Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 480
1.
Nucleic Acids Res ; 50(2): e9, 2022 01 25.
Article En | MEDLINE | ID: mdl-34718755

Epigenetic therapy has significant potential for cancer treatment. However, few small potent molecules have been identified against DNA or RNA modification regulatory proteins. Current approaches for activity detection of DNA/RNA methyltransferases and demethylases are time-consuming and labor-intensive, making it difficult to subject them to high-throughput screening. Here, we developed a fluorescence polarization-based 'High-Throughput Methyl Reading' (HTMR) assay to implement large-scale compound screening for DNA/RNA methyltransferases and demethylases-DNMTs, TETs, ALKBH5 and METTL3/METTL14. This assay is simple to perform in a mix-and-read manner by adding the methyl-binding proteins MBD1 or YTHDF1. The proteins can be used to distinguish FAM-labelled substrates or product oligonucleotides with different methylation statuses catalyzed by enzymes. Therefore, the extent of the enzymatic reactions can be coupled with the variation of FP binding signals. Furthermore, this assay can be effectively used to conduct a cofactor competition study. Based on the assay, we identified two natural products as candidate compounds for DNMT1 and ALKBH5. In summary, this study outlines a powerful homogeneous approach for high-throughput screening and evaluating enzymatic activity for DNA/RNA methyltransferases and demethylases that is cheap, easy, quick, and highly sensitive.


DNA Modification Methylases/metabolism , Drug Discovery/methods , High-Throughput Screening Assays/methods , Methyltransferases/metabolism , Oxidoreductases, N-Demethylating/metabolism , Carrier Proteins/metabolism , DNA Methylation , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/standards , Humans , Methyltransferases/antagonists & inhibitors , Nucleotides/metabolism , Oxidoreductases, N-Demethylating/antagonists & inhibitors , RNA/metabolism
2.
Pharmacogenet Genomics ; 31(3): 60-67, 2021 04 01.
Article En | MEDLINE | ID: mdl-33075016

OBJECTIVES: Chemotherapy-induced hematological toxicities are potentially life-threatening adverse drug reactions that vary between individuals. Recently, JMJD1C has been associated with gemcitabine/carboplatin-induced thrombocytopenia in non-small-cell lung cancer patients, making it a candidate marker for predicting the risk of toxicity. This study investigates if JMJD1C knockdown affects gemcitabine/carboplatin-sensitivity in cell lines. METHODS: Lentiviral transduction-mediated shRNA knockdown of JMJD1C in the cell lines K562 and MEG-01 were performed using shRNA#32 and shRNA#33. The knockdown was evaluated using qPCR. Cell proliferation, viability, and gemcitabine/carboplatin-sensitivity were subsequently determined using cell counts, trypan blue, and the MTT assay. RESULTS: ShRNA#33 resulted in JMJD1C downregulation by 56.24% in K562 and 68.10% in MEG-01. Despite incomplete knockdown, proliferation (reduction of cell numbers by 61-68%, day 7 post-transduction) and viability (reduction by 21-53%, day 7 post-transduction) were impaired in K562 and MEG-01 cells. Moreover, JMJD1C knockdown reduced the gemcitabine IC50-value for K562 cells (P < 0.01) and MEG-01 cells (P < 0.05) compared to scrambled shRNA control transduced cells. CONCLUSIONS: Our results suggest that JMJD1C is essential for proliferation, survival, and viability of K562 and MEG-01 cells. Further, JMJD1C also potentially affects the cells gemcitabine/carboplatin-sensitivity. Although further research is required, the findings show that JMJD1C could have an influential role for gemcitabine/carboplatin-sensitivity.


Cell Proliferation/drug effects , Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Oxidoreductases, N-Demethylating/genetics , Carboplatin/adverse effects , Carboplatin/pharmacology , Cell Survival/drug effects , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Gene Knockdown Techniques , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Myeloid Cells/drug effects , Myeloid Cells/pathology , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Gemcitabine
3.
FEBS Open Bio ; 11(1): 265-277, 2021 01.
Article En | MEDLINE | ID: mdl-33289299

Leukemic stem cells (LSCs) comprise a very rare cell population that results in the development of acute myeloid leukemia. The selective targeting of drivers in LSCs with small molecule inhibitors holds promise for treatment of acute myeloid leukemia. Recently, we reported the identification of inhibitors of the histone lysine demethylase JMJD1C that preferentially kill MLL rearranged acute leukemia cells. Here, we report the identification of jumonji domain modulator #7 (JDM-7). Surface plasmon resonance analysis showed that JDM-7 binds to JMJD1C and its family homolog JMJD1B. JDM-7 did not significantly suppress cell proliferation in liquid cell culture at higher doses, although it led to a significant decrease in semi-solid colony formation experiments at lower concentrations. Moreover, low doses of JDM-7 did not suppress the proliferation of erythroid progenitor cells. We identified that JDM-7 downregulates the LSC self-renewal gene HOXA9 in leukemia cells. We further found that the structure of JDM-7 is similar to that of tadalafil, a drug approved by the US Food and Drug Administration. Molecular docking and surface plasmon resonance analysis showed that tadalafil binds to JMJD1C. Moreover, similar to JDM-7, tadalafil suppressed colony formation of leukemia cells in semi-solid cell culture at a concentration that did not affect primary umbilical cord blood cells. In summary, we have identified JDM-7 and tadalafil as potential JMJD1C modulators that selectively inhibit the growth of LSCs.


Antineoplastic Agents/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/drug effects , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor , Gene Expression Regulation, Leukemic/drug effects , Homeodomain Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Primary Cell Culture , Tadalafil/pharmacology , Tadalafil/therapeutic use
4.
Cancer Biol Med ; 17(1): 9-19, 2020 02 15.
Article En | MEDLINE | ID: mdl-32296573

The biological roles of N6 methylation of nucleic acids have been extensively studied. Adenine methylation of RNA is the most prevalent RNA modification and has widespread effects on RNA splicing, translation, localization, and stability. Aberrant dynamic regulation of RNA N6-methyladenosine (m6A) has been reported in numerous human diseases, including several cancers. In recent years, eukaryotic DNA N6-methyladenosine (6mA) has also been reported and implicated in cancer progression and tumorigenesis. In this review, we summarize the contributions of N6-methyladenosine modification to cancer biology and pathogenesis in the context of both RNA and DNA. We also highlight the clinical relevance of targeting these modifications as a therapeutic strategy for cancer.


DNA Methylation/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasms/genetics , RNA Processing, Post-Transcriptional/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/genetics , DNA Methylation/drug effects , Disease Progression , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/pathology , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/metabolism , RNA Processing, Post-Transcriptional/drug effects , RNA Splicing Factors , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism
5.
J Recept Signal Transduct Res ; 40(4): 339-347, 2020 Aug.
Article En | MEDLINE | ID: mdl-32122211

Pathological cardiac hypertrophy is a major risk factor for cardiovascular morbidity and mortality. Histone demethylases (KDMs) are emerging regulators of transcriptional reprograming in cancer, however, their potential role in abnormal heart growth and fibrosis remains largely unknown. The aim of this current study was to examine the role of JMJD1C, an H3K9me2 specific demethylase, in angiotensin II (Ang II) induced cardiac hypertrophy and fibrosis. In this study, we observed that Ang II could increase the expression of JMJD1C detected by Western blot and RT-qPCR in vitro and in vivo. Immunofluorescence staining showed that the treatment of Ang II could increase cardiomyocyte size. RT-qPCR results have shown that Ang II could increase the expression of cell hypertrophic and fibrotic markers in H9c2 cells. Whereas, inhibition of JMJD1C by shRNA and JIB-04, a small molecule histone demethylase inhibitor, significantly reduced Ang II-induced cell hypertrophy, and hypertrophic and fibrotic marker overexpression. Furthermore, cardiomyocyte JMJD1C knockdown decreased Tissue Inhibitor of Metalloproteinases 1 (TIMP1) transcription with pro-fibrotic activity. In conclusion, JMJD1C plays an important role in Ang II-induced cardiac hypertrophy and fibrosis by activating TIMP1 transcription, targeting of JMJD1C may be an effective strategy for the treatment of Ang II-associated cardiac diseases.


Cardiomegaly/drug therapy , Fibrosis/drug therapy , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-1/genetics , Aminopyridines/pharmacology , Angiotensin II/adverse effects , Angiotensin II/genetics , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Disease Models, Animal , Fibrosis/genetics , Fibrosis/pathology , Gene Expression Regulation/drug effects , Heart/drug effects , Humans , Hydrazones/pharmacology , Jumonji Domain-Containing Histone Demethylases/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Oxidoreductases, N-Demethylating/genetics , RNA, Small Interfering/pharmacology , Rats , Signal Transduction/drug effects
7.
Int J Cancer ; 146(2): 400-412, 2020 01 15.
Article En | MEDLINE | ID: mdl-31271662

Histone demethylases are promising therapeutic targets as they play fundamental roles for survival of Mixed lineage leukemia rearranged acute leukemia (MLLr AL). Here we focused on the catalytic Jumonji domain of histone H3 lysine 9 (H3K9) demethylase JMJD1C to screen for potential small molecular modulators from 149,519 natural products and 33,765 Chinese medicine components via virtual screening. JMJD1C Jumonji domain inhibitor 4 (JDI-4) and JDI-12 that share a common structural backbone were detected within the top 15 compounds. Surface plasmon resonance analysis showed that JDI-4 and JDI-12 bind to JMJD1C and its family homolog KDM3B with modest affinity. In vitro demethylation assays showed that JDI-4 can reverse the H3K9 demethylation conferred by KDM3B. In vivo demethylation assays indicated that JDI-4 and JDI-12 could induce the global increase of H3K9 methylation. Cell proliferation and colony formation assays documented that JDI-4 and JDI-12 kill MLLr AL and other malignant hematopoietic cells, but not leukemia cells resistant to JMJD1C depletion or cord blood cells. Furthermore, JDI-16, among multiple compounds structurally akin to JDI-4/JDI-12, exhibits superior killing activities against malignant hematopoietic cells compared to JDI-4/JDI-12. Mechanistically, JDI-16 not only induces apoptosis but also differentiation of MLLr AL cells. RNA sequencing and quantitative PCR showed that JDI-16 induced gene expression associated with cell metabolism; targeted metabolomics revealed that JDI-16 downregulates lactic acids, NADP+ and other metabolites. Moreover, JDI-16 collaborates with all-trans retinoic acid to repress MLLr AML cells. In summary, we identified bona fide JMJD1C inhibitors that induce preferential death of MLLr AL cells.


Antineoplastic Agents/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Leukemia, Biphenotypic, Acute/drug therapy , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Adult , Aged , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bone Marrow/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Demethylation/drug effects , DNA Methylation/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Female , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Biphenotypic, Acute/pathology , Male , Middle Aged , Molecular Docking Simulation , Oxidoreductases, N-Demethylating/chemistry , Oxidoreductases, N-Demethylating/metabolism , Protein Domains , Structure-Activity Relationship , Surface Plasmon Resonance , Tretinoin/pharmacology , Tretinoin/therapeutic use
8.
Nat Med ; 24(9): 1407-1417, 2018 09.
Article En | MEDLINE | ID: mdl-30082863

Trimethylamine N-oxide (TMAO) is a gut microbiota-derived metabolite that enhances both platelet responsiveness and in vivo thrombosis potential in animal models, and TMAO plasma levels predict incident atherothrombotic event risks in human clinical studies. TMAO is formed by gut microbe-dependent metabolism of trimethylamine (TMA) moiety-containing nutrients, which are abundant in a Western diet. Here, using a mechanism-based inhibitor approach targeting a major microbial TMA-generating enzyme pair, CutC and CutD (CutC/D), we developed inhibitors that are potent, time-dependent, and irreversible and that do not affect commensal viability. In animal models, a single oral dose of a CutC/D inhibitor significantly reduced plasma TMAO levels for up to 3 d and rescued diet-induced enhanced platelet responsiveness and thrombus formation, without observable toxicity or increased bleeding risk. The inhibitor selectively accumulated within intestinal microbes to millimolar levels, a concentration over 1-million-fold higher than needed for a therapeutic effect. These studies reveal that mechanism-based inhibition of gut microbial TMA and TMAO production reduces thrombosis potential, a critical adverse complication in heart disease. They also offer a generalizable approach for the selective nonlethal targeting of gut microbial enzymes linked to host disease limiting systemic exposure of the inhibitor in the host.


Gastrointestinal Microbiome , Thrombosis/microbiology , Animals , Bacteria/drug effects , Bacteria/metabolism , Choline/pharmacology , Diet , Gastrointestinal Microbiome/drug effects , Hexanols/pharmacology , Mice, Inbred C57BL , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/metabolism , Platelet Aggregation/drug effects
9.
Int J Mol Sci ; 17(5)2016 Apr 29.
Article En | MEDLINE | ID: mdl-27136547

Perilipins are evolutionarily conserved from Drosophila to humans, the lipid storage droplet 1 (Lsd1) is a Drosophila homolog of human perilipin 1. The function of Lsd1 as a regulator of lipolysis in Drosophila has been demonstrated, as the Lsd1 mutant causes an increase of lipid droplet size. However, the functions of this gene during development are still under investigation. In order to determine the function of Lsd1 during development, Lsd1 was knocked down in Drosophila using the GAL4-UAS system. Selective knockdown of Lsd1 in the dorsal wing disc caused an atrophied wing phenotype. The generation of reactive oxygen species in the wing pouch compartment of the Lsd1-knockdown flies was significantly higher than in the control. Immunostaining with caspase-3 antibody revealed a greater number of apoptotic cells in Lsd1-knockdown wing discs than in the control. Cell death by autophagy was also induced in the knockdown flies. Moreover, cells deprived of Lsd1 showed mitochondrial expansion and decreased ATP levels. These results strongly suggest that knockdown of Lsd1 induces mitochondrial stress and the production of reactive oxygen species that result in cell death, via apoptosis and the autophagy pathway. These results highlight the roles of Drosophila Lsd1 during wing development.


Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Oxidoreductases, N-Demethylating/metabolism , Adenosine Triphosphate/metabolism , Animals , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Autophagy , Caspase 3/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Immunohistochemistry , Lipids/chemistry , Microscopy, Fluorescence , Mitochondria/metabolism , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/genetics , Phenotype , RNA Interference , Reactive Oxygen Species/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wings, Animal/growth & development , Wings, Animal/metabolism
10.
Sci Transl Med ; 6(265): 265ra169, 2014 Dec 03.
Article En | MEDLINE | ID: mdl-25473037

Herpesviruses are highly prevalent and maintain lifelong latent reservoirs, thus posing challenges to the control of herpetic disease despite the availability of antiviral pharmaceuticals that target viral DNA replication. The initiation of herpes simplex virus infection and reactivation from latency is dependent on a transcriptional coactivator complex that contains two required histone demethylases, LSD1 (lysine-specific demethylase 1) and a member of the JMJD2 family (Jumonji C domain-containing protein 2). Inhibition of either of these enzymes results in heterochromatic suppression of the viral genome and blocks infection and reactivation in vitro. We demonstrate that viral infection can be epigenetically suppressed in three animal models of herpes simplex virus infection and disease. Treating animals with the monoamine oxidase inhibitor tranylcypromine to inhibit LSD1 suppressed viral lytic infection, subclinical shedding, and reactivation from latency in vivo. This phenotypic suppression was correlated with enhanced epigenetic suppression of the viral genome and suggests that, even during latency, the chromatin state of the virus is dynamic. Therefore, epi-pharmaceuticals may represent a promising approach to treat herpetic diseases.


Epigenesis, Genetic , Herpesviridae Infections/metabolism , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/physiology , Animals , Disease Models, Animal , Female , Genome, Viral , Guinea Pigs , Histone Demethylases , Mice , Mice, Inbred BALB C , Monoamine Oxidase Inhibitors/chemistry , Phenotype , Protein Structure, Tertiary , Rabbits , Recurrence , Tranylcypromine/chemistry , Vagina/virology , Virus Activation , Virus Latency , Virus Replication/drug effects , Virus Shedding
11.
Blood ; 123(12): 1870-82, 2014 Mar 20.
Article En | MEDLINE | ID: mdl-24501218

Epigenetic regulatory mechanisms are implicated in the pathogenesis of acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL). Recent progress suggests that proteins involved in epigenetic control are amenable to drug intervention, but little is known about the cancer-specific dependency on epigenetic regulators for cell survival and proliferation. We used a mouse model of human AML induced by the MLL-AF9 fusion oncogene and an epigenetic short hairpin RNA (shRNA) library to screen for novel potential drug targets. As a counter-screen for general toxicity of shRNAs, we used normal mouse bone marrow cells. One of the best candidate drug targets identified in these screens was Jmjd1c. Depletion of Jmjd1c impairs growth and colony formation of mouse MLL-AF9 cells in vitro as well as establishment of leukemia after transplantation. Depletion of JMJD1C impairs expansion and colony formation of human leukemic cell lines, with the strongest effect observed in the MLL-rearranged ALL cell line SEM. In both mouse and human leukemic cells, the growth defect upon JMJD1C depletion appears to be primarily due to increased apoptosis, which implicates JMJD1C as a potential therapeutic target in leukemia.


Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia, Myeloid, Acute/genetics , Oxidoreductases, N-Demethylating/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Epigenesis, Genetic , Gene Knockdown Techniques , Genes, myb , Genes, myc , Histone-Lysine N-Methyltransferase/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/pathology , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Oxidoreductases, N-Demethylating/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Tumor Stem Cell Assay
12.
Stem Cell Res ; 11(3): 1299-313, 2013 Nov.
Article En | MEDLINE | ID: mdl-24084696

MicroRNA-137 (miR-137) has been shown to play an important role in the differentiation of neural stem cells. Embryonic stem (ES) cells have the potential to differentiate into different cell types including neurons; however, the contribution of miR-137 in the maintenance and differentiation of ES cells remains unknown. Here, we show that miR-137 is mainly expressed in ES cells at the mitotic phase of the cell cycle and highly upregulated during differentiation. We identify that ES cell transcription factors, Klf4 and Tbx3, are downstream targets of miR-137, and we show that endogenous miR-137 represses the 3' untranslated regions of Klf4 and Tbx3. Transfection of ES cells with mature miR-137 RNA duplexes led to a significant reduction in cell proliferation and the expression of Klf4, Tbx3, and other self-renewal genes. Furthermore, we demonstrate that increased miR-137 expression accelerates differentiation of ES cells in vitro. Loss of miR-137 during ES cell differentiation significantly impeded neuronal gene expression and morphogenesis. Taken together, our results suggest that miR-137 regulates ES cell proliferation and differentiation by repressing the expression of downstream targets, including Klf4 and Tbx3.


Cell Differentiation , Embryonic Stem Cells/metabolism , Kruppel-Like Transcription Factors/antagonists & inhibitors , MicroRNAs/metabolism , T-Box Domain Proteins/antagonists & inhibitors , 3' Untranslated Regions , Animals , Base Sequence , Cell Division , Cell Lineage , Cell Proliferation , Embryonic Stem Cells/cytology , Histone Demethylases , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , MicroRNAs/genetics , Mitosis , Neurons/cytology , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transfection , Up-Regulation
13.
Chem Biol Interact ; 205(1): 11-9, 2013 Sep 05.
Article En | MEDLINE | ID: mdl-23777987

The present study was performed to evaluate the potency and specificity of sibutramine as an inhibitor of the activities of nine human CYP isoforms in liver microsomes. Using a cocktail assay, the effects of sibutramine on specific marker reactions of the nine CYP isoforms were measured in human liver microsomes. Sibutramine showed potent inhibition of CYP2B6-mediated bupropion 6-hydroxylation with an IC50 value of 1.61µM and Ki value of 0.466µM in a competitive manner at microsomal protein concentrations of 0.25mg/ml; this was 3.49-fold more potent than the typical CYP2B6 inhibitor thio-TEPA (Ki=1.59µM). In addition, sibutramine slightly inhibited CYP2C19 activity (Ki=16.6µM, noncompetitive inhibition) and CYP2D6 activity (Ki=15.7µM, noncompetitive inhibition). These observations indicated 35.6- and 33.7-fold decreases in inhibition potency, respectively, compared with that of CYP2B6 by sibutramine. However, no inhibition of CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2D6, or CYP2E1 activities was observed. In addition, the CYP2B6 inhibitory potential of sibutramine was enhanced at a lower microsomal protein concentration of 0.05mg/ml. After 30min preincubation of human liver microsomes with sibutramine in the presence of NADPH, no shift in IC50 was observed in terms of inhibition of the activities of the nine CYPs, suggesting that sibutramine is not a time-dependent inactivator. These observations suggest that sibutramine is a selective and potent inhibitor of CYP2B6 in vitro, whereas inhibition of other CYPs is substantially lower. These in vitro data support the use of sibutramine as a well-known inhibitor of CYP2B6 for routine screening of P450 reversible inhibition when human liver microsomes are used as the enzyme source.


Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Cyclobutanes/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Cyclobutanes/pharmacokinetics , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2D6 Inhibitors , Cytochrome P-450 Enzyme Inhibitors , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Female , Humans , Kinetics , Male , Microsomes, Liver/metabolism , Thiotepa/pharmacology
14.
Cell Rep ; 3(4): 1071-9, 2013 Apr 25.
Article En | MEDLINE | ID: mdl-23545502

The functional organization of eukaryotic genomes correlates with specific patterns of histone methylations. Regulatory regions in genomes such as enhancers and promoters differ in their extent of methylation of histone H3 at lysine-4 (H3K4), but it is largely unknown how the different methylation states are specified and controlled. Here, we show that the Kdm5c/Jarid1c/SMCX member of the Kdm5 family of H3K4 demethylases can be recruited to both enhancer and promoter elements in mouse embryonic stem cells and in neuronal progenitor cells. Knockdown of Kdm5c deregulates transcription via local increases in H3K4me3. Our data indicate that by restricting H3K4me3 modification at core promoters, Kdm5c dampens transcription, but at enhancers Kdm5c stimulates their activity. Remarkably, an impaired enhancer function activates the intrinsic promoter activity of Kdm5c-bound distal elements. Our results demonstrate that the Kdm5c demethylase plays a crucial and dynamic role in the functional discrimination between enhancers and core promoters.


Histones/metabolism , Oxidoreductases, N-Demethylating/metabolism , Animals , Binding Sites , Cells, Cultured , Embryonic Stem Cells , Histone Demethylases , Histones/genetics , Methylation , Mice , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/genetics , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism
15.
Comp Biochem Physiol C Toxicol Pharmacol ; 157(4): 329-36, 2013 May.
Article En | MEDLINE | ID: mdl-23474500

The measurement of enzymatic activities involved in xenobiotic biotransformation was carried out in adults of Solea solea and Solea senegalensis. The hepatic enzymes analysed were cytochrome P450 (CYP) related activities using eight fluorometric substrates and carboxylesterases (CbE). The conjugating activities of glutathione S-transferase (GST) and UPD-glucuronosyltransferase (UDPGT) were also assessed. Specific mammalian inhibitors were used as diagnostic tools for related activities of CYP1A (α-naphthoflavone; αNF), CYP2B6 and CYP2C19 (ticlopidine) and CYP3A4 (ketoconazole). The in vitro sensitivity to organophosphorous pesticides (OP) was tested in the S10 homogenate of brain (acetylcholinesterase-AChE) and liver (CbE). Furthermore, the pesticide chlorpyrifos oxon (CLPO) was used to explore the OP sensitivity of CbE of both species in two subcellular fractions (microsomes and cytosol), using two substrates. Overall, only two parameters confirmed species differences: EROD and cytosolic CbE being significantly elevated (p < 0.05) in the common sole, S. solea. A high inhibition of CYP1A related activities using several fluorometric substrates (ER, MR and CEC) after in vitro incubation with αNF confirmed all measure CYP1A1-related activities whereas ketoconazole was more specific for BFCOD (CYP3A4). Pesticide sensitivity was similar for brain AChE but hepatic CbE had a protective role that was species and pesticide dependent.


Flatfishes/metabolism , Pesticides/pharmacology , Xenobiotics/metabolism , Acetylcholinesterase/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/metabolism , Brain/drug effects , Brain/enzymology , Chlorpyrifos/analogs & derivatives , Chlorpyrifos/pharmacology , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP2B6 , Enzyme Activation , Female , Glutathione Transferase/metabolism , Ketoconazole/pharmacology , Liver/drug effects , Liver/enzymology , Male , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/metabolism , Pesticides/metabolism , Species Specificity , Ticlopidine/pharmacology
16.
J Clin Pharmacol ; 53(5): 550-8, 2013 May.
Article En | MEDLINE | ID: mdl-23381968

In this study, we assessed the effects of clopidogrel and clarithromycin, known CYP2B6 and CYP3A inhibitors, respectively, on the enantioselective disposition of racemic sibutramine in conjunction with CYP2B6 polymorphisms in humans. Sibutramine showed enantioselective plasma profiles with consistently higher concentrations of R-enantiomers. Clopidogrel and clarithromycin significantly increased the sibutramine plasma concentration, but their effects differed between enantiomers; a 2.2-fold versus 4.1-fold increase in the AUC in S-enantiomer and 1.8-fold versus 2.0-fold for the R-enantiomer, respectively. The AUCs of S- and R-desmethyl metabolites changed significantly during the clopidogrel phase (P < .001 and P < .001, respectively) but not during the clarithromycin phase (P = .099 and P = .090, respectively). Exposure to sibutramine was higher in subjects with the CYP2B6*6/*6 genotype, but no statistical difference was observed among the CYP2B6 genotypes. These results suggest that the enantioselective disposition of sibutramine and its active metabolites are influenced by the altered genetic and environmental factors of CYP2B6 and CYP3A activity in vivo.


Appetite Depressants/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Clarithromycin/pharmacology , Cyclobutanes/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Ticlopidine/analogs & derivatives , Adult , Appetite Depressants/chemistry , Aryl Hydrocarbon Hydroxylases/genetics , Clopidogrel , Cross-Over Studies , Cyclobutanes/blood , Cyclobutanes/chemistry , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A , Genotype , Humans , Male , Microsomes/metabolism , Oxidoreductases, N-Demethylating/genetics , Polymorphism, Genetic , Stereoisomerism , Ticlopidine/pharmacology , Young Adult
17.
J Clin Pharmacol ; 53(3): 305-13, 2013 Mar.
Article En | MEDLINE | ID: mdl-23361846

Methadone N-demethylation in vitro is catalyzed by hepatic cytochrome P4502B6 (CYP2B6) and CYP3A4, but clinical disposition is often attributed to CYP3A4. This investigation tested the hypothesis that CYP2B6 is a prominent CYP isoform responsible for clinical methadone N-demethylation and clearance, using the in vivo mechanism-based CYP2B6 inhibitor ticlopidine, given orally for 4 days. A preliminary clinical investigation with the CYP3A4/5 substrate probe alfentanil established that ticlopidine did not inhibit intestinal or hepatic CYP3A4/5. Subjects received intravenous plus oral (deuterium-labeled) racemic methadone before and after ticlopidine. Ticlopidine significantly and stereoselectively (S > R) inhibited methadone N-demethylation, decreasing plasma metabolite/methadone area under the curve ratios and metabolite formation clearances. Ticlopidine also significantly increased the dose-adjusted plasma area under the curve for R- and S-methadone by 20% and 60%, respectively, after both intravenous and oral dosing. CYP2B6 inhibition reduces methadone N-demethylation and clearance, and alters methadone concentrations, demonstrating an important role for CYP2B6 in clinical methadone disposition.


Analgesics, Opioid/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/metabolism , Methadone/pharmacokinetics , Oxidoreductases, N-Demethylating/metabolism , Administration, Intravenous , Administration, Oral , Adult , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/blood , Analgesics, Opioid/urine , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Cytochrome P-450 CYP2B6 , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Methadone/administration & dosage , Methadone/blood , Methadone/urine , Middle Aged , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Ticlopidine/pharmacology , Young Adult
18.
Xenobiotica ; 43(2): 211-8, 2013 Feb.
Article En | MEDLINE | ID: mdl-22830954

Plasma concentrations of sibutramine and its two active metabolites after single oral dose of sibutramine were determined in Korean healthy male subjects with different CYP2B6 genotypes (CYP2B6*1/*1, *1/*6 and *6/*6), either alone or after four-day pretreatment with clopidogrel or clarithromycin. The pretreatment with clopidogrel and clarithromycin raised the mean area under the concentration-time curve (AUC) of sibutramine by 163% and 255%, respectively. Co-administration of clarithromycin, combined with CYP2B6*6/*6 genotype, led to highest concentration of sibutramine. The molar sum AUC (M1 + M2) was raised by 35% in the clopidogrel phase but not significantly affected by clarithromycin or CYP2B6 genotype. The CYP2B6*6/*6 subjects in the clopidogrel phase showed the highest molar AUC (M1 + M2) among three genotype groups throughout the three phases. The exposure of sibutramine and its metabolites seemed to be associated with the CYP2B6 genotype. The treatment of clopidogrel significantly altered the disposition of active metabolites as well as sibutramine, but clarithromycin only affects the disposition of sibutramine. These results suggest that the perturbation of CYP2B6 activity may contribute to the inter-individual variation of sibutramine drug responses although the clinical relevance is remained to be established.


Appetite Depressants/metabolism , Aryl Hydrocarbon Hydroxylases/genetics , Clarithromycin/pharmacology , Cyclobutanes/metabolism , Oxidoreductases, N-Demethylating/genetics , Ticlopidine/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Appetite Depressants/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/metabolism , Asian People , Clopidogrel , Cyclobutanes/pharmacokinetics , Cytochrome P-450 CYP2B6 , Humans , Male , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/metabolism , Platelet Aggregation Inhibitors/pharmacology , Polymorphism, Genetic , Republic of Korea , Ticlopidine/pharmacology
19.
Eur J Clin Pharmacol ; 69(4): 867-75, 2013 Apr.
Article En | MEDLINE | ID: mdl-23099620

PURPOSE: We assessed possible drug interactions of tramadol given concomitantly with the potent CYP2B6 inhibitor ticlopidine, alone or together with the potent CYP3A4 and P-glycoprotein inhibitor itraconazole. METHODS: In a randomized, placebo-controlled cross-over study, 12 healthy subjects ingested 50 mg of tramadol after 4 days of pretreatment with either placebo, ticlopidine (250 mg twice daily) or ticlopidine plus itraconazole (200 mg once daily). Plasma and urine concentrations of tramadol and its active metabolite O-desmethyltramadol (M1) were monitored over 48 h and 24 h, respectively. RESULTS: Ticlopidine increased the mean area under the plasma concentration-time curve (AUC0-∞) of tramadol by 2.0-fold (90 % confidence interval (CI) 1.6-2.4; p < 0.001) and Cmax by 1.4-fold (p < 0.001), and reduced its oral and renal clearance (p < 0.01). Ticlopidine reduced the AUC0-3 of M1 (p < 0.001) and the ratio of the AUC0-∞ of M1 to that of tramadol, but did not influence the AUC0-∞ of M1. Tramadol or M1 pharmacokinetics did not differ between the ticlopidine alone and ticlopidine plus itraconazole phases. CONCLUSIONS: Ticlopidine increased exposure to tramadol, reduced its renal clearance and inhibited the formation of M1, most likely via inhibition of CYP2B6 and/or CYP2D6. The addition of itraconazole to ticlopidine did not modify the outcome of the drug interaction. Concomitant clinical use of ticlopidine and tramadol may enhance the risk of serotonergic effects, especially when higher doses of tramadol are used.


Analgesics, Opioid/pharmacokinetics , Itraconazole/pharmacology , Kidney/metabolism , Ticlopidine/pharmacology , Tramadol/analogs & derivatives , Adult , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/blood , Analgesics, Opioid/urine , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Biotransformation , Cross-Over Studies , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inhibitors , Dose-Response Relationship, Drug , Drug Interactions , Drug Therapy, Combination , Female , Humans , Itraconazole/administration & dosage , Male , Metabolic Clearance Rate , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Ticlopidine/administration & dosage , Tramadol/administration & dosage , Tramadol/blood , Tramadol/pharmacokinetics , Tramadol/urine , Young Adult
20.
Drug Metab Dispos ; 40(12): 2256-66, 2012 Dec.
Article En | MEDLINE | ID: mdl-22936314

Selegiline, the R-enantiomer of deprenyl, is used in the treatment of Parkinson's disease. Bupropion, an antidepressant, often used to treat patients in conjunction with selegiline, is metabolized primarily by CYP2B6. The effect of selegiline on the enzymatic activity of human cytochrome CYP2B6 in a reconstituted system and its effect on the metabolism of bupropion were examined. Selegiline was found to be a mechanism-based inactivator of the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation (7-EFC) activity of CYP2B6 as well as bupropion metabolism. The inactivations were time-, concentration-, and NADPH-dependent and were characterized by K(I) values of 0.14 and 0.6 µM, k(inact) values of 0.022 and 0.029 min⁻¹, and t(½) values of 31.5 and 24 min, respectively. In standard inhibition assays, selegiline increased the K(m) of CYP2B6 for bupropion from 10 to 92 µM and decreased the k(cat) by ∼50%. The reduced carbon-monoxide difference spectrum revealed over a 50% loss in the cytochrome P450 spectrum in the inactivated sample, with no loss in heme, and there was ∼70% loss in enzyme activity. Trapping of the reactive metabolite using GSH led to the identification of a GSH-selegiline conjugate with a m/z 528 that could be explained by hydroxylation of selegiline followed by the addition of glutathione to the propargyl moiety after oxygenation to form the ketene intermediate. Liquid chromatography-tandem mass spectrometry analysis of the labeled protein following digestion with trypsin revealed the peptide 64DVFTVHLGPR7³ as the peptide modified by the reactive metabolite of selegiline and the site of adduct formation is Asp64.


Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Bupropion/metabolism , Glutathione/metabolism , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Peptides/metabolism , Selegiline/pharmacology , Amino Acid Sequence , Antidepressive Agents, Second-Generation/metabolism , Antiparkinson Agents/pharmacology , Aryl Hydrocarbon Hydroxylases/metabolism , Chromatography, Liquid/methods , Cytochrome P-450 CYP2B6 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Escherichia coli/metabolism , Heme/metabolism , Humans , Hydroxylation/drug effects , Molecular Sequence Data , NADP/metabolism , Oxidoreductases, N-Demethylating/metabolism , Tandem Mass Spectrometry/methods
...