin activation. Moreover, activating Wnt/ß-catenin pathway blocked anti-proliferative effect induced by mannose and TMZ, which was further suppressed by overexpressed MGMT. Mannose inhibited glioma growth, suppressed Ki67 and downregulated MGMT and ß-catenin in vivo. CONCLUSION: Mannose inhibited MGMT to enhance sensitivity of glioma cells to TMZ, with Wnt/ß-catenin pathway involvement. Our data suggested that mannose could be an innovative agent to improve glioma treatment, particularly in TMZ-resistant glioma with high MGMT.


Assuntos
Neoplasias Encefálicas , Glioma , Apoptose , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Humanos , Manose/farmacologia , Manose/uso terapêutico , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Via de Sinalização Wnt , beta Catenina/metabolismo
9.
Cell Death Dis ; 13(4): 293, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35365623

RESUMO

Overexpression of histone deacetylases (HDACs) in cancer commonly causes resistance to genotoxic-based therapies. Here, we report on the novel mechanism whereby overexpressed class I HDACs increase the resistance of glioblastoma cells to the SN1 methylating agent temozolomide (TMZ). The chemotherapeutic TMZ triggers the activation of the DNA damage response (DDR) in resistant glioma cells, leading to DNA lesion bypass and cellular survival. Mass spectrometry analysis revealed that the catalytic activity of class I HDACs stimulates the expression of the E3 ubiquitin ligase RAD18. Furthermore, the data showed that RAD18 is part of the O6-methylguanine-induced DDR as TMZ induces the formation of RAD18 foci at sites of DNA damage. Downregulation of RAD18 by HDAC inhibition prevented glioma cells from activating the DDR upon TMZ exposure. Lastly, RAD18 or O6-methylguanine-DNA methyltransferase (MGMT) overexpression abolished the sensitization effect of HDAC inhibition on TMZ-exposed glioma cells. Our study describes a mechanism whereby class I HDAC overexpression in glioma cells causes resistance to TMZ treatment. HDACs accomplish this by promoting the bypass of O6-methylguanine DNA lesions via enhancing RAD18 expression. It also provides a treatment option with HDAC inhibition to undermine this mechanism.


Assuntos
Neoplasias Encefálicas , Glioma , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Glioma/tratamento farmacológico , Glioma/genética , Glioma/metabolismo , Histona Desacetilases/farmacologia , Humanos , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Ubiquitina-Proteína Ligases/farmacologia
10.
ACS Chem Biol ; 15(4): 1004-1015, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32162909

RESUMO

Most cell behaviors are the outcome of processing information from multiple signals generated upon cell stimulation. Thus, a systematic understanding of cellular systems requires methods that allow the activation of more than one specific signaling molecule or pathway within a cell. However, the construction of tools suitable for such multiplexed signal control remains challenging. In this work, we aimed to develop a platform for chemically manipulating multiple signaling molecules/pathways in living mammalian cells based on self-localizing ligand-induced protein translocation (SLIPT). SLIPT is an emerging chemogenetic tool that controls protein localization and cell signaling using synthetic self-localizing ligands (SLs). Focusing on the inner leaflet of the plasma membrane (PM), where there is a hub of intracellular signaling networks, here we present the design and engineering of two new PM-specific SLIPT systems based on an orthogonal eDHFR and SNAP-tag pair. These systems rapidly induce translocation of eDHFR- and SNAP-tag-fusion proteins from the cytoplasm to the PM specifically in a time scale of minutes upon addition of the corresponding SL. We then show that the combined use of the two systems enables chemically inducible, individual translocation of two distinct proteins in the same cell. Finally, by integrating the orthogonal SLIPT systems with fluorescent reporters, we demonstrate simultaneous multiplexed activation and fluorescence imaging of endogenous ERK and Akt activities in a single cell. Collectively, orthogonal PM-specific SLIPT systems provide a powerful new platform for multiplexed chemical signal control in living single cells, offering new opportunities for dissecting cell signaling networks and synthetic cell manipulation.


Assuntos
Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Membrana/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Transporte Proteico/efeitos dos fármacos , Pirimidinas/farmacologia , Tetra-Hidrofolato Desidrogenase/farmacologia , Membrana Celular/metabolismo , Escherichia coli/enzimologia , Células HeLa , Humanos , Proteínas de Membrana/genética , O(6)-Metilguanina-DNA Metiltransferase/química , O(6)-Metilguanina-DNA Metiltransferase/genética , Engenharia de Proteínas , Pirimidinas/química , Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/genética
11.
Proc Natl Acad Sci U S A ; 111(1): 409-14, 2014 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-24367102

RESUMO

A hypoxic microenvironment induces resistance to alkylating agents by activating targets in the mammalian target of rapamycin (mTOR) pathway. The molecular mechanisms involved in this mTOR-mediated hypoxia-induced chemoresistance, however, are unclear. Here we identify the mTOR target N-myc downstream regulated gene 1 (NDRG1) as a key determinant of resistance toward alkylating chemotherapy, driven by hypoxia but also by therapeutic measures such as irradiation, corticosteroids, and chronic exposure to alkylating agents via distinct molecular routes involving hypoxia-inducible factor (HIF)-1alpha, p53, and the mTOR complex 2 (mTORC2)/serum glucocorticoid-induced protein kinase 1 (SGK1) pathway. Resistance toward alkylating chemotherapy but not radiotherapy was dependent on NDRG1 expression and activity. In posttreatment tumor tissue of patients with malignant gliomas, NDRG1 was induced and predictive of poor response to alkylating chemotherapy. On a molecular level, NDRG1 bound and stabilized methyltransferases, chiefly O(6)-methylguanine-DNA methyltransferase (MGMT), a key enzyme for resistance to alkylating agents in glioblastoma patients. In patients with glioblastoma, MGMT promoter methylation in tumor tissue was not more predictive for response to alkylating chemotherapy in patients who received concomitant corticosteroids.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Proteínas de Ciclo Celular/metabolismo , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioma/tratamento farmacológico , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Reparo do DNA , Glioblastoma/metabolismo , Glioma/metabolismo , Humanos , Hipóxia , Immunoblotting , Lentivirus/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Plasmídeos/metabolismo , Fatores de Tempo
12.
Clin Cancer Res ; 15(2): 502-10, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19147755

RESUMO

PURPOSE: Despite objective response rates of only approximately 13%, temozolomide remains one of the most effective single chemotherapy agents against metastatic melanoma, second only to dacarbazine, the current standard of care for systemic treatment of melanoma. The goal of this study was to identify molecular and/or genetic markers that correlate with, and could be used to predict, response to temozolomide-based treatment regimens and that reflect the intrinsic properties of a patient's tumor. EXPERIMENTAL DESIGN: Using a panel of 26 human melanoma-derived cell lines, we determined in vitro temozolomide sensitivity, O(6)-methylguanine-DNA methyltransferase (MGMT) activity, MGMT protein expression and promoter methylation status, and mismatch repair proficiency, as well as the expression profile of 38,000 genes using an oligonucleotide-based microarray platform. RESULTS: The results showed a broad spectrum of temozolomide sensitivity across the panel of cell lines, with IC(50) values ranging from 100 micromol/L to 1 mmol/L. There was a significant correlation between measured temozolomide sensitivity and a gene expression signature-derived prediction of temozolomide sensitivity (P < 0.005). Notably, MGMT alone showed a significant correlation with temozolomide sensitivity (MGMT activity, P < 0.0001; MGMT expression, P

Assuntos
Antineoplásicos Alquilantes/farmacologia , Dacarbazina/análogos & derivados , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Área Sob a Curva , Linhagem Celular Tumoral , Metilação de DNA , Dacarbazina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genômica , Humanos , Concentração Inibidora 50 , Repetições de Microssatélites , Metástase Neoplásica , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Temozolomida
13.
Carcinogenesis ; 28(5): 1111-6, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17116724

RESUMO

The DNA repair protein O(6)-methylguanine-DNA methyltransferase (MGMT) protects from toxicity and mutations incurred following alkylating agents by removing O(6)-alkylguanine lesions. Using Mgmt-/- mice, we examined MGMT's role in protecting from in vivo mutations induced by three different alkylating agents, temozolomide (TMZ), 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) and cyclophosphamide. Mutant frequencies were determined in the hypoxanthine-guanine phosphoribosyltransferase gene of splenic T-lymphocytes from C57BL/6 mice (Mgmt+/+ and Mgmt-/-) following TMZ, BCNU or cyclophosphamide. Following TMZ, the mutation frequency was significantly greater in Mgmt-/- mice (5.5 and 9.8 x 10(-6) for 7 and 10 mg/kg TMZ, respectively) compared with vehicle-treated mice (1.0 x 10(-6), P

Assuntos
Alquilantes/toxicidade , Mutação , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Animais , Carmustina/toxicidade , Linhagem Celular Tumoral , Ciclofosfamida/toxicidade , Dacarbazina/análogos & derivados , Dacarbazina/toxicidade , Guanina/análogos & derivados , Guanina/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Testes de Mutagenicidade , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Temozolomida , Transfecção
14.
ACS Chem Biol ; 1(9): 575-84, 2006 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-17168553

RESUMO

We introduce a strategy for evolving protein substrate specificity by the insertion of random amino acid loops into the protein backbone. Application of this strategy to human O6-alkylguanine-DNA alkyltransferase (AGT) led to the isolation of mutants that react with the non-natural substrate O6-propargylguanine. Libraries generated by conventional random or targeted saturation mutagenesis, by contrast, did not yield any mutants with activity towards this new substrate. The strategy of loop insertion to alter enzyme specificity should be general and applicable to other classes of proteins. An important application of the isolated AGT mutant is in molecular imaging, where the mutant and parental AGTs are used to label two different AGT fusion proteins with different fluorophores in the same living cell or in vitro . This allowed the establishment of fluorescence-based assays to detect protein-protein interactions and measure enzymatic activities.


Assuntos
Microscopia de Fluorescência/métodos , Mutagênese Sítio-Dirigida , O(6)-Metilguanina-DNA Metiltransferase/química , Sequência de Aminoácidos , Sequência de Bases , Transferência Ressonante de Energia de Fluorescência , Humanos , Cinética , Modelos Químicos , Dados de Sequência Molecular , Mutação , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Biblioteca de Peptídeos , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Especificidade por Substrato
15.
J Neurosurg ; 101(4): 659-63, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15481722

RESUMO

OBJECT: The DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) inhibits the cytotoxic effect of alkylating agents on tumor cells. The presence of two nonconsensus glucocorticoid-responsive elements in the human MGMT promoter region indicates the potential regulation of MGMT expression by glucocorticoid agents. This study was performed to elucidate whether dexamethasone affects the expression of MGMT in glioblastoma multiforme (GBM) cells, thereby limiting the benefit of chemotherapeutic alkylating agents. METHODS: Four GBM cell lines (A172, T98G, U138MG, and U87MG) were exposed to the alkylating agent 1-(4-amino-2-methyl-5-pyrimidinyl) methyl-3-(2-chloroethyl)-3-nitrosourea hydrochloride (ACNU) with or without dexamethasone. The expression levels of MGMT were correlated with the cytotoxic effects of ACNU in GBM cells. In the presence of ACNU alone, dexamethasone alone, and the combination of both agents, messenger RNA expression of MGMT was induced to varying degrees with the highest increases seen in the later conditions. This dexamethasone-dependent induction of the MGMT gene was even observed in U87MG cells in which the promoter is methylated, although the absolute expression of MGMT mRNA was the lowest in that cell line. The induction of MGMT by dexamethasone was associated with an increased resistance of these cells to ACNU. CONCLUSIONS: These results indicate that dexamethasone-mediated upregulation of MGMT limits the efficiency of alkylating agents in the treatment of malignant gliomas.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias Encefálicas/genética , Dexametasona/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Antineoplásicos/farmacologia , Neoplasias Encefálicas/patologia , Resistencia a Medicamentos Antineoplásicos , Indução Enzimática , Glioblastoma/patologia , Humanos , Nimustina/farmacologia , RNA Mensageiro/análise , Células Tumorais Cultivadas , Regulação para Cima
16.
Mol Cancer Ther ; 3(10): 1215-20, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15486188

RESUMO

Temozolomide is an alkylating agent that mediates its cytotoxic effects via O(6)-methylguanine (O(6)-meG) adducts in DNA. O(6)-alkylguanine-DNA-alkyltransferase (MGMT) can repair such adducts and therefore constitutes a major resistance mechanism to the drug. MGMT activity can be attenuated in vitro and in vivo by the pseudosubstrate O(6)-(4-bromothenyl)guanine (PaTrin-2, Patrin, Lomeguatrib), which in clinical trials is in combination with temozolomide. Resistance to cytotoxic agents can also be mediated by the Bcl-2 protein, which inhibits apoptosis and is frequently up-regulated in tumor cells. Attenuation of Bcl-2 expression can be affected by treatment of cells with the antisense oligonucleotide, oblimersen sodium (Genasense), currently in phase III clinical trials in combination with the methylating agent dacarbazine. Using a human ovarian cancer cell line (A2780) that expresses both Bcl-2 and MGMT, we show that cells treated with active dose levels of either oblimersen (but not control reverse sequence or mismatch oligonucleotides) or PaTrin-2 are substantially sensitized to temozolomide. Furthermore, the exposure of oblimersen-pretreated cells to PaTrin-2 leads to an even greater sensitization of these cells to temozolomide. Thus, growth of cells treated only with temozolomide (5 microg/mL) was 91% of control growth, whereas additional exposure to PaTrin-2 alone (10 micromol/L) or oblimersen alone (33 nmol/L) reduced this to 81% and 66%, respectively, and the combination of PaTrin-2 (10 micromol/L) and oblimersen (33 nmol/L) reduced growth to 25% of control. These results suggest that targeting both Bcl-2 with oblimersen and MGMT with PaTrin-2 would markedly enhance the antitumor activity of temozolomide and merits testing in clinical trials.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Reparo do DNA , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Humanos , Oligonucleotídeos/química , Neoplasias Ovarianas/tratamento farmacológico , Temozolomida , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Tionucleotídeos/farmacologia , Fatores de Tempo , Regulação para Cima
17.
Cancer Epidemiol Biomarkers Prev ; 13(2): 320-3, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14973087

RESUMO

PURPOSE: The objective of the study was to examine the association of three exon 5 variants in the O(6)-alkylguanine DNA alkyltransferase (AGT) gene involved in the repair of the mutagenic DNA lesion O(6)-alkylguanine formed by nitrosamines, with lung cancer risk in never-smokers. EXPERIMENTAL DESIGN: Exon 5 of the AGT gene was sequenced in genomic DNA from 136 cases and 133 hospital- or population-based controls for whom questionnaire information on second-hand smoke and diet was available to determine the frequencies of the Gly(160)Arg, Ile(143)Val, and Lys(178)Arg variant alleles. RESULTS: No codon (160)Arg variant alleles were found in the study population. The codon (143)Val and (178)Arg variant alleles, present at allele frequencies of 0.07, showed 100% linkage. The odds ratio (OR) of lung cancer for these variant carriers was 2.05 [95% confidence interval (CI) 1.03-4.07]. The risk varied between the different lung cancer pathologies with an increased risk for adenocarcinoma (OR 2.67, 95% CI 1.21-5.87) or small cell carcinoma (OR 4.83, 95% CI 0.91-25.7) but not for squamous cell carcinoma (OR 1.07, 95% CI 0.27-4.18). Compared with individuals carrying the mutant alleles unexposed to second-hand smoke, the OR for exposed variant carriers was 1.95 (95% CI 0.53-1.15); a similar interaction, although not significative, was observed for low consumption of cruciferous vegetables and for green vegetables and tomatoes. CONCLUSIONS: These results point toward a role of AGT polymorphisms in lung cancer susceptibility among never-smokers, in particular among subjects exposed to environmental carcinogens.


Assuntos
Adenocarcinoma/etiologia , Adenocarcinoma/genética , Carcinoma Pulmonar de Células não Pequenas/etiologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma de Células Escamosas/etiologia , Carcinoma de Células Escamosas/genética , Predisposição Genética para Doença , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/genética , O(6)-Metilguanina-DNA Metiltransferase/genética , Polimorfismo Genético , Poluição por Fumaça de Tabaco/efeitos adversos , Idoso , Alelos , Estudos de Casos e Controles , Dieta , Éxons , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Reação em Cadeia da Polimerase , Medição de Risco , Análise de Sequência de DNA
18.
J Hematother Stem Cell Res ; 12(4): 375-87, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12965075

RESUMO

Optimized hematopoietic gene therapy requires vectors with strong expression in the desired target cell population and the ability to select for the expressing transduced cells. In the context of drug resistance selection of repopulating hematopoietic stem cells in the mouse, we examined tissue expression after transduced marrow transplantation of the drug selection gene, G156A mutant O6-methylguanine-DNA methyltransferase (G156A MGMT). To gain more experience with the rigor of the impact of selection on tissue-specific gene expression, we also asked whether there are expression differences between three different onco-retroviral backbones--MPSV, SF, and MFG. MGMT expression was compared after O6-benzylguanine (BG) and 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) drug selection in vivo. After mice were transplanted with cells transduced with MPSV, MFG, or SF retroviral vectors expressing G156A MGMT and drug treated, nearly complete replacement by transduced progenitors was observed in the marrow. Each backbone supported MGMT expression in all four hematopoietic lineages in vivo indicating that MGMT-mediated selection is indeed robust. Expression in marrow, spleen, and thymus was very similar between the vectors and differences were most likely due to differences in gene copy number per selected cell. In primary and secondary recipients, the highest expression was observed in MFG and this was the vector that transduced at the greatest proviral copy number per cell. These data indicate that strong selection pressure using the MGMT gene to protect primary and secondary repopulating murine stem cells from the toxicity of BCNU. Regardless of the vector backbone used, multiorgan expression was observed without evidence of gene silencing. These data help establish mutant, BG-resistant MGMT as a potent selection gene for stem cell selection in vivo.


Assuntos
Guanina/análogos & derivados , Células-Tronco Hematopoéticas/metabolismo , Mutação , O(6)-Metilguanina-DNA Metiltransferase/genética , Proteínas Oncogênicas de Retroviridae/genética , Transgenes , Animais , Antineoplásicos/farmacologia , Células da Medula Óssea/citologia , Linhagem Celular , Linhagem da Célula , Separação Celular , Clonagem Molecular , Relação Dose-Resposta a Droga , Citometria de Fluxo , Inativação Gênica , Vetores Genéticos , Genoma , Guanina/farmacologia , Humanos , Células K562 , Camundongos , Camundongos Endogâmicos C3H , Modelos Genéticos , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Reação em Cadeia da Polimerase , Células-Tronco/citologia , Fatores de Tempo
19.
Cancer Gene Ther ; 10(8): 603-10, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12872142

RESUMO

Chemotherapeutic DNA alkylating agents are common weapons employed to fight both pediatric and adult cancers. In addition to cancerous cells, nontarget tissues are subjected to the cytotoxicity of these agents, and dose-limiting toxicity in the form of myelosuppression is a frequent result of treatment. One approach to prevent myelosuppression that results from the use of chemotherapeutic agents is to increase the levels of DNA repair proteins in bone marrow cells. Here we report our second successful attempt to create a fusion protein that possesses both direct reversal and base excision repair pathway DNA repair activities. The chimeric protein is composed of the human O(6)-Methylguanine-DNA Methyltransferase (MGMT) and the yeast Apn1 proteins and retains both MGMT and AP endonuclease activities as determined by biochemical analysis. We have also demonstrated that the chimeric protein is able to protect mammalian cells from the DNA alkylating agents 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) and methyl methanesulfonate (MMS). The protection by the chimeric protein against BCNU is even greater than MGMT alone, which has potential translational significance given that MGMT is currently in clinical trials. Additionally, we show that the chimeric MGMT-Apn1 protein can protect mammalian cells from dual treatments of BCNU and MMS and that this effect is greater than that provided by MGMT alone. The data support our previous finding that a protein with multiple DNA repair activities can be constructed and that this and other constructs may play an important clinical role in guarding against dose-limiting effects of chemotherapy, particularly in situations of multiple drug use.


Assuntos
Alquilantes/efeitos adversos , Reparo do DNA , Endodesoxirribonucleases/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Proteínas de Saccharomyces cerevisiae/farmacologia , Carmustina/efeitos adversos , Sobrevivência Celular , Citoproteção , Enzimas Reparadoras do DNA , Humanos , Células K562 , Metanossulfonato de Metila/farmacologia
20.
J Chemother ; 15(2): 173-83, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12797396

RESUMO

O6-alkylguanine-DNA alkyltransferase (OGAT) and the mismatch repair system (MRS) play a crucial role in the susceptibility of tumor cells to the cytotoxic effects of agents that generate O6-methylguanine in DNA, including the triazene compound temozolomide (TMZ). Studies performed with peripheral blood mononuclear cells (MNC) showed that TMZ was scarcely active on lymphocyte functions not dependent on cell proliferation (e.g. NK activity and cytokine-mediated induction of CD1b molecule in adherent MNC). In contrast, TMZ depressed proliferation and lymphokine activated killer (LAK) cell generation in response to IL-2. In this case, a reasonably good inverse relationship was found between OGAT levels of MNC and their susceptibility to TMZ. This study also analyzed the ratio of the toxic effect of TMZ on MNC and on tumor cells (i.e. "Tumor-Immune Function Toxicity Index", TIFTI). A particularly favorable TIFTI can be obtained when OGAT levels are extremely high in MNC and markedly low in tumor cells. This holds true for MRS-proficient neoplastic cells, but not for MRS-deficient tumors. In conclusion, strategies aimed at modulating OGAT and MRS may improve the clinical response to TMZ. However, the use of OGAT inhibitors to potentiate the antitumor activity of TMZ might result in a concomitant increase of the immunosuppressive effects of the drug, thus reducing the relative TIFTI.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Reparo do DNA , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Linfoma de Burkitt/patologia , Divisão Celular , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Humanos , Interleucina-2/farmacologia , Células Matadoras Ativadas por Linfocina , Leucemia Eritroblástica Aguda/patologia , Leucócitos Mononucleares/fisiologia , Linfócitos/fisiologia , Melanoma/patologia , O(6)-Metilguanina-DNA Metiltransferase/efeitos dos fármacos , Neoplasias Cutâneas/patologia , Temozolomida , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...