Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Sci Rep ; 11(1): 21133, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702850

ABSTRACT

Chemotherapeutic drugs such as the alkylating agent Temozolomide (TMZ), in addition to reducing tumor mass, can also sensitize tumors to immune recognition by transient upregulation of multiple stress induced NKG2D ligands (NKG2DL). However, the potential for an effective response by innate lymphocyte effectors such as NK and γδ T cells that recognize NKG2DL is limited by the drug's concomitant lymphodepleting effects. We have previously shown that modification of γδ T cells with a methylguanine DNA methyltransferase (MGMT) transgene confers TMZ resistance via production of O6-alkylguanine DNA alkyltransferase (AGT) thereby enabling γδ T cell function in therapeutic concentrations of TMZ. In this study, we tested this strategy which we have termed Drug Resistant Immunotherapy (DRI) to examine whether combination therapy of TMZ and MGMT-modified γδ T cells could improve survival outcomes in four human/mouse xenograft models of primary and refractory GBM. Our results confirm that DRI leverages the innate response of γδ T cells to chemotherapy-induced stress associated antigen expression and achieves synergies that are significantly greater than either individual approach.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Immunotherapy , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocytes , Temozolomide/pharmacology , Transgenes , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Humans , Mice, Nude , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/economics , T-Lymphocytes/enzymology , T-Lymphocytes/transplantation , Xenograft Model Antitumor Assays
2.
Tumour Biol ; 36(8): 5839-48, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25716203

ABSTRACT

Emerging evidence indicates that O(6)-methylguanine-DNA methyltransferase (MGMT) is a candidate for tumor suppression in several types of human tumors including colorectal cancer (CRC). However, the correlation between MGMT hypermethylation and clinicopathological characteristics of CRC remains unclear. In this study, we conducted a systematic review and meta-analysis to quantitatively evaluate the effects of MGMT hypermethylation on the incidence of CRC and clinicopathological characteristics. A comprehensive literature search was done from Web of Science, the Cochrane Library Database, PubMed, EMBASE, CINAHL, and the Chinese Biomedical Database for related research publications written in English and Chinese. Methodological quality of the studies was also evaluated. Analyses of pooled data were performed with Review Manager 5.2. Odds ratio (OR) and hazard ratio (HR) were calculated and summarized, respectively. Final analysis from 28 eligible studies was performed. MGMT hypermethylation is found to be significantly higher in CRC than in normal colorectal mucosa, the pooled OR from 13 studies including 1085 CRC and 899 normal colorectal mucosa, OR = 6.04, 95 % confidence interval (CI) = 4.69-7.77, p < 0.00001. MGMT hypermethylation is also significantly higher in colorectal adenoma than in normal colorectal mucosa, but it is significantly less compared to that in CRC patients. Interestingly, MGMT hypermethylation is correlated with sex status and is significantly higher in female than in male. MGMT hypermethylation is also associated with high levels of microsatellite instability (MSI). The pooled HR for overall survival (OS) shows that MGMT hypermethylation is not associated with worse survival in CRC patients. The results of this meta-analysis suggest that MGMT hypermethylation is associated with an increased risk and high levels of MSI and may play an important role in CRC initiation. However, MGMT hypermethylation may play an important role in the early stage of CRC progression and development, as well as having limited value in prediction of prognosis in CRC patients. We also discussed that MGMT may serve as a potential drug target of CRC.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation/genetics , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Prognosis , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Guanine/analogs & derivatives , Guanine/metabolism , Humans , Mutation , Neoplasm Staging , O(6)-Methylguanine-DNA Methyltransferase/genetics , Promoter Regions, Genetic
3.
Virchows Arch ; 465(5): 579-86, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25031012

ABSTRACT

The O (6)-methylguanine-DNA-methyltransferase (MGMT) gene encodes for a DNA repairing enzyme of which silencing by promoter methylation is involved in brain tumorigenesis. MGMT promoter methylation represents a favorable prognostic factor and has been associated with a better response to alkylating agents in glioma and systemic lymphoma. Primary central nervous system lymphoma (PCNSL) is a rare and aggressive extranodal malignant lymphoma. The current standard of care, based on high-dose methotrexate chemotherapy, has improved prognosis but outcome remains poor for a majority of patients. Therapeutic progress in this field is conditioned by limited biological and molecular knowledge about the disease. Temozolomide has recently emerged as an alternative option for PCNSL treatment. We aimed to analyze the MGMT gene methylation status in a series of 24 PCNSLs, to investigate the relationship between methylation status of the gene and immunohistochemical expression of MGMT protein and to evaluate the possible prognostic significance of these biomarkers. Our results confirm that methylation of the MGMT gene and loss of MGMT protein are frequent events in these lymphomas (54 % of our cases) and suggest that they are gender and age related. MGMT methylation showed high correlation with loss of protein expression (concordance correlation coefficient = -0.49; Fisher exact test: p < 0.01), different from what has been observed in other brain tumors. In the subgroup of ten patients who received high dose chemotherapy, the presence of methylated MGMT promoter (n = 4), seems to be associated with a prolonged overall survival (>60 months in three of four patients). The prognostic significance of these molecular markers in PCNSL needs to be further studied in groups of patients treated in a homogeneous way.


Subject(s)
Central Nervous System Neoplasms/metabolism , Lymphoma/metabolism , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/genetics , Promoter Regions, Genetic , Adult , Aged , Biomarkers, Tumor/metabolism , Central Nervous System Neoplasms/genetics , DNA Methylation , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Female , Humans , Immunohistochemistry , Lymphoma/genetics , Male , Middle Aged , Temozolomide
4.
Neuropathology ; 34(3): 268-76, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24397721

ABSTRACT

Temozolomide (TMZ) is an oral alkylating agent which is widely used in the treatment of glioblastoma (GBM) and is composed of astrocytic and/or oligodendroglial tumors, and the evaluation of O(6) -methylguanine DNA methyltransferase (MGMT) expression is important to predict the response to TMZ therapy. In this study, we conducted immunohistochemical analysis of 117 cases of Japanese GBM including 19 cases of GBM with oligodendroglioma component (GBMO), using a scoring system for quantitative evaluation of staining intensity and proportion of MGMT, and performed survival analysis of these patients. Immunohistochemically, 55 cases (47%) were positive for MGMT with various intensities and proportions (total score (TS) ≥ 2), while 62 cases (53%) were negative (TS = 0). The distribution of MGMT expression pattern was not affected by any clinicopathological parameters such as the histological subtype (GBM vs. GBMO), age and gender. The survival analysis of these patients revealed that the minimal expression of MGMT (TS ≥ 2) was a significant unfavorable prognostic factor (P < 0.001) as well as resectability (P = 0.004). Moreover, multivariate analysis showed that minimal MGMT expression in GBM was the most potent independent predictor for progression free survival (P < 0.001) and also overall patient survival (P < 0.001). This is the first report employing the scoring system for both staining intensity and proportion to evaluate immunohistochemical MGMT expression in GBM. In addition, our results emphases the clinicopathological values of the immunohistochemical approach for MGMT expression in glioma patients as a routine laboratory examination.


Subject(s)
Biomarkers, Tumor/biosynthesis , Brain Neoplasms/enzymology , Gene Expression Regulation, Neoplastic , Glioblastoma/enzymology , O(6)-Methylguanine-DNA Methyltransferase/analysis , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Female , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Male , Middle Aged , Survival Rate/trends
6.
Nucleic Acids Res ; 40(16): 7806-20, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22669901

ABSTRACT

An adaptive response to alkylating agents based upon the conformational change of a methylphosphotriester (MPT) DNA repair protein to a transcriptional activator has been demonstrated in a number of bacterial species, but this mechanism appears largely absent from eukaryotes. Here, we demonstrate that the human pathogen Aspergillus fumigatus elicits an adaptive response to sub-lethal doses of the mono-functional alkylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). We have identified genes that encode MPT and O(6)-alkylguanine DNA alkyltransferase (AGT) DNA repair proteins; deletions of either of these genes abolish the adaptive response and sensitize the organism to MNNG. In vitro DNA repair assays confirm the ability of MPT and AGT to repair methylphosphotriester and O(6)-methylguanine lesions respectively. In eukaryotes, the MPT protein is confined to a select group of fungal species, some of which are major mammalian and plant pathogens. The evolutionary origin of the adaptive response is bacterial and rooted within the Firmicutes phylum. Inter-kingdom horizontal gene transfer between Firmicutes and Ascomycete ancestors introduced the adaptive response into the Fungal kingdom. Our data constitute the first detailed characterization of the molecular mechanism of the adaptive response in a lower eukaryote and has applications for development of novel fungal therapeutics targeting this DNA repair system.


Subject(s)
Alkylating Agents/toxicity , Aspergillus fumigatus/genetics , DNA Repair , Methyltransferases/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Adaptation, Physiological , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/enzymology , DNA Damage , Gene Deletion , Methylnitronitrosoguanidine/toxicity , Methyltransferases/metabolism , Methyltransferases/physiology , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Phylogeny
7.
Oncol Rep ; 28(2): 654-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22614944

ABSTRACT

The O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation status is a predictive parameter for the response of malignant gliomas to alkylating agents such as temozolomide. First clinical trials with temozolomide plus bevacizumab therapy in metastatic melanoma patients are ongoing, although the predictive value of the MGMT promoter methylation status in this setting remains unclear. We assessed MGMT promoter methylation in formalin-fixed, primary tumor tissue of metastatic melanoma patients treated with first-line temozolomide and bevacizumab from the trial SAKK 50/07 by methylation-specific polymerase chain reaction. In addition, the MGMT expression levels were also analyzed by MGMT immunohistochemistry. Eleven of 42 primary melanomas (26%) revealed a methylated MGMT promoter. Promoter methylation was significantly associated with response rates CR + PR versus SD + PD according to RECIST (response evaluation criteria in solid tumors) (p<0.05) with a trend to prolonged median progression-free survival (8.1 versus 3.4 months, p>0.05). Immunohistochemically different protein expression patterns with heterogeneous and homogeneous nuclear MGMT expression were identified. Negative MGMT expression levels were associated with overall disease stabilization CR+PR+SD versus PD (p=0.05). There was only a poor correlation between MGMT methylation and lack of MGMT expression. A significant proportion of melanomas have a methylated MGMT promoter. The MGMT promoter methylation status may be a promising predictive marker for temozolomide therapy in metastatic melanoma patients. Larger sample sizes may help to validate significant differences in survival type endpoints.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA Methylation , Melanoma/drug therapy , Melanoma/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Epigenomics , Female , Humans , Immunohistochemistry , Male , Melanoma/enzymology , Melanoma/pathology , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Predictive Value of Tests , Promoter Regions, Genetic , Temozolomide , Treatment Outcome
8.
Cancer ; 118(18): 4545-54, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22359215

ABSTRACT

BACKGROUND: O(6) -methylguanine-DNA methyltransferase (MGMT) promoter methylation status was proposed as a prognostic biomarker for patients with glioblastoma. However, the prognostic impact of MGMT in patients with newly diagnosed glioblastoma who receive carmustine-releasing wafers (Gliadel) along with temozolomide (TMZ) is still unknown. METHODS: MGMT promoter methylation status and protein expression were analyzed in formalin-fixed, paraffin-embedded tumor specimens obtained from 111 French patients with newly diagnosed glioblastoma. Patients received the Gliadel wafers followed by radiotherapy plus concomitant and adjuvant TMZ chemotherapy while they were enrolled in a French multicenter prospective study. RESULTS: For the whole cohort, the median overall survival (OS) was 17.5 months, and the progression-free survival was 10.3 months. Patients with tumors that harbored MGMT methylation had a significantly longer OS compared with patients who had wild-type MGMT (21.7 months vs 15.1 months; P = .025). Similarly, patients who had low MGMT protein expression (≤15%) had a significantly improved OS compared with patients who had high MGMT expression (27.0 months vs 15.1 months; P = .021). The extent of resection was the strongest clinical predictor of outcome. In multivariate Cox models that were adjusted for sex, performance status, and extent of surgery, both MGMT methylation and protein expression were identified as independent prognosticators, and the finding was validated internally using a bootstrap resampling technique. Discrepancies were identified between protein expression and MGMT methylation status, thus suggesting that the 2 assays probably assess different biologic features. CONCLUSIONS: MGMT promoter methylation status and low MGMT expression both were identified as positive prognosticators in patients with newly diagnosed glioblastoma who underwent surgical resection and received Gliadel wafer implants followed by adjuvant radiotherapy and concomitant oral TMZ chemotherapy (the Stupp protocol).


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/therapy , DNA Methylation , Glioblastoma/genetics , Glioblastoma/therapy , O(6)-Methylguanine-DNA Methyltransferase/genetics , Promoter Regions, Genetic , Adult , Aged , Antineoplastic Agents, Alkylating/therapeutic use , Biomarkers, Tumor , Carmustine/therapeutic use , Chemoradiotherapy , Chemotherapy, Adjuvant , Combined Modality Therapy , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Disease-Free Survival , Female , Humans , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Prognosis , Temozolomide
9.
Med Hypotheses ; 77(5): 857-60, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21864987

ABSTRACT

Why does O(6)-methylguanine-DNA methyltransferase (MGMT), an indispensable DNA repair enzyme, have a mechanism which seems to run counter to its importance? This enzyme is key to the removal of detrimental alkyl adducts from guanine bases. Although the mechanism is well known, an unusual feature surrounds its mode of action, which is its so-called suicidal endpoint. In addition, induction of MGMT is highly variable and its kinetics is atypical. These features raise some questions on the seemingly paradoxical mechanism. In this manuscript we point out that, although there is ample literature regarding the "how" of the MGMT enzyme, we found a lack of information on "why" this specific mechanism is in place. We then ask whether we know all there is to know about MGMT, or if perhaps there is a further as yet unknown function for MGMT, or if the suicidal mechanism may play some kind of protective role in the cell.


Subject(s)
Cell Death , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Enzyme Induction , Humans , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/genetics
10.
Melanoma Res ; 21(3): 206-16, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21460749

ABSTRACT

The efficacy of temozolomide in melanoma treatment is low (response rate <20%) and may depend on the activity of O-methylguanine DNA methyltransferase (MGMT) and mismatch repair. We identified melanoma cell lines with different sensitivities to single versus prolonged clinical dosing regimens of temozolomide treatment and assessed a variety of potential resistance mechanisms using this model. We measured mRNA expression and promoter methylation of MGMT and essential mismatch repair genes (MLH1, MSH2). Cell cycle distribution, apoptosis/necrosis induction, O-methylguanine-adduct formation, and ABCB1 gene expression were assessed. We found that three cell lines, MelA, MelB, and MelC, were more sensitive to a single dose regimen than to a prolonged regimen, which would be expected to exhibit higher cytotoxicity. KAII and LIBR cell sensitivity was higher with regard to the prolonged treatment regimen, as expected. Only MelC expressed MGMT. Gene expression correlated well with promoter methylation. Temozolomide exposure did not alter mRNA expression. Different sensitivities to temozolomide were caused neither by delayed apoptosis induction due to early cell cycle arrest nor by O-methylguanine-adduct formation or efflux transporter expression. MelC was the most resistant cell line with rapid elimination of O-methylguanine adducts. This was in good agreement with its MGMT expression. The sensitive cell lines KAII and LIBR accumulated O-methylguanine adducts after a second treatment cycle with temozolomide in contrast with the other three cell lines. We conclude that MGMT expression and DNA adduct accumulation are relevant factors in temozolomide chemosensitivity. Considering individualized temozolomide treatment regimens either by quantification of DNA adducts or by chemosensitivity testing seems worthwhile clinically.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Damage , Dacarbazine/analogs & derivatives , Melanoma/drug therapy , Melanoma/genetics , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , DNA Methylation , DNA Repair , Dacarbazine/pharmacology , Drug Resistance, Neoplasm , Gene Expression/drug effects , Humans , Melanoma/metabolism , Melanoma/pathology , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Promoter Regions, Genetic , Temozolomide
11.
Br J Cancer ; 103(4): 498-504, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20628383

ABSTRACT

BACKGROUND: O(6)-methylguanine-DNA methyltransferase (MGMT) expression in glioblastoma correlates with temozolomide resistance. Dose-intense temozolomide schedules deplete MGMT activity in peripheral blood mononuclear cells; however, no published data exist evaluating the effect of temozolomide schedules on intracranial tumour MGMT activity. METHODS: Human glioblastoma cells (GBM43) with an unmethylated MGMT promoter were implanted intracranially in immunodeficient rodents. Three weeks later, animals received temozolomide 200 mg m(-2) for 5 days (schedule A, standard dose) or 100 mg m(-2) for 21 days (schedule B, dose intense). RESULTS: Tumour MGMT activity was depleted by day 6 in both treatment groups compared with baseline. O(6)-methylguanine-DNA methyltransferase activity returned to baseline by day 22 in the schedule A group, but remained suppressed in the schedule B group. By day 29, MGMT activity had returned to baseline in both groups. Mean tumour volume was significantly decreased compared with untreated controls with either schedule (P<0.01), although neither schedule was superior (P=0.60). Median survival was 64, 42, and 28 days for schedule A, schedule B, and no drug, respectively (P<0.001 A or B vs control, P=NS A vs B). CONCLUSIONS: Dose-intense temozolomide prolongs tumour MGMT activity depletion compared with standard dosing, however, survival was not improved in this model.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/metabolism , Dacarbazine/analogs & derivatives , Glioblastoma/metabolism , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Animals , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/mortality , Cell Line, Tumor , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Dose-Response Relationship, Drug , Glioblastoma/mortality , Humans , Rats , Survival Analysis , Temozolomide , Tumor Burden/drug effects
12.
J Vis Exp ; (39)2010 May 17.
Article in English | MEDLINE | ID: mdl-20485262

ABSTRACT

SNAP-tag and CLIP-tag protein labeling systems enable the specific, covalent attachment of molecules, including fluorescent dyes, to a protein of interest in live cells. These systems offer a broad selection of fluorescent substrates optimized for a range of imaging instrumentation. Once cloned and expressed, the tagged protein can be used with a variety of substrates for numerous downstream applications without having to clone again. There are two steps to using this system: cloning and expression of the protein of interest as a SNAP-tag fusion, and labeling of the fusion with the SNAP-tag substrate of choice. The SNAP-tag is a small protein based on human O(6)-alkylguanine-DNA-alkyltransferase (hAGT), a DNA repair protein. SNAP-tag labels are dyes conjugated to guanine or chloropyrimidine leaving groups via a benzyl linker. In the labeling reaction, the substituted benzyl group of the substrate is covalently attached to the SNAP-tag. CLIP-tag is a modified version of SNAP-tag, engineered to react with benzylcytosine rather than benzylguanine derivatives. When used in conjunction with SNAP-tag, CLIP-tag enables the orthogonal and complementary labeling of two proteins simultaneously in the same cells.


Subject(s)
Fluorescent Dyes/chemistry , Image Enhancement/methods , Recombinant Fusion Proteins/chemistry , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Microscopy, Fluorescence/methods , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/chemistry , O(6)-Methylguanine-DNA Methyltransferase/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transfection
13.
J Pharmacol Exp Ther ; 333(3): 782-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20308330

ABSTRACT

O(6)-methylguanine-DNA methyltransferase (MGMT) plays a crucial role in the defense against the alkylating agent-induced cytotoxic lesion O(6)-alkylguanine in DNA. Although a significant circadian variation in MGMT activity has been found in the liver of mice, the exact mechanism of the variation remains poorly understood. In this study, we present evidence that glucocorticoids were required for the 24-h oscillation of MGMT expression in mouse liver. The exposure of mouse hepatic cells (Hepa1-6) to dexamethasone (DEX) significantly increased the mRNA levels of MGMT in a dose-dependent manner. The DEX-induced increase in MGMT expression was reversed by concomitant treatment with RU486 [11beta-[p-(dimethylamino) phenyl]-17beta-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one], a glucocorticoid receptor antagonist. The mRNA levels of MGMT and its enzymatic activity in the liver of mice showed significant 24-h oscillations, which were not observed in adrenalectomized mice. A single administration of DEX to adrenalectomized mice significantly increased the mRNA levels of MGMT in the liver. These findings suggest that the 24-h oscillation in the hepatic expression of MGMT is caused by the endogenous rhythm of glucocorticoid secretion. Dacarbazine (DTIC), a potent O(6)-guanine-alkylating agent, causes serious hepatotoxicity accompanied by hepatocellular necrosis and hepatic vein thrombosis. DTIC-induced hepatotoxicity in mice was attenuated by administering the drug at the time of day when MGMT expression was abundant. The present findings suggest that glucocorticoid-regulated oscillation in the hepatic MGMT expression is the underlying cause of dosing time-dependent changes in DTIC-induced hepatotoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Dacarbazine/toxicity , Glucocorticoids/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/genetics , Adrenalectomy , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/pathology , Circadian Rhythm/drug effects , Corticosterone/blood , Dose-Response Relationship, Drug , Hormone Antagonists/pharmacology , Male , Mice , Mice, Inbred ICR , Mifepristone/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
14.
Cancer Prev Res (Phila) ; 3(1): 92-100, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20051376

ABSTRACT

Folate deficiency is implicated in human colon cancer. The effects of feeding rats a folate-deficient diet for 24 weeks on DNA damage (8-oxo-7,8-dihydroguanine), DNA repair [O(6)-methylguanine-DNA methyltransferase (MGMT) and 8-oxoguanine-DNA glycosylase (OGG-1) activity], and epigenetic parameters (genome-wide cytosine methylation and indices of cellular methylation status) were investigated. Relative to control diet, the folate-deficient diet resulted in significantly reduced levels of serum ( approximately 80%; P < 0.0001), whole blood ( approximately 40%; P < 0.0001), and tissue folate (between 25% and 60% depending on the tissue sampled; P < 0.05); increased plasma total homocysteine ( approximately 35%; P < 0.05); and decreased S-adenosylmethionine to S-adenosylhomocysteine concentrations ( approximately 11%; P < 0.05). There was no significant change in the levels of 5-methyldeoxycytidine in liver or colon DNA, nor in the activity of liver DNA cytosine methyltransferase. However, there were significant increases in 8-oxo-7,8-dihydroguanine (P < 0.001) in lymphocyte DNA and in levels of the DNA repair proteins OGG-1 ( approximately 27%; P < 0.03) and MGMT ( approximately 25%; P < 0.003) in the liver, but not in the colon. This may reflect the ability of the liver, but not the colon, to upregulate DNA repair enzymes in response to either elevated DNA damage or an imbalance in the nucleotide precursor pool. These results show that folate deficiency can significantly modulate DNA damage and DNA repair, providing mechanisms by which it plays a role in the etiology of human cancer. We speculate that the inability of colon tissue to respond to folate deficiency occurs in humans and may increase the potential for malignant transformation.


Subject(s)
Colon/metabolism , DNA Glycosylases/biosynthesis , DNA Methylation/physiology , Folic Acid Deficiency/metabolism , Liver/metabolism , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Animals , DNA Damage/physiology , DNA Repair/physiology , Male , Rats
15.
Anticancer Res ; 29(10): 3741-50, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19846904

ABSTRACT

O(6)-Methylguanine-DNA methyltransferase (MGMT), a ubiquitous DNA repair protein, reverses mutagenic and cytotoxic effects of O(6)-alkylguanine in DNA induced by chemotherapeutic N-alkyl N-nitrosourea and procarbazine type drugs by dealkylating the adduct. MGMT expression is down-regulated by wild-type p53 (WTp53) in human tumor cells. Here we report that p53 sequesters the Sp1 transcription factor to prevent its binding to the cognate cis elements in the MGMT promoter and thus inhibits MGMT expression. Sp1 overexpression abrogated the inhibitory effect of p53 on the MGMT promoter activity in a dose-dependent manner. Stable interaction of Sp1 with WTp53 was observed in HCT116 cells. Moreover, WTp53 overexpression reduced the binding of the nuclear extract to the Sp1 consensus sequence, even though recombinant p53 alone did not bind to the same sequence. Taken together, these results suggest that sequestration of Sp1 could be one of the mechanisms by which p53 negatively regulates MGMT expression, thus enhancing sensitivity of tumor cells to O(6)-alkylguanine generating drugs.


Subject(s)
DNA Repair , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Sp1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Consensus Sequence , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Down-Regulation , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , HCT116 Cells , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/genetics , Promoter Regions, Genetic , Protein Binding , Sp1 Transcription Factor/genetics , Temozolomide , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
16.
Eur J Endocrinol ; 161(4): 553-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19589911

ABSTRACT

CONTEXT: Crooke's cell adenoma (CCA), characterized by massive Crooke's hyaline change in corticotroph adenoma, causes a rare subtype of Cushing's disease. In contrast to ordinary corticotroph adenomas, CCAs are generally aggressive and present as invasive macroadenomas, which are refractory to both surgery and radiotherapy and have a high-recurrence rate. Moreover, some patients with CCA present with distant or craniospinal metastases. Currently, there are no effective standard therapies for CCA. OBJECTIVE: We report a patient with Crooke's cell carcinoma who presented with local invasion and liver metastases, which was refractory to conventional therapeutic modalities including transsphenoidal surgery, radiosurgery, medications, and hepatic transcatheter arterial embolization. After all these treatments failed, the patient had monthly temozolomide administrations, resulting in gradual clinical improvement and biochemical data that were consistent with tumor shrinkage. In glioblastoma, low O(6)-methylguanine DNA methyltransferase (MGMT) expression is associated with epigenetic gene silencing and predicts a better response to temozolomide. METHODS: We thus investigated MGMT expression, immunohistochemically, in seven CCAs (five invasive macroadenomas and two invasive microadenomas) and 17 ordinary-type adenomas (OTAs; three noninvasive macroadenomas, 12 noninvasive microadenomas, and two invasive microadenomas) from patients with Cushing's disease. RESULTS: In seven CCAs, all five invasive macroadenomas exhibited low MGMT expression, defined as <5% nuclear MGMT staining. In 17 OTAs, only one adenoma showed low MGMT expression. CONCLUSION: In Cushing's disease, invasive macroadenomas including CCA usually have low-MGMT expression. Temozolomide thus may be a new therapeutic option for invasive macroadenomas such as CCA particularly when conventional treatments are ineffective.


Subject(s)
O(6)-Methylguanine-DNA Methyltransferase/deficiency , O(6)-Methylguanine-DNA Methyltransferase/genetics , Pituitary ACTH Hypersecretion/enzymology , Pituitary Neoplasms/enzymology , Adolescent , Adrenocorticotropic Hormone/blood , Adult , Aged , Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Female , Humans , Hydrocortisone/blood , Immunohistochemistry , Liver Neoplasms/secondary , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Pituitary ACTH Hypersecretion/pathology , Pituitary ACTH Hypersecretion/surgery , Pituitary Neoplasms/pathology , Pituitary Neoplasms/surgery , Temozolomide , Tomography, X-Ray Computed , Young Adult
17.
Gynecol Oncol ; 112(1): 224-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18973931

ABSTRACT

OBJECTIVE: O(6)-methylguanine-DNA methyltransferase (MGMT) acts to repair DNA damaged by alkylation of guanine residues. MGMT promoter methylation and gene silencing is seen in a variety of cancers and pre-cancerous changes [Ogino S, Meyerhardt JA, Kawasaki T, et al. CpG island methylation, response to combination chemotherapy, and patient survival in advanced microsatellite stable colorectal carcinoma. Virchows Arch 2007;450:529-37; Rodriguez MJ, Acha A, Ruesga MT, Rodriguez C, Rivera JM, Aguirre JM. Loss of expression of DNA repair enzyme MGMT in oral leukoplakia and early oral squamous cell carcinoma. A prognostic tool? Cancer Lett 2007;245:263-8; Ishii T, Murakami J, Notohara K, et al. Oesophageal squamous cell carcinoma may develop within a background of accumulating DNA methylation in normal and dysplastic mucosa. Gut 2007;56:13-9]. The loss of MGMT activity and promoter methylation is associated with increased sensitivity to alkylating agents and is a favorable prognostic indicator in gliomas [Weaver KD, Grossman SA, Herman JG. Methylated tumor-specific DNA as a plasma biomarker in patients with glioma. Cancer Invest 2006;24:35-40; Esteller M, Garcia-Foncillas J, Andion E, et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med 2000;343:1350-4; Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 2005;352:997-1003]. We sought to determine if MGMT promoter methylation plays a role in endometrial cancer. METHODS: One hundred and twenty primary endometrial cancers were analyzed for MGMT promoter methylation by combined bisulfite restriction analysis (COBRA). The cohort included 77 endometrioid endometrial cancers, 43 endometrial tumors of adverse histologic type, and 6 endometrial cancer cell lines. Twenty-one endometrioid and mixed endometrioid ovarian cancers were also analyzed. A subset of the primary tumors was analyzed for MGMT expression by immunohistochemistry. RESULTS: No MGMT promoter methylation was seen in the 120 endometrial cancers evaluated or the 6 endometrial cancer cell lines. One of the 21 endometrioid ovarian cancers showed methylation. Immunohistochemistry revealed moderate to high level expression of MGMT in the primary endometrial tumors. CONCLUSION: MGMT promoter methylation is an infrequent event in endometrial cancer. MGMT expression and the ability to repair damaged alkylguanine residues could in part explain the limited response of endometrial tumors to alkylating chemotherapy.


Subject(s)
DNA Methylation , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , DNA, Neoplasm/genetics , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , Neoplasm Staging , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Promoter Regions, Genetic
18.
Neuropathology ; 29(4): 379-88, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19019175

ABSTRACT

We studied the expression of O(6)-methylguanine-DNA methyltransferase (O(6)-MGMT), P-glycoprotein (Pgp), and multidrug resistance protein-1 (MRP-1) in 23 glioblastomas using RT-PCR, methylation-specific PCR, and immunohistochemistry, and analyzed their association with overall patient survival. Univariate analysis of collected data demonstrated that the expressions of O(6)-MGMT and MRP-1 detected by immunohistochemistry, in addition to the consistent factors, including preoperative Karnofsky performance scale (KPS), radical surgery, and tumor location and extension, were significant prognostic factors for the overall survival (OS) of patients with glioblastoma, who received nimustine (ACNU)-based chemotherapy in association with surgery and radiotherapy. Among them, following multivariate analysis, preoperative KPS, radical surgery, tumor location, and the expression of O(6)-MGMT remained as significant prognostic factors. These findings suggest that immunohistochemical analysis of O(6)-MGMT in patients with glioblastoma can be a useful method to predict the effects of chemotherapy and identify alternative chemotherapeutic regimens for O(6)-MGMT-positive patients.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Biomarkers, Tumor/biosynthesis , Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Female , Glioblastoma/diagnosis , Glioblastoma/enzymology , Glioblastoma/genetics , Humans , Immunohistochemistry , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/genetics , Predictive Value of Tests , Prognosis , Young Adult
19.
Ukr Biokhim Zh (1999) ; 80(6): 60-5, 2008.
Article in Russian | MEDLINE | ID: mdl-19351058

ABSTRACT

The influence of plant lectins on primary DNA damage repair and on expression of repair enzyme alkylguanine-DNA alkyltransferase (MGMT) in mammalian cells in vitro has been investigated. Those lectins have been shown to modify DNA damage repair process, and to induce the MGMT expression increasing its level in the used model system.


Subject(s)
DNA Damage , DNA Repair , Mesenchymal Stem Cells/drug effects , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Plant Lectins/pharmacology , Animals , Blotting, Western , Cell Culture Techniques , Cell Line, Tumor , Cricetinae , Cricetulus , Enzyme Induction , Humans , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/metabolism
20.
Zhonghua Fu Chan Ke Za Zhi ; 42(9): 600-4, 2007 Sep.
Article in Chinese | MEDLINE | ID: mdl-17983514

ABSTRACT

OBJECTIVE: To evaluate the relationship between combined multigene detection and response to chemotherapy and prognosis in epithelial ovarian carcinomas (EOCS). METHODS: A total of 80 ovarian tissue samples taken from the surgical specimens of patients with EOCS of our hospital in the last two decades who had received chemotherapy after surgery were paraffin-embedded. The samples were divided into 2 groups, good prognosis group (patients who survived more than 2 years, n = 46) and poor prognosis group (patients who survived less than 2 years, n = 34). The expression levels of ToPo-II, Ki-67, MGMT, PCNA, p27, p53, p16, P-gp, LRP, GST-pi, bcl-2, C-myc, Fas, bax, MSH2, MRP and BCRP were investigated by the combination of tissue arrays and immunohistochemical streptavidin-biotin peroxidase (SP) method in all samples. Data were analysed with SPSS 12.0 for windows. RESULTS: There were statistically significant differences in the positive expression levels of P-gp, BCRP, MGMT, MSH2, p27 and p16 (62%, 50% and 50% in poor prognosis group vs 33%, 28% and 28% respectively, P < 0.05) in the good prognosis group, suggesting that the positive expression levels of P-gp, BCRP, MGMT, MSH2, p27 and p16 were related to the response to chemotherapy and prognosis in EOCS. And the positive expression of P-gp, BCRP and MSH2 (43%, 54% and 43%) indicated poor prognosis, while the positive expression of MGMT, p27 and p16 (18%, 29% and 24%, P < 0.05) indicated good prognosis. Cox multigene expression analysis confirmed that the positive expression levels of MRP, C-myc, LRP, p16, p27, MGMT, ToPo-II, P-gp and GST-pi were related to the response to chemotherapy and prognosis in EOCS. And the positive expression of MRP, C-myc, LRP, ToPo-II, P-gp and GST-pi indicated poor prognosis, while the positive expression of MGMT, p27 and p16 indicated good prognosis. Combined multigene detections were conducted among P-gp, BCRP, MGMT, MSH2, p27 and p16, and there were statistically significant differences in the positive coexpression of P-gp plus MGMT in the two groups (P < 0.05); indicating that the combined multigene expression were related to the response to chemotherapy and prognosis in EOCS. The predictive value to response to chemotherapy and prognosis of the positive coexpression of P-gp plus MGMT was 70%. CONCLUSIONS: By univariate and multivariate analyses, the positive expression of P-gp, MGMT, p27 and p16 are related to the response to chemotherapy and prognosis in EOCS. The combined multigene expression of P-gp plus MGMT are related to the response to chemotherapy and prognosis and could predict prognosis more effectively.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Cyclin-Dependent Kinase Inhibitor Proteins/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Adult , Aged , Epithelial Cells/pathology , Female , Glutathione S-Transferase pi/biosynthesis , Humans , Immunohistochemistry , Logistic Models , Middle Aged , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Prognosis , Survival Analysis , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...