Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Drug Dev Res ; 84(8): 1724-1738, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37756467

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) and STAT5 are the transcription factors that have been studied extensively in relevance to the development of cancers in humans. Suppression of either STAT3 or STAT5-mediated signaling events has been demonstrated to be effective in inducing cytotoxicity in cancer cells. Herein, new hybrids of triazolyl-indolo-quinoxaline are synthesized and examined for their effect on the activation of STAT3 and STAT5 pathways in gastric cancer (GC) cells. Among the newly synthesized compounds, 2,3-difluoro-6-((1-(3-fluorophenyl)-1H-1,2,3-triazol-5-yl)methyl)-6H-indolo[2,3-b]quinoxaline (DTI) displayed selective cytotoxicity against GC cells over their normal counterpart. Flow cytometric analysis, annexin-V-fluorescein isothiocyanate staining, terminal deoxynucleotidyl transferase dUTP nick end labeling assay, live and dead assay, and caspase activation experiments suggested DTI as a potent inducer of apoptosis. The mechanistic approach revealed that DTI imparts cytotoxicity via downregulating the phosphorylation of STAT3Y705 and STAT5Y694/699 . DTI significantly reduced the nuclear pool of STAT3/STAT5 and reduced the DNA interaction ability of STAT3/STAT5 as evidenced by immunofluorescence and electrophoretic mobility shift assay. Further investigation revealed that inhibitory effects towards STAT proteins were mediated through the suppression of upstream kinases such as JAK1, JAK2, and Src. Treatment of GC cells with pervanadate counteracted the DTI-driven STAT3/STAT5 inhibition suggesting the involvement of tyrosine phosphatase. Upon DTI exposure, there was a significant upregulation in the mRNA and protein expression of PTPεC, which is a negative regulator of the JAK-STAT pathway. Knockdown of PTPεC suppressed the DTI-induced STATs inhibition in GC cells. Taken together, triazolyl-indolo-quinoxaline is presented as a new inhibitor of the STAT3/STAT5 pathway in GC cells.


Subject(s)
Signal Transduction , Stomach Neoplasms , Humans , STAT5 Transcription Factor/metabolism , STAT5 Transcription Factor/pharmacology , STAT3 Transcription Factor/metabolism , DNA-Binding Proteins/metabolism , Trans-Activators , Up-Regulation , Quinoxalines/pharmacology , Janus Kinases/metabolism , Janus Kinases/pharmacology , STAT Transcription Factors/metabolism , STAT Transcription Factors/pharmacology , Phosphorylation , Apoptosis
2.
Anticancer Agents Med Chem ; 23(17): 1924-1931, 2023.
Article in English | MEDLINE | ID: mdl-37469157

ABSTRACT

BACKGROUND: Indazoles are known for their anti-cancer properties. OBJECTIVE: The current investigation was on the synthesis and evaluation of novel indazole derivatives for their anticancer properties. METHODS: A series of novel indazoles were synthesized and characterized by IR, NMR and LCMS. We performed cytotoxic studies for all synthesized compounds on different cell lines such as HeLa, MCF-7 and EAC using MTT assay. The lead compound was tested further for its anti-tumor and anti-angiogenic effect on EAT tumor model. RESULTS: Amongst the series of compounds synthesized, compound KA8 showed potent antiproliferative effect against Hela, MCF-7 and EAC cell lines with IC50 values 10.4 to 11.5 and 13.5 µM respectively. In addition, our compound KA8 significantly decreased the cell viability, body weight, ascites volume and it also showed superior survival ability of mice compared to control groups. Furthermore, it suppressed the formation of neovasculature in the peritoneum of EAT-bearing mice. CONCLUSION: The findings reveal that the lead compound KA8 possesses potent anti-tumor and anti-angiogenic properties thereby promising it to be developed as a novel anticancer agent with further mechanistic studies.


Subject(s)
Antineoplastic Agents , Carcinoma, Ehrlich Tumor , Animals , Mice , Cell Line, Tumor , Indazoles/chemistry , Ascites/drug therapy , Cell Proliferation , Antineoplastic Agents/chemistry , Carcinoma, Ehrlich Tumor/drug therapy , Drug Screening Assays, Antitumor , Molecular Structure , Structure-Activity Relationship
3.
RSC Adv ; 13(8): 4910-4916, 2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36762078

ABSTRACT

Highly regioselective synthesis of 2-acyl-4-(het)arylthiazoles and thioethers by the reaction between α-oxothioamides and α-bromoketones in the absence of base in DMF and in the presence of triethylamine in acetonitrile, respectively, has been reported. This thiazole synthesis is an important extended work of the Hantzsch thiazole synthesis, which overcomes the drawbacks of earlier reported methods. The probable mechanisms for the formation of thiazoles and thioethers are also presented.

4.
FEBS J ; 290(3): 796-820, 2023 02.
Article in English | MEDLINE | ID: mdl-36048168

ABSTRACT

Mercaptopyrimidine derivatives are heterocyclic compounds with potent biological activities including antiproliferative, antibacterial, and anti-inflammatory properties. The present study describes the synthesis and characterization of several mercaptopyrimidine derivatives through condensation of 5,6-diamino-2-mercaptopyrimidin-4-ol with various heterocyclic and aromatic aldehydes. Previous studies have shown that SCR7, synthesized from 5,6-diamino-2-mercaptopyrimidin-4-ol, induced cytotoxicity by targeting cancer cells by primarily inhibiting DNA Ligase IV involved in nonhomologous end joining, one of the major DNA double-strand break repair pathways. Inhibition of DNA repair pathways is considered as an important strategy for cancer therapy. Due to limitations of SCR7 in terms of IC50 in cancer cells, here we have designed, synthesized, and characterized potent derivatives of SCR7 using 5,6-diamino-2-mercaptopyrimidin-4-ol as the starting material. Several synthesized imine compounds exhibited significant improvement in inhibition of end joining and cytotoxicity up to 27-fold lower concentrations than SCR7. Among these, two compounds, SCR116 and SCR132, showed increased cancer cell death in a Ligase IV-dependent manner. Treatment with the compounds also led to reduction in V(D)J recombination efficiency, cell cycle arrest at G2/M phase, accumulation of double-strand breaks inside cells, and improved anti-cancer potential when combined with γ-radiation and radiomimetic drugs. Thus, we describe novel inhibitors of NHEJ with higher efficacy and potential, which can be developed as cancer therapeutics.


Subject(s)
DNA End-Joining Repair , Neoplasms , Humans , Neoplasms/genetics , DNA Repair , DNA Breaks, Double-Stranded , DNA/metabolism
5.
J Org Chem ; 86(8): 5530-5537, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33826333

ABSTRACT

Carbonylation of (hetero)aryl iodides/bromides with highly deactivated 2-aminopyridines using Pd-Co(CO)4 bimetallic catalysis is accomplished. The use of Co2(CO)8 as a solid CO(g) source enhanced reaction rates observed when compared to CO(g), and excellent yields highlight the versatility of the developed protocol. A wide range of electronically and sterically demanding heterocyclic amines and (hetero)aryl iodides/bromides employed for this study resulted in excellent yields of amino carbonylated products. The developed methodology was further extended to synthesize Trypanosome brucie and luciferase inhibitors.


Subject(s)
Amines , Palladium , Bromides , Catalysis , Iodides
6.
Sci Rep ; 10(1): 15188, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32938954

ABSTRACT

Small molecule inhibitors targeting BCL2 are explored as anticancer therapeutics. Previously, we have reported identification and characterization of a novel BCL2 inhibitor, Disarib. Disarib induced cancer cell death in a BCL2 dependent manner in different cancer cell lines and mouse tumor models when it was administered intraperitoneally. In the present study, using two syngeneic mouse models, breast adenocarcinoma (EAC) and Dalton's lymphoma (DLA), we show that oral administration of Disarib resulted in significant tumor regression in a concentration dependent manner. Importantly, tumor developed in both female and male mice were equally sensitive to Disarib. Further, we have investigated the toxicity of Disarib in normal cells. Single dose toxicity analysis of Disarib in male and female mice after oral administration revealed no significant variations compared to control group for parameters such as body weight, food and water consumption and behavioural changes which were analysed for the entire period of study. Haematological and histopathological analyses also did not show any significant difference from the control groups. Thus, our results reveal safe use of Disarib as a small molecule inhibitor and provide the foundation for investigation of other preclinical studies.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Drug-Related Side Effects and Adverse Reactions/diagnosis , Indoles/therapeutic use , Lymphoma/drug therapy , Mammary Glands, Human/drug effects , Mammary Neoplasms, Experimental/drug therapy , Thiadiazoles/therapeutic use , Administration, Oral , Animals , Blood Cell Count , Body Weight/drug effects , Cell Line, Tumor , Female , Hematopoiesis/drug effects , Humans , Indoles/pharmacology , Male , Mammary Glands, Human/pathology , Mammary Neoplasms, Experimental/diagnosis , Mice , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Thiadiazoles/pharmacology
7.
Mol Carcinog ; 59(6): 618-628, 2020 06.
Article in English | MEDLINE | ID: mdl-32189406

ABSTRACT

Targeting DNA repair with small-molecule inhibitors is an attractive strategy for cancer therapy. Majority of DNA double-strand breaks in mammalian cells are repaired through nonhomologous end-joining (NHEJ). It has been shown that small-molecule inhibitors of NHEJ can block efficient repair inside cancer cells, leading to cell death. Previously, we have reported that SCR7, an inhibitor of NHEJ can induce tumor regression in mice. Later studies have shown that different forms of SCR7 can inhibit DNA end-joining in Ligase IV-dependent manner. Recently, we have derivatized SCR7 by introducing spiro ring into core structure. Here, we report the identification of a novel inhibitor of NHEJ, named SCR130 with 20-fold higher efficacy in inducing cytotoxicity in cancer cell lines. SCR130 inhibited DNA end-joining catalyzed by rat tissue extract. Specificity analysis revealed that while SCR130 was specific to Ligase IV, it showed minimal or no effect on Ligase III and Ligase I mediated joining. Importantly, SCR130 exhibited the least cytotoxicity in Ligase IV-null cell line as compared with wild type, confirming Ligase IV-specificity. Furthermore, we demonstrate that SCR130 can potentiate the effect of radiation in cancer cells when used in combination with γ-radiation. Various cellular assays in conjunction with Western blot analysis revealed that treatment with SCR130 led to loss of mitochondrial membrane potential leading to cell death by activating both intrinsic and extrinsic pathways of apoptosis. Thus, we describe a novel inhibitor of NHEJ with higher efficacy and may have the potential to be developed as cancer therapeutic.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death , DNA Breaks, Double-Stranded/drug effects , DNA End-Joining Repair/drug effects , DNA Ligase ATP/antagonists & inhibitors , Pyrimidines/pharmacology , Schiff Bases/pharmacology , Small Molecule Libraries/pharmacology , Animals , HeLa Cells , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Rats
8.
Org Biomol Chem ; 18(14): 2678-2684, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32202293

ABSTRACT

The present work describes an unexpected and unique protocol for the iodine catalysed synthesis of o-ureidobenzonitriles using o-aminobenzamides and isothiocyanates via intramolecular rearrangement. The metal-free route achieved here is insensitive to moisture and applicable to the synthesis of a wide variety of o-ureidobenzonitriles with excellent yields even in a scalable fashion.

9.
Acta Crystallogr E Crystallogr Commun ; 76(Pt 1): 18-24, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31921446

ABSTRACT

Three title compounds, namely, 2-(4-chloro-benz-yl)-5-[(1H-indol-3-yl)meth-yl]-6-phenyl-imidazo[2,1-b][1,3,4]thia-diazole, C26H19ClN4S, (I), 2-(4-chloro-benz-yl)-6-(4-fluoro-phen-yl)-5-[(1H-indol-3-yl)meth-yl]imidazo[2,1-b][1,3,4]thia-diazole, C26H18ClFN4S, (II), and 6-(4-bromo-phen-yl)-2-(4-chloro-benz-yl)-5-[(1H-indol-3-yl)meth-yl]imidazo[2,1-b][1,3,4]thia-diazole, C26H18BrClN4S, (III), have been prepared using a reductive condensation of indole with the corresponding 6-aryl-2-(4-chloro-benz-yl)imidazo[2,1-b][1,3,4]thia-diazole-5-carbaldehydes (aryl = phenyl, 4-fluoro-phenyl or 4-bromo-phen-yl), and their crystal structures have been determined. The asymmetric unit of compound (I) consists of two independent mol-ecules and one of the mol-ecules exhibits disorder of the 4-chloro-benzyl substituent with occupancies 0.6289 (17) and 0.3711 (17). Each type of mol-ecule forms a C(8) chain motif built from N-H⋯N hydrogen bonds, which for the fully ordered mol-ecule is reinforced by C-H⋯π inter-actions. In compound (II), the chloro-benzyl unit is again disordered, with occupancies 0.822 (6) and 0.178 (6), and the mol-ecules form C(8) chains similar to those in (I), reinforced by C-H⋯π inter-actions involving only the major disorder component. The chloro-benzyl unit in compound (III) is also disordered with occupancies of 0.839 (5) and 0.161 (5). The mol-ecules are linked by a combination of one N-H⋯N hydrogen bond and four C-H⋯π inter-actions, forming a three-dimensional framework.

10.
FEBS J ; 285(21): 3959-3976, 2018 11.
Article in English | MEDLINE | ID: mdl-30230716

ABSTRACT

Nonhomologous DNA end joining (NHEJ) is the major DNA double-strand break (DSB) repair pathway in mammals. Previously, we have described a small molecule inhibitor, SCR7, which can inhibit NHEJ in a Ligase IV-dependent manner. Administration of SCR7 within the cells resulted in the accumulation of DNA breaks, cell death, and inhibition of tumor growth in mice. In the present study, we report that parental SCR7, which is unstable, can be autocyclized into a stable form. Both parental SCR7 and cyclized SCR7 possess the same molecular weight (334.09) and molecular formula (C18 H14 N4 OS), whereas its oxidized form, SCR7-pyrazine, possesses a different molecular formula (C18 H12 N4 OS), molecular weight (332.07), and structure. While cyclized form of SCR7 showed robust inhibition of NHEJ in vitro, both forms exhibited efficient cytotoxicity. Cyclized and oxidized forms of SCR7 inhibited DNA end joining catalyzed by Ligase IV, whereas their impact was minimal on Ligase III, Ligase I, and T4 DNA Ligase-mediated joining. Importantly, both forms inhibited V(D)J recombination, although the effect was more pronounced for SCR7-cyclized. Both forms blocked NHEJ in a Ligase IV-dependent manner leading to the accumulation of DSBs within the cells. Although cytotoxicity due to SCR7-cyclized was Ligase IV specific, the pyrazine form exhibited nonspecific cytotoxicity at higher concentrations in Ligase IV-null cells. Finally, we demonstrate that both forms can potentiate the effect of radiation. Thus, we report that cyclized and oxidized forms of SCR7 can inhibit NHEJ in a Ligase IV-dependent manner, although SCR7-pyrazine is less specific to Ligase IV inside the cell.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA End-Joining Repair/drug effects , DNA Ligase ATP/chemistry , DNA Ligase ATP/metabolism , Neoplasms/pathology , Pyrimidines/pharmacology , Schiff Bases/pharmacology , Cell Death/drug effects , HeLa Cells , Humans , MCF-7 Cells , Neoplasms/drug therapy , Neoplasms/genetics , Oxidation-Reduction , V(D)J Recombination
11.
Target Oncol ; 11(5): 655-665, 2016 10.
Article in English | MEDLINE | ID: mdl-27188390

ABSTRACT

BACKGROUND: Cancer is a multifactorial disease, which makes it difficult to cure. Since more than one defective cellular component is often involved during oncogenesis, combination therapy is gaining prominence in the field of cancer therapeutics. OBJECTIVE: The purpose of this study was to investigate the combinatorial effects of a novel PARP inhibitor, P10, and HDAC inhibitor, SAHA, in leukemic cells. METHODS: Combinatorial effects of P10 and SAHA were tested using propidium iodide staining in different leukemic cells. Further, flowcytometry-based assays such as calcein-AM/ethidium homodimer staining, annexin-FITC/PI staining, and JC-1 staining were carried out to elucidate the mechanism of cell death. In addition, cell-cycle analysis, immunocytochemistry studies, and western blotting analysis were conducted to check the combinatorial effect in Nalm6 cells. RESULTS: Propidium iodide staining showed that P10 in combination with SAHA induced cell death in Nalm6 cells, in which PARP expression and activity is high with a combination index of <0.2. Annexin-FITC/PI staining, JC-1 staining, and other biochemical assays revealed that P10 in combination with SAHA induced apoptosis by causing a change in mitochondrial membrane potential in >65 % cells. Importantly, combinatorial treatment induced S phase arrest in 40-45 % cells due to DNA damage and plausible replicative stress. Finally, we demonstrated that treatment with P10 led to DNA strand breaks, which were further potentiated by SAHA (p < 0.01), leading to activation of apoptosis and increased cell death in PARP-positive leukemic cells. CONCLUSIONS: Our study reveals that coadministration of PARP inhibitor with SAHA could be used as a combination therapy against leukemic cells that possess high levels of intrinsic PARP activity.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Leukemia/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Cell Line, Tumor , Cell Proliferation , Histone Deacetylase Inhibitors/pharmacology , Humans , Leukemia/pathology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
12.
Org Lett ; 16(22): 5910-3, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25413125

ABSTRACT

Azomethine ylides are accessed under mild conditions via benzoic acid catalyzed condensations of 1,2,3,4-tetrahydroisoquinolines or tryptolines with aldehydes bearing a pendent dipolarophile. These intermediates undergo intramolecular [3 + 2]-cycloadditions in a highly diastereoselective fashion to form polycyclic amines with four new stereogenic centers. Challenging substrates such as piperidine, morpholine, and thiomorpholine undergo the corresponding reactions at elevated temperatures.


Subject(s)
Amines/chemistry , Azo Compounds/chemical synthesis , Benzoic Acid/chemistry , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Tetrahydroisoquinolines/chemistry , Thiosemicarbazones/chemical synthesis , Aldehydes/chemistry , Amines/chemical synthesis , Azo Compounds/chemistry , Catalysis , Cyclization , Cycloaddition Reaction , Heterocyclic Compounds, 4 or More Rings/chemistry , Molecular Structure , Oxidation-Reduction , Stereoisomerism , Thiosemicarbazones/chemistry
13.
BMC Cancer ; 13: 37, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23356739

ABSTRACT

BACKGROUND: Post-translational modifications (PTMs) of histones and other proteins are perturbed in tumours. For example, reduced levels of acetylated H4K16 and trimethylated H4K20 are associated with high tumour grade and poor survival in breast cancer. Drug-like molecules that can reprogram selected histone PTMs in tumour cells are therefore of interest as potential cancer chemopreventive agents. In this study we assessed the effects of the phytocompounds garcinol and curcumin on histone and p53 modification in cancer cells, focussing on the breast tumour cell line MCF7. METHODS: Cell viability/proliferation assays, cell cycle analysis by flow cytometry, immunodetection of specific histone and p53 acetylation marks, western blotting, siRNA and RT-qPCR. RESULTS: Although treatment with curcumin, garcinol or the garcinol derivative LTK-14 hampered MCF7 cell proliferation, differential effects of these compounds on histone modifications were observed. Garcinol treatment resulted in a strong reduction in H3K18 acetylation, which is required for S phase progression. Similar effects of garcinol on H3K18 acetylation were observed in the osteosarcoma cells lines U2OS and SaOS2. In contrast, global levels of acetylated H4K16 and trimethylated H4K20 in MCF7 cells were elevated after garcinol treatment. This was accompanied by upregulation of DNA damage signalling markers such as γH2A.X, H3K56Ac, p53 and TIP60. In contrast, exposure of MCF7 cells to curcumin resulted in increased global levels of acetylated H3K18 and H4K16, and was less effective in inducing DNA damage markers. In addition to its effects on histone modifications, garcinol was found to block CBP/p300-mediated acetylation of the C-terminal activation domain of p53, but resulted in enhanced acetylation of p53K120, and accumulation of p53 in the cytoplasmic compartment. Finally, we show that the elevation of H4K20Me3 levels by garcinol correlated with increased expression of SUV420H2, and was prevented by siRNA targeting of SUV420H2. CONCLUSION: In summary, although garcinol and curcumin can both inhibit histone acetyltransferase activities, our results show that these compounds have differential effects on cancer cells in culture. Garcinol treatment alters expression of chromatin modifying enzymes in MCF7 cells, resulting in reprogramming of key histone and p53 PTMs and growth arrest, underscoring its potential as a cancer chemopreventive agent.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Curcumin/pharmacology , Histones/metabolism , Protein Processing, Post-Translational/drug effects , Terpenes/pharmacology , Tumor Suppressor Protein p53/metabolism , Acetylation , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CREB-Binding Protein/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Flow Cytometry , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunohistochemistry , Lysine Acetyltransferase 5 , MCF-7 Cells , Methylation , Polymerase Chain Reaction , RNA Interference , Time Factors , Transfection
14.
Chem Biol ; 17(8): 903-13, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20797619

ABSTRACT

Altered histone acetylation is associated with several diseases, including cancer. We report here that, unlike in most cancers, histones are found to be highly hyperacetylated in oral squamous cell carcinoma (OSCC; oral cancer) patient samples. Mechanistically, overexpression, as well as enhanced autoacetylation, of p300 induced by nucleophosmin (NPM1) and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) causes the hyperacetylation, which is nitric oxide (NO) signal dependent. Inhibition of the histone acetyltransferase (HAT) activity of p300 by a water-soluble, small molecule inhibitor, Hydrazinocurcumin (CTK7A), substantially reduced the xenografted oral tumor growth in mice. These results, therefore, not only establish an epigenetic target for oral cancer, but also implicate a HAT inhibitor (HATi) as a potential therapeutic molecule.


Subject(s)
Curcumin/analogs & derivatives , Histone Acetyltransferases/antagonists & inhibitors , Histones/metabolism , Hydrazines/chemistry , Hydrazines/pharmacology , Mouth Neoplasms/metabolism , Nitric Oxide/metabolism , Water/chemistry , Acetylation/drug effects , Animals , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Curcumin/chemistry , Curcumin/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , HeLa Cells , Histone Acetyltransferases/metabolism , Humans , Mice , Mice, Nude , Mouth Neoplasms/enzymology , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Nuclear Proteins/metabolism , Nucleophosmin , Solubility , Up-Regulation/drug effects , p300-CBP Transcription Factors/metabolism
15.
J Biol Chem ; 285(10): 7143-52, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20022955

ABSTRACT

Methylation of the arginine residues of histones by methyltransferases has important consequences for chromatin structure and gene regulation; however, the molecular mechanism(s) of methyltransferase regulation is still unclear, as is the biological significance of methylation at particular arginine residues. Here, we report a novel specific inhibitor of coactivator-associated arginine methyltransferase 1 (CARM1; also known as PRMT4) that selectively inhibits methylation at arginine 17 of histone H3 (H3R17). Remarkably, this plant-derived inhibitor, called TBBD (ellagic acid), binds to the substrate (histone) preferentially at the signature motif, "KAPRK," where the proline residue (Pro-16) plays a critical role for interaction and subsequent enzyme inhibition. In a promoter-specific context, inhibition of H3R17 methylation represses expression of p21, a p53-responsive gene, thus implicating a possible role for H3 Arg-17 methylation in tumor suppressor function. These data establish TBBD as a novel specific inhibitor of arginine methylation and demonstrate substrate sequence-directed inhibition of enzyme activity by a small molecule and its physiological consequence.


Subject(s)
Arginine/metabolism , Ellagic Acid/metabolism , Histones/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/metabolism , Amino Acid Sequence , Animals , Cell Line , Ellagic Acid/chemistry , Gene Expression Regulation , Histones/chemistry , Histones/genetics , Humans , Lythraceae/chemistry , Methylation , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutagenesis, Site-Directed , Proline/metabolism , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/genetics , Thermodynamics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...