Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
BMC Cancer ; 21(1): 493, 2021 May 03.
Article in English | MEDLINE | ID: mdl-33941107

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the deadliest and the most common primary brain tumor in adults. The invasiveness and proliferation of GBM cells can be decreased through the inhibition of Wnt/ß-catenin pathway. In this regard, celecoxib is a promising agent, but other COXIBs and 2,5-dimethylcelecoxib (2,5-DMC) await elucidation. Thus, the aim of this study was to analyze the impact of celecoxib, 2,5-DMC, etori-, rofe-, and valdecoxib on GBM cell viability and the activity of Wnt/ß-catenin pathway. In addition, the combination of the compounds with temozolomide (TMZ) was also evaluated. Cell cycle distribution and apoptosis, MGMT methylation level, COX-2 and PGE2 EP4 protein levels were also determined in order to better understand the molecular mechanisms exerted by these compounds and to find out which of them can serve best in GBM therapy. METHODS: Celecoxib, 2,5-DMC, etori-, rofe- and valdecoxib were evaluated using three commercially available and two patient-derived GBM cell lines. Cell viability was analyzed using MTT assay, whereas alterations in MGMT methylation level were determined using MS-HRM method. The impact of COXIBs, in the presence and absence of TMZ, on Wnt pathway was measured on the basis of the expression of ß-catenin target genes. Cell cycle distribution and apoptosis analysis were performed using flow cytometry. COX-2 and PGE2 EP4 receptor expression were evaluated using Western blot analysis. RESULTS: Wnt/ß-catenin pathway was attenuated by COXIBs and 2,5-DMC irrespective of the COX-2 expression profile of the treated cells, their MGMT methylation status, or radio/chemoresistance. Celecoxib and 2,5-DMC were the most cytotoxic. Cell cycle distribution was altered, and apoptosis was induced after the treatment with celecoxib, 2,5-DMC, etori- and valdecoxib in T98G cell line. COXIBs and 2,5-DMC did not influence MGMT methylation status, but inhibited COX-2/PGE2/EP4 pathway. CONCLUSIONS: Not only celecoxib, but also 2,5-DMC, etori-, rofe- and valdecoxib should be further investigated as potential good anti-GBM therapeutics.


Subject(s)
Brain Neoplasms/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Glioblastoma/metabolism , Neoplasm Proteins/drug effects , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Wnt Signaling Pathway/drug effects , Aged , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Celecoxib/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cyclooxygenase 2/metabolism , DNA Modification Methylases/drug effects , DNA Modification Methylases/metabolism , DNA Repair Enzymes/drug effects , DNA Repair Enzymes/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Etoricoxib/pharmacology , Female , Glioblastoma/drug therapy , Humans , Isoxazoles/pharmacology , Lactones/pharmacology , Male , Methylation , Middle Aged , Neoplasm Proteins/metabolism , Receptors, Prostaglandin E, EP4 Subtype/drug effects , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sulfones/pharmacology , Temozolomide/pharmacology , Tumor Suppressor Proteins/drug effects , Tumor Suppressor Proteins/metabolism , beta Catenin/drug effects , beta Catenin/metabolism
2.
Clin Epigenetics ; 13(1): 35, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33588926

ABSTRACT

BACKGROUND: Epigenetic inactivation of O6-methylguanine DNA-methyltransferase (MGMT) is associated with increased sensitivity to alkylating chemotherapeutic agents in glioblastoma patients. The genetic background underlying MGMT gene methylation may explain individual differences in treatment response and provide a clue to a personalized treatment strategy. Making use of the longitudinal twin design, we aimed, for the first time, to estimate the genetic contributions to MGMT methylation in a Danish twin cohort. METHODS: DNA-methylation from whole blood (18 monozygotic (MZ) and 25 dizygotic (DZ) twin pairs) repeated 10 years apart from the Longitudinal Study of Aging Danish Twins (LSADT) were used to search for genetic and environmental contributions to DNA-methylation at 170 CpG sites of across the MGMT gene. Both univariate and bivariate twin models were applied. The intraclass correlations, performed on cross-sectional data (246 MZ twin pairs) from an independent study population, the Middle-Aged Danish Twins (MADT), were used to assess the genetic influence at each CpG site of MGMT for replication. RESULTS: Univariate twin model revealed twelve CpG sites showing significantly high heritability at intake (wave 1, h2 > 0.43), and seven CpG sites with significant heritability estimates at end of follow-up (wave 2, h2 > 0.5). There were six significant CpG sites, located at the gene body region, that overlapped among the two waves (h2 > 0.5), of which five remained significant in the bivariate twin model, which was applied to both waves. Within MZ pair correlation in these six CpGs from MADT demarks top level of genetic influence. There were 11 CpGs constantly have substantial common environmental component over the 10 years. CONCLUSIONS: We have identified 6 CpG sites linked to the MGMT gene with strong and persistent genetic control based on their DNA methylation levels. The genetic basis of MGMT gene methylation could help to explain individual differences in glioblastoma treatment response and most importantly, provide references for mapping the methylation Quantitative Trait Loci (meQTL) underlying the genetic regulation.


Subject(s)
DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , DNA/metabolism , Glioblastoma/genetics , Guanine/analogs & derivatives , Tumor Suppressor Proteins/genetics , Aged , Aging/genetics , Antineoplastic Agents, Alkylating/therapeutic use , Cohort Studies , CpG Islands/genetics , Cross-Sectional Studies , DNA Methylation , DNA Modification Methylases/drug effects , DNA Repair Enzymes/drug effects , Denmark/epidemiology , Environment , Epigenomics , Female , Gene Silencing/drug effects , Glioblastoma/drug therapy , Guanine/metabolism , Humans , Longitudinal Studies , Male , Methyltransferases , Middle Aged , Tumor Suppressor Proteins/drug effects , Twins, Dizygotic/genetics , Twins, Monozygotic/genetics
3.
CNS Neurosci Ther ; 27(5): 552-563, 2021 05.
Article in English | MEDLINE | ID: mdl-33460245

ABSTRACT

BACKGROUND: Temozolomide (TMZ) is a first-line chemotherapy drug for the treatment of malignant glioma and resistance to it poses a major challenge. Receptor-interacting protein 2 (RIP2) is associated with the malignant character of cancer cells. However, it remains unclear whether RIP2 is involved in TMZ resistance in glioma. METHODS: RIP2 expression was inhibited in TMZ-resistant glioma cells and normal glioma cells by using small interfering RNA (siRNA) against RIP2. Plasmid transfection method was used to overexpress RIP2. Cell counting kit-8 assays were performed to evaluate cell viability. Western blotting or immunofluorescence was performed to determine RIP2, NF-κB, and MGMT expression in cells. Flow cytometry was used to investigate cell apoptosis. TMZ-resistant glioma xenograft models were established to evaluate the role of the RIP2/NF-κB/MGMT signaling pathway in drug resistance. RESULTS: We observed that RIP2 expression was upregulated in TMZ-resistant glioma cells, whereas silencing of RIP2 expression enhanced cellular sensitivity to TMZ. Similarly, upon the induction of RIP2 overexpression, glioma cells developed resistance to TMZ. The molecular mechanism underlying the process indicated that RIP2 can activate the NF-κB signaling pathway and upregulate the expression of O6-methylguanine-DNA methyltransferase (MGMT), following which the glioma cells develop drug resistance. In the TMZ-resistant glioma xenograft model, treatment with JSH-23 (an NF-κB inhibitor) and lomeguatrib (an MGMT inhibitor) could enhance the sensitivity of the transplanted tumor to TMZ. CONCLUSION: We report that the RIP2/NF-κB/MGMT signaling pathway is involved in the regulation of TMZ resistance. Interference with NF-κB or MGMT activity could constitute a novel strategy for the treatment of RIP2-positive TMZ-resistant glioma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , DNA Modification Methylases/drug effects , DNA Modification Methylases/genetics , DNA Repair Enzymes/drug effects , DNA Repair Enzymes/genetics , Drug Resistance, Neoplasm/genetics , Glioma/drug therapy , NF-kappa B/drug effects , NF-kappa B/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/drug effects , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Signal Transduction/drug effects , Temozolomide/pharmacology , Tumor Suppressor Proteins/drug effects , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/drug effects , Brain Neoplasms/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Humans , Male , Mice , Mice, Inbred BALB C , NF-kappa B/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
4.
Nutrients ; 12(3)2020 Mar 13.
Article in English | MEDLINE | ID: mdl-32183060

ABSTRACT

Breast cancer is a common cancer that occurs due to different epigenetic alterations and genetic mutations. Various epidemiological studies have demonstrated an inverse correlation between breast cancer incidence and flavonoid intake. The anti-cancer action of flavonoids, a class of polyphenolic compounds that are present in plants, as secondary metabolites has been a major topic of research for many years. Our review analysis demonstrates that flavonoids exhibit anti-cancer activity against breast cancer occurring in different ethnic populations. Breast cancer subtype and menopausal status are the key factors in inducing the flavonoid's anti-cancer action in breast cancer. The dose is another key factor, with research showing that approximately 10 mg/day of isoflavones is required to inhibit breast cancer occurrence. In addition, flavonoids also influence the epigenetic machinery in breast cancer, with research demonstrating that epigallocatechin, genistein, and resveratrol all inhibited DNA methyltransferase and altered chromatin modification in breast cancer. These flavonoids can induce the expression of different tumor suppressor genes that may contribute to decreasing breast cancer progression and metastasis. Additional studies are required to confirm the contribution of epigenetic modifications by flavonoids to breast cancer prevention.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Epigenesis, Genetic/drug effects , Flavonoids/pharmacology , Polyphenols/pharmacology , Breast Neoplasms/prevention & control , Breast Neoplasms/therapy , Catechin/analogs & derivatives , Catechin/pharmacology , Chromatin/drug effects , DNA Modification Methylases/drug effects , Female , Genes, Tumor Suppressor/drug effects , Genistein/pharmacology , Humans , Resveratrol/pharmacology
5.
Br J Haematol ; 186(1): 91-100, 2019 07.
Article in English | MEDLINE | ID: mdl-30891745

ABSTRACT

Sickle cell disease (SCD) affects over 2 million people worldwide with high morbidity and mortality in underdeveloped countries. Therapeutic interventions aimed at reactivating fetal haemoglobin (HbF) is an effective approach for improving survival and ameliorating the clinical severity of SCD. A class of agents that inhibit DNA methyltransferase (DNMT) activity show promise as HbF inducers because off-target effects are not observed at low concentrations. However, these compounds are rapidly degraded by cytidine deaminase when taken by oral administration, creating a critical barrier to clinical development for SCD. We previously demonstrated that microRNA29B (MIR29B) inhibits de novo DNMT synthesis, therefore, the goal of our study was to determine if MIR29 mediates HbF induction. Overexpression of MIR29B in human KU812 cells and primary erythroid progenitors significantly increased the percentage of HbF positive cells, while decreasing the expression of DNMT3A and the HBG repressor MYB. Furthermore, HBG promoter methylation levels decreased significantly following MIR29B overexpression in human erythroid progenitors. We subsequently, observed higher MIR29B expression in SCD patients with higher HbF levels compared to those with low HbF. Our findings provide evidence for the ability of MIR29B to induce HbF and supports further investigation to expand treatment options for SCD.


Subject(s)
Anemia, Sickle Cell/genetics , Epigenesis, Genetic/drug effects , Fetal Hemoglobin/genetics , MicroRNAs/physiology , Transcriptional Activation/drug effects , gamma-Globins/genetics , Cell Line , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/drug effects , DNA Methyltransferase 3A , DNA Modification Methylases/biosynthesis , DNA Modification Methylases/drug effects , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/metabolism , Humans , MicroRNAs/antagonists & inhibitors
6.
Curr Top Med Chem ; 18(28): 2380-2394, 2018.
Article in English | MEDLINE | ID: mdl-30430946

ABSTRACT

Epigenetics process is the heritable change in gene function that does not involve changes in the DNA sequence. Until now, several types of epigenetic mechanisms have been characterized, including DNA methylation, histone modification (acetylation, methylation, etc.), nucleosome remodeling, and noncoding RNAs. With the biological investigations of these modifiers, some of them are identified as promoters in the process of various diseases, such as cancer, cardiovascular disease and virus infection. Epigenetic changes may serve as potential "first hits" for tumorigenesis. Hence, targeting epigenetic modifiers is being considered as a promising way for disease treatment. To date, six agents in two epigenetic target classes (DNMT and HDAC) have been approved by the US Food and Drug Administration (FDA). Most of these drugs are applied in leukemia, lymphoma therapy, or are combined with other drugs for the treatment of solid tumor. Due to the rapid development of epigenetics and epigenetics targeted drugs, it is becoming an emerging area in targeted drug design.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Design , Epigenesis, Genetic/drug effects , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Carcinogenesis , DNA Modification Methylases/drug effects , Histone Deacetylases/drug effects , Humans , Neoplasms/enzymology , Neoplasms/genetics
7.
World Neurosurg ; 116: e147-e161, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29709748

ABSTRACT

BACKGROUND: There is limited information on O6-methylguanine DNA methyltransferase (MGMT) status, extent of surgical resection, and its impact on overall outcomes in patients with glioblastoma (GBM). METHODS: After institutional review board approval, 233 newly diagnosed patients with GBM with known MGMT status (2009-2015) were included in our analysis. Clinical, imaging, and follow-up data were collected from the database. Overall survival (OS) and progression-free survival (PFS) were the primary and secondary end points, respectively. RESULTS: Of patients, 51.9% were younger than 65 years and 44.2% were noted to have promoter methylation of MGMT. Median residual tumor volume was 1.1 cm3 and extent of complete resection of enhancing tumor on imaging was 96%. Estimated median OS and PFS were 10.9 months and 5.4 months, respectively. MGMT status was an independent predictor of PFS (hazard ratio [HR], 0.52; P = 0.005) but only marginally associated with OS (P = 0.059). In MGMT methylated patients, extent of resection (≥86%) and good performance status (Karnofsky Performance Status ≥70) were independently associated with PFS and OS, respectively (PFS: HR, 0.21; P = 0.015; OS: HR, 0.05; P = 0.002). In MGMT promoter unmethylated patients, extent of resection (≥86%) was independently associated with OS (P = 0.039). Concurrent chemoradiotherapy was associated with OS/PFS irrespective of age and MGMT status. CONCLUSIONS: Greater extent of resection of enhancing tumor was associated with improved PFS in MGMT promoter methylated patients, OS regardless of MGMT status. Elderly patients with methylated MGMT promoter were found to have improved PFS whereas younger patients had improved OS with MGMT promoter methylated status.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Glioblastoma/pathology , Guanine/analogs & derivatives , Methyltransferases/metabolism , Neoplasm, Residual/pathology , Adult , Aged , Biomarkers, Tumor/genetics , Brain Neoplasms/surgery , DNA Modification Methylases/drug effects , DNA Modification Methylases/metabolism , DNA Repair Enzymes/drug effects , DNA Repair Enzymes/metabolism , Disease-Free Survival , Female , Glioblastoma/genetics , Glioblastoma/surgery , Guanine/pharmacology , Humans , Male , Middle Aged
8.
Carcinogenesis ; 39(4): 601-613, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29452350

ABSTRACT

Targeting tumor-initiating, drug-resistant populations of cancer stem cells (CSC) with phytochemicals is a novel paradigm for cancer prevention and treatment. We herein employed a phenotypic drug discovery approach coupled to mechanism-of-action profiling and target deconvolution to identify phenolic components of extra virgin olive oil (EVOO) capable of suppressing the functional traits of CSC in breast cancer (BC). In vitro screening revealed that the secoiridoid decarboxymethyl oleuropein aglycone (DOA) could selectively target subpopulations of epithelial-like, aldehyde dehydrogenase (ALDH)-positive and mesenchymal-like, CD44+CD24-/low CSC. DOA could potently block the formation of multicellular tumorspheres generated from single-founder stem-like cells in a panel of genetically diverse BC models. Pretreatment of BC populations with noncytotoxic doses of DOA dramatically reduced subsequent tumor-forming capacity in vivo. Mice orthotopically injected with CSC-enriched BC-cell populations pretreated with DOA remained tumor-free for several months. Phenotype microarray-based screening pointed to a synergistic interaction of DOA with the mTOR inhibitor rapamycin and the DNA methyltransferase (DNMT) inhibitor 5-azacytidine. In silico computational studies indicated that DOA binds and inhibits the ATP-binding kinase domain site of mTOR and the S-adenosyl-l-methionine (SAM) cofactor-binding pocket of DNMTs. FRET-based Z-LYTE™ and AlphaScreen-based in vitro assays confirmed the ability of DOA to function as an ATP-competitive mTOR inhibitor and to block the SAM-dependent methylation activity of DNMTs. Our systematic in vitro, in vivo and in silico approaches establish the phenol-conjugated oleoside DOA as a dual mTOR/DNMT inhibitor naturally occurring in EVOO that functionally suppresses CSC-like states responsible for maintaining tumor-initiating cell properties within BC populations.


Subject(s)
Acetates/pharmacology , Breast Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Olive Oil/chemistry , Plant Extracts/pharmacology , Pyrans/pharmacology , Animals , Cyclopentane Monoterpenes , DNA Modification Methylases/drug effects , Female , Humans , Mice , TOR Serine-Threonine Kinases/drug effects , Xenograft Model Antitumor Assays
9.
Neuro Oncol ; 20(2): 236-248, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29016925

ABSTRACT

Background: Glioblastoma (GBM) is almost invariably fatal due to failure of standard therapy. The relapse of GBM following surgery, radiation, and systemic temozolomide (TMZ) is attributed to the ability of glioma stem cells (GSCs) to survive, evolve, and repopulate the tumor mass, events on which therapy exerts a poorly understood influence. Methods: Here we explore the molecular and cellular evolution of TMZ resistance as it emerges in vivo (xenograft models) in a series of human GSCs with either proneural (PN) or mesenchymal (MES) molecular characteristics. Results: We observed that the initial response of GSC-initiated intracranial xenografts to TMZ is eventually replaced by refractory growth pattern. Individual tumors derived from the same isogenic GSC line expressed divergent and complex profiles of TMZ resistance markers, with a minor representation of O6-methylguanine DNA methyltransferase (MGMT) upregulation. In several independent TMZ-resistant tumors originating from MES GSCs we observed a consistent diminution of mesenchymal features, which persisted in cell culture and correlated with increased expression of Nestin, decline in transglutaminase 2 and sensitivity to radiation. The corresponding mRNA expression profiles reflective of TMZ resistance and stem cell phenotype were recapitulated in the transcriptome of exosome-like extracellular vesicles (EVs) released by GSCs into the culture medium. Conclusions: Intrinsic changes in the tumor-initiating cell compartment may include loss of subtype characteristics and reciprocal alterations in sensitivity to chemo- and radiation therapy. These observations suggest that exploiting therapy-induced changes in the GSC phenotype and alternating cycles of therapy may be explored to improve GBM outcomes.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Drug Resistance, Neoplasm/drug effects , Extracellular Vesicles/drug effects , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Temozolomide/pharmacology , Animals , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Modification Methylases/drug effects , DNA Modification Methylases/genetics , Extracellular Vesicles/metabolism , Humans , Mice , Neoplastic Stem Cells/metabolism
11.
Toxicology ; 361-362: 62-72, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27396502

ABSTRACT

Mammalian spermatogenesis is a progressive process comprising spermatogonial proliferation, spermatocytic meiosis, and later spermiogenesis, which is considered to be under the regulation of epigenetic parameters. To gain insights into the significance of DNA methylation in early spermatogenesis, 5-azadC was used as a molecular biological tool to mimic the level of DNA methylation in vivo. Since the drug is incorporated into DNA during the S-phase, spermatogonia and spermatocytes would be affected primarily in mouse spermatogenesis. Adult male ICR mice were intraperitoneally injected with 5-azadC at a dose of 0.25mg/kg/day for 10 consecutive days, allowing us to examine its maximum effect on the kinetics of spermatogonia and spermatocytes. In this short-term protocol, 5-azadC induced significant histological abnormalities, such as a marked increase in apoptosis of spermatogonia and spermatocytes, followed by severe loss of spermatids, while after termination of 5-azadC treatment, normal histology was restored in the testis within 35days. Quantification of the methylation level of CCGG sites as well as whole DNA showed spermatogonial hypomethylation, which correlated with increased apoptosis of spermatogonia. Interestingly, the hypomethylated cells were simultaneously positive for tri-methylated histone H3 at K4. On the other hand, no changes in methylation level were found in spermatocytes, but PCNA staining clearly showed disordered accumulation of S-phase spermatocytes, which increased their apoptosis in stage XII. In addition, different immunohistochemical staining pattern was found for DNA methyltransferases (DNMTs); DNMT1was expressed in the majority of all germ cells, but DNMT3a and b were only expressed in spermatogonia. Our results indicate that 5-azadC caused DNA hypomethylation in spermatogonia, but induced prolongation of S-phase in spermatocytes, resulting in the induction of apoptosis in both cases. Thus, 5-azadC affects spermatogenesis at more than one differentiation stage with different mechanisms, probably due to the specific usage of DNMTs.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Azacitidine/analogs & derivatives , DNA Modification Methylases/metabolism , Spermatogenesis/drug effects , Animals , Azacitidine/toxicity , Cell Differentiation/drug effects , DNA Methylation/drug effects , DNA Modification Methylases/drug effects , Decitabine , Germ Cells/drug effects , Germ Cells/ultrastructure , Male , Mice , Mice, Inbred ICR , Seminiferous Tubules/drug effects , Seminiferous Tubules/ultrastructure , Spermatocytes/drug effects , Spermatocytes/metabolism , Spermatogonia/drug effects , Spermatogonia/metabolism , Testis/drug effects , Testis/metabolism
12.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 36(10): 1219-1223, 2016 10.
Article in Chinese | MEDLINE | ID: mdl-30641010

ABSTRACT

Objective To observe the effect of Wenhua Juanbi Recipe (WJR) on the expres- sions of DNA methyltransferases (DNMTs) in peripheral blood mononuclear cells (PBMCs) of collagen- inducing arthritis (CIA) , and to study its mechanism for treating CIA. Methods Totally 90 Wistar rats were randomly divided into the model group (n =80) and the normal control group (n = 10). Rats of the model group were injected with type II collagen of bovine (BC II) emulsion from the tail to establish CIA model. Successfully modeled 50 CIA rats were randomly divided into five groups, i.e., the model group, the methotrexate (MTX) group, the low dose WJR group, the middle dose WJR group, the high dose WJR group, 10 in each group. Rats in the model group were administered with normal saline by gastrogavage, once per day. Rats in low, middle, and high dose WJR groups were administered with WJR by gas- trogavage at the daily dose of 22. 9, 45. 8, 68. 7 g/kg, respectively (once per day). Rats in the MTX group were administered with MTX suspension (0.78 mg/kg) by gastrogavage, once per week for 30 successive days. The paw swelling was evaluated using volume method (draining volume). PBMCs were extrac- ted from each group after intervention. mRNA expression levels of DNMTs (DNMT1 , DNMT3a, DNMT3b) were detected by real-time quantitative PCR. Results Compared with the normal group, the paws were obviously swollen in the model group (P <0. 01). Compared with the model group, swollen paws were obviously alleviated in low, middle, and high dose WJR groups, and the MTX group (P <0.01). Compared with before treatment in the same group, swollen paws were obviously alleviated in low, middle, and high dose WJR groups, and the MTX group (P <0. 01 ). Compared with the normal group, expression levels of DNMT1, DNMT3a, and DNMT3b in PBMCs were obviously lowered in the model group (P <0.01). Compared with the model group, expression levels of DNMT1 , DNMT3a, DNMT3b in PBMCs were obviously elevated in low, middle, and high dose WJR groups, and the MTX group (all P <0. 01). There was no sig- nificant difference in expression levels of DNMT1, DNMT3a, or DNMT3b in PBMCs among low, middle, and high dose WJR groups (P>0.05). Conclusions Expression levels of DNMTs in PBMCs of CIA rats decreased. WJR up-regulated the expression level of DNMTs in PBMCs of CIA rats in no obvious dose de- pendent way. One of WJR's mechanisms for treating CIA might be up-regulating expression levels of DN- MTs, and adjusting the state of DNA methylation.


Subject(s)
Arthritis , DNA Methylation , DNA Modification Methylases , Leukocytes, Mononuclear , Animals , Arthritis/drug therapy , Arthritis/metabolism , Cattle , Collagen , DNA , DNA Methylation/drug effects , DNA Modification Methylases/drug effects , DNA Modification Methylases/metabolism , Drugs, Chinese Herbal/pharmacology , Rats , Rats, Wistar
13.
Endocrinology ; 156(12): 4672-83, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26372177

ABSTRACT

The increasing incidence of reproductive anomalies, described as testicular dysgenesis syndrome, is thought to be related to the exposure of the population to chemicals in the environment. Bisphenol A (BPA) and di(2-ethylhexyl)phthalate (DEHP), which have hormonal and antihormonal activity, have attracted public attention due to their presence in consumer products. The present study investigated the effects of BPA and DEHP on reproductive development. Timed-pregnant female rats were exposed to BPA and DEHP by gavage from gestational days 12 to 21. Results showed that prenatal exposures to test chemicals exerted variable effects on steroidogenic factor 1 and GATA binding protein 4 protein expression and increased (P < .05) sex-determining region Y-box 9 and antimüllerian hormone protein in the infantile rat testis compared with levels in the control unexposed animals. Pituitary LHß and FSHß subunit protein expression was increased (P < .05) in BPA- and DEHP-exposed prepubertal male rats but were decreased (P < .05) in adult animals relative to control. Exposure to both BPA and DEHP in utero inhibited (P < .05) global DNA hydroxymethylation in the adult testis in association with altered DNA methyltransferase protein expression. Together the present data suggest that altered developmental programming in the testes associated with chemical exposures are related to the disruption of sexual differentiation events and DNA methylation patterns. The chemical-induced effects impact the development of steroidogenic capacity in the adult testis.


Subject(s)
Benzhydryl Compounds/pharmacology , Diethylhexyl Phthalate/pharmacology , Environmental Pollutants/pharmacology , Estrogens, Non-Steroidal/pharmacology , Phenols/pharmacology , Plasticizers/pharmacology , Sex Differentiation/drug effects , Testis/drug effects , Animals , Anti-Mullerian Hormone/metabolism , DNA Methylation/drug effects , DNA Modification Methylases/drug effects , DNA Modification Methylases/metabolism , Endocrine Disruptors/pharmacology , Female , Follicle Stimulating Hormone, beta Subunit/drug effects , Follicle Stimulating Hormone, beta Subunit/metabolism , GATA4 Transcription Factor/drug effects , GATA4 Transcription Factor/metabolism , Gonadal Dysgenesis , Luteinizing Hormone, beta Subunit/drug effects , Luteinizing Hormone, beta Subunit/metabolism , Male , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Sex-Determining Region Y Protein/drug effects , Sex-Determining Region Y Protein/metabolism , Steroidogenic Factor 1/drug effects , Steroidogenic Factor 1/metabolism , Testicular Diseases , Testis/metabolism
14.
Neurochem Res ; 37(6): 1192-200, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22228201

ABSTRACT

Glioblastoma multiforme (GBM) is recognized as the most common and lethal form of central nervous system cancer. Currently used surgical techniques, chemotherapeutic agents, and radiotherapy strategies have done very little in extending the life expectancies of patients diagnosed with GBM. The difficulty in treating this malignant disease lies both in its inherent complexity and numerous mechanisms of drug resistance. In this review, we summarize several of the primary mechanisms of drug resistance. We reviewed available published literature in the English language regarding drug resistance in glioblastoma. The reasons for drug resistance in glioblastoma include drug efflux, hypoxic areas of tumor cells, cancer stem cells, DNA damage repair, and miRNAs. Many potential therapies target these mechanisms, including a series of investigated alternative and plant-derived agents. Future research and clinical trials in glioblastoma patients should pursue combination of therapies to help combat drug resistance. The emerging new data on the potential of plant-derived therapeutics should also be closely considered and further investigated.


Subject(s)
Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Allyl Compounds/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Cell Cycle Checkpoints/drug effects , Combined Modality Therapy , DNA Modification Methylases/drug effects , DNA Repair/drug effects , DNA Repair Enzymes/drug effects , Flavonoids/therapeutic use , Garlic/chemistry , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , MicroRNAs/therapeutic use , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-bcl-2/drug effects , RNA, Small Interfering/therapeutic use , Retinoids/therapeutic use , Sulfides/therapeutic use , Tumor Suppressor Proteins/drug effects
16.
J Neurooncol ; 92(2): 227-32, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19037596

ABSTRACT

We report a case of a 51-year-old woman with newly diagnosed glioblastoma multiforme (GBM) who was treated with surgery followed by the standard concomitant temozolomide (TMZ) and radiotherapy (RT). Although TMZ is generally safe and well-tolerated, she developed a sudden onset of prolonged and severe thrombocytopenia as the most prominent event of pancytopenia during the combined treatment, leading to discontinuation of the combined therapy. Thrombocytopenia lasted for more than 2 months with intensive, intermittent platelet transfusions. A bone marrow aspiration and biopsy performed after recovery of severe suppression still revealed reduced number of megakaryocytes. O(6)-methylguanine-DNA methyltransferase (MGMT) analyses showed methylated MGMT promoter in GBM, but unmethylated promoters in both peripheral blood leukocytes and bone marrow cells. This is the first report suggesting the irrelevance of MGMT status of normal hematopoietic cells to TMZ-induced severe thrombocytopenia and pancytopenia.


Subject(s)
Brain Neoplasms/therapy , DNA Modification Methylases/drug effects , DNA Repair Enzymes/drug effects , Glioblastoma/therapy , Pancytopenia/etiology , Thrombocytopenia/etiology , Tumor Suppressor Proteins/drug effects , Antineoplastic Agents, Alkylating/adverse effects , Base Sequence , Blotting, Western , Brain Neoplasms/pathology , Combined Modality Therapy , DNA Methylation/drug effects , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/adverse effects , Dacarbazine/analogs & derivatives , Female , Glioblastoma/pathology , Humans , Magnetic Resonance Imaging , Middle Aged , Neurosurgical Procedures , Polymerase Chain Reaction , Radiotherapy/adverse effects , Temozolomide , Tumor Suppressor Proteins/genetics
17.
Cleft Palate Craniofac J ; 45(5): 545-51, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18788878

ABSTRACT

OBJECTIVES: The aim of this study was to analyze epigenetic (specifically, DNA methylation) participation in the mechanisms of cleft palate only induced by maternal exposure to all-trans retinoic acid in mice. DESIGN: Cleft palate only was induced in fetuses by maternal exposure to all-trans retinoic acid. Their secondary palates were excised for analysis. Cytosine extension assay and restriction landmark genomic scanning were performed to analyze DNA methylation status. The expression levels of the DNA methyltransferases were examined by real-time reverse transcriptase-polymerase chain reaction. RESULTS: Using cytosine extension assay, on gestation day 14.5, the status of DNA methylation within CpG islands and in global DNA was decreased significantly in all-trans retinoic acid-treated groups compared with the controls (p < .01 and p < .05). In the controls, the status within CpG islands on gestation day 14.5 was significantly increased compared with gestation days 13.5 and 18.5 (p < .01). Using real-time reverse transcriptase-polymerase chain reaction, there was no significant change in the expression of DNA methyltransferases, except on gestation day 18.5. Using restriction landmark genomic scanning on gestation day 18.5, five spots (0.49%) in the controls and one spot (0.1%) in all-trans retinoic acid-treated groups were specifically detected. CONCLUSIONS: These results indicate that changes in DNA methylation may play an important role in the manifestation of cleft palate only caused by environmental factors such as maternal exposure to all-trans retinoic acid.


Subject(s)
Cleft Palate/chemically induced , DNA Methylation/drug effects , Tretinoin/adverse effects , Animals , Cleft Palate/embryology , CpG Islands/drug effects , Cytosine/analysis , DNA/analysis , DNA/drug effects , DNA Modification Methylases/analysis , DNA Modification Methylases/drug effects , Epigenesis, Genetic/drug effects , Female , Gestational Age , Maternal Exposure/adverse effects , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Palate/drug effects , Palate/embryology , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction
18.
Carcinogenesis ; 29(6): 1267-75, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18204074

ABSTRACT

Nickel (Ni) compounds are potent carcinogens and can induce malignant transformation of rodent and human cells. To uncover the molecular mechanisms of nickel sulfide (NiS)-induced cell transformation, we investigated epigenetic alterations in a set of DNA repair genes. The silencing of the O(6)-methylguanine DNA methyltransferase (MGMT) gene locus and upregulation of DNA methyltransferase 1 (DNMT1) expression was specifically detected in NiS-transformed human bronchial epithelial (16HBE) cells. In addition, we noted epigenetic alterations including DNA hypermethylation, reduced histone H4 acetylation and a decrease in the ratio of Lys-9 acetylated/methylated histone H3 at the MGMT CpG island in NiS-transformed 16HBE cells. Meanwhile, we identified concurrent binding of methyl-CpG-binding protein 2, methylated DNA-binding domain protein 2 and DNMT1 to the CpG island of the MGMT promoter, demonstrating that these components collaborate to maintain MGMT methylation in NiS-transformed cells. Moreover, depletion of DNMT1 by introduction of a small hairpin RNA construct into NiS-transformed cells resulted in a 30% inhibition of cell proliferation and led to increased MGMT gene expression by reversion of the epigenetic modifications at the MGMT promoter region. MGMT suppression and hypermethylation at the CpG island of the MGMT promoter occurred 6 days after NiS treatment, indicating that epigenetic modifications of MGMT might be an early event in tumorigenesis. Taken together, these observations demonstrate that epigenetic silencing of MGMT is associated with DNA hypermethylation, histone modifications and DNMT1 upregulation, which contribute to NiS-induced malignant transformation.


Subject(s)
Carcinogens/toxicity , Cell Transformation, Neoplastic/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Gene Silencing , Nickel/toxicity , Tumor Suppressor Proteins/genetics , Blotting, Western , Cell Line , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/drug effects , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Modification Methylases/drug effects , DNA Modification Methylases/metabolism , DNA Repair Enzymes/drug effects , DNA Repair Enzymes/metabolism , Gene Expression , Histones/genetics , Humans , Immunoprecipitation , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Suppressor Proteins/drug effects , Tumor Suppressor Proteins/metabolism
19.
Clin Adv Hematol Oncol ; 5(7): 544-52, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17679928

ABSTRACT

Aberrant DNA methylation is one of the molecular hallmarks of cancer and leukemia. By repressing gene expression, it is considered a functional equivalent to the physical inactivation of tumor suppressor genes by deletions or mutations. To clinically exploit this process, compounds with DNA hypomethylating properties have been evaluated both in the laboratory and the clinic. Two such agents, 5-azacytidine and 5-aza-2'-deoxycytidine, are currently approved by the US Food and Drug Administration for the treatment of patients with myelodysplastic syndromes. Ongoing studies are evaluating alternative dosing schedules for these drugs and the activity and safety of this class of agent in combination with histone deacetylase inhibitors. Here we summarize the experience of hypomethylating agents in myelodysplastic syndromes.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , DNA Methylation/drug effects , DNA Modification Methylases/drug effects , Myelodysplastic Syndromes/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Decitabine , Histone Deacetylase Inhibitors , Humans , Randomized Controlled Trials as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...