Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Anticancer Drugs ; 33(1): e235-e246, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34419964

ABSTRACT

One cost-effective way for identifying novel cancer therapeutics is in the repositioning of available drugs for which current therapies are inadequate. Levofloxacin prevents DNA duplication in bacteria by inhibiting the activity of DNA helicase. As eukaryotic cells have similar intracellular biologic characteristics as prokaryotic cells, we speculate that antibiotics inhibiting DNA duplication in bacteria may also affect the survival of cancer cells. Here we report that levofloxacin significantly inhibited the proliferation and clone formation of cancer cells and xenograft tumor growth through cell cycle arrest at G2/M and by enhancing apoptosis. Levofloxacin significantly altered gene expression in a direction favoring anticancer activity. THBS1 and LAPTM5 were dose-dependently upregulated whereas SRD5A3, MFAP5 and P4HA1 were downregulated. Pathway analysis revealed that levofloxacin significantly regulated canonical oncogenic pathways. Specific network enrichment included a MAPK/apoptosis/cytokine-cytokine receptor interaction pathway network that associates with cell growth, differentiation, cell death, angiogenesis and development and repair processes and a bladder cancer/P53 signaling pathway network mediating the inhibition of angiogenesis and metastasis. THBS1 overlapped in 16 of the 22 enriched apoptotic pathways and the 2 pathways in the bladder cancer/P53 signaling pathway network. P4HA1 enriched in 7 of the top 10 molecular functions regulated by differential downregulated genes. Our results indicate that levofloxacin has broad-spectrum anticancer activity with the potential to benefit cancer patients already treated or requiring prophylaxis for an infectious syndrome. The efficacy we find with levofloxacin may provide insight into the discovery and the design of novel less toxic anticancer drugs.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Levofloxacin/pharmacology , Animals , Apoptosis/drug effects , Cell Adhesion Molecules/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/drug effects , DNA Helicases/drug effects , Dose-Response Relationship, Drug , Humans , Male , Membrane Proteins/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinases/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
2.
Mol Biol Cell ; 32(21): ar18, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34432484

ABSTRACT

Stress granules (SGs) are ribonucleoprotein functional condensates that form under stress conditions in all eukaryotic cells. Although their stress-survival function is far from clear, SGs have been implicated in the regulation of many vital cellular pathways. Consequently, SG dysfunction is thought to be a mechanistic point of origin for many neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). Additionally, SGs are thought to play a role in pathogenic pathways as diverse as viral infection and chemotherapy resistance. There is a growing consensus on the hypothesis that understanding the mechanistic regulation of SG physical properties is essential to understanding their function. Although the internal dynamics and condensation mechanisms of SGs have been broadly investigated, there have been fewer investigations into the timing of SG formation and clearance in live cells. Because the lifetime of SG persistence can be a key factor in their function and tendency toward pathological dysregulation, SG clearance mechanisms deserve particular attention. Here we show that resveratrol and its analogues piceatannol, pterostilbene, and 3,4,5,4'-tetramethoxystilbene induce G3BP-dependent SG formation with atypically rapid clearance kinetics. Resveratrol binds to G3BP, thereby reducing its protein-protein association valency. We suggest that altering G3BP valency is a pathway for the formation of uniquely transient SGs.


Subject(s)
DNA Helicases/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Resveratrol/pharmacology , Stress Granules/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , DNA Helicases/drug effects , HEK293 Cells , HeLa Cells , Humans , Kinetics , Poly-ADP-Ribose Binding Proteins/drug effects , RNA Helicases/drug effects , RNA Recognition Motif Proteins/drug effects , Ribonucleoproteins/metabolism , Stress Granules/drug effects
3.
SAR QSAR Environ Res ; 32(1): 51-70, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33401979

ABSTRACT

A Förster resonance energy transfer (FRET)-based assay was used to screen the FDA-approved compound library against the MERS-CoV helicase, an essential enzyme for virus replication within the host cell. Five compounds inhibited the helicase activity with submicromolar potencies (IC50, 0.73-1.65 µM) and ten compounds inhibited the enzyme with micromolar potencies (IC50, 19.6-502 µM). The molecular operating environment (MOE) was used to dock the identified inhibitors on the MERS-CoV helicase nucleotide binding. Strong inhibitors docked well in the nucleotide-binding site and established interactions with some of the essential residues. There was a reasonable correlation between the observed IC50 values and the MOE docking scores of the strong inhibitors (r 2 = 0.74), indicating the ability of the in silico docking model to predict the binding of strong inhibitors. In silico docking could be a useful complementary tool used with the FRET-based assay to predict new MERS-CoV helicase inhibitors. The identified inhibitors could potentially be used in the clinical development of new antiviral treatment for MERS-CoV and other coronavirus related diseases, including coronavirus disease 2019 (COVID-19).


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Coronavirus Infections/drug therapy , DNA Helicases/drug effects , Enzyme Inhibitors/pharmacokinetics , Middle East Respiratory Syndrome Coronavirus/drug effects , Humans , Quantitative Structure-Activity Relationship , SARS-CoV-2/drug effects , Virus Replication/drug effects
4.
Article in English | MEDLINE | ID: mdl-31382687

ABSTRACT

Maternal exposure to polychlorinated biphenyls (PCBs) results in abnormal fetal development, possibly because of epigenetic alterations. However, the association between PCB levels in cord serum with fetal DNA methylation status in cord tissue is unclear. This study aims to identify alterations in DNA methylation in cord tissue potentially associated with PCB levels in cord serum from a birth cohort in Chiba, Japan (male neonates = 32, female neonates = 43). Methylation array analysis identified five sites for female neonates (cg09878117, cg06154002, cg06289566, cg12838902, cg01083397) and one site for male neonates (cg13368805) that demonstrated a change in the methylation degree. This result was validated by pyrosequencing analysis, showing that cg06154002 (tudor domain containing 9: TDRD9) in cord tissue from female neonates is significantly correlated with total PCB levels in cord serum. These results indicate that exposure to PCBs may alter TDRD9 methylation levels, although this hypothesis requires further validation using data obtained from female neonates. However, since the present cohort is small, further studies with larger cohorts are required to obtain more data on the effects of PCB exposure and to identify corresponding biomarkers.


Subject(s)
DNA Methylation/drug effects , Fetal Blood/metabolism , Maternal Exposure , Polychlorinated Biphenyls/blood , Umbilical Cord/metabolism , Biomarkers , Cohort Studies , DNA Helicases/drug effects , Female , Fetal Development/drug effects , Humans , Infant, Newborn , Japan , Male
5.
Biomed Res Int ; 2018: 5753804, 2018.
Article in English | MEDLINE | ID: mdl-30246023

ABSTRACT

The hepatitis E virus- (HEV-) helicase as a novel drug-target was evaluated. While cell culture model was used for mutational characterization of helicase, in silico protein modeling and virtual screening were employed to identify helicase inhibitors. None of the saturation mutant replicons significantly affected RNA replication. Notably, mutants encompassing the Walker motifs replicated as wild-type, showing indispensability of nucleotides conservation in viability compared to known criticality of amino acids. A 3D modeling of HEV-helicase and screening of a compound dataset identified ten most promising inhibitors with drug likeness, notably, JFD02650, RDR03130, and HTS11136 that interacted with Walker A residues Gly975, Gly978, Ser979, and Gly980. Our model building and virtual identification of novel helicase inhibitors warrant further studies towards developing anti-HEV drugs.


Subject(s)
DNA Helicases , Hepatitis E virus/enzymology , Mutation , DNA Helicases/drug effects , DNA Helicases/genetics , Hepatitis E , Virus Replication
6.
Mol Cell Neurosci ; 87: 77-85, 2018 03.
Article in English | MEDLINE | ID: mdl-29249293

ABSTRACT

We are beginning to appreciate the complex mechanisms by which epigenetic proteins control chromatin dynamics to tightly regulate normal development. However, the interaction between these proteins, particularly in the context of neuronal function, remains poorly understood. Here, we demonstrate that the activity of histone deacetylases (HDACs) opposes that of a chromatin remodeling enzyme at the Drosophila neuromuscular junction (NMJ). Pharmacological inhibition of HDAC function reverses loss of function phenotypes associated with Kismet, a chromodomain helicase DNA-binding (CHD) protein. Inhibition of HDACs suppresses motor deficits, overgrowth of the NMJ, and defective neurotransmission associated with loss of Kismet. We hypothesize that Kismet and HDACs may converge on a similar set of target genes in the nervous system. Our results provide further understanding into the complex interactions between epigenetic protein function in vivo.


Subject(s)
DNA Helicases/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Neuromuscular Junction/drug effects , Synaptic Transmission/drug effects , Animals , Chromatin , DNA Helicases/genetics , Histone Deacetylases/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Neuromuscular Junction/genetics , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/genetics
7.
J Microbiol Biotechnol ; 27(11): 2070-2073, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-28910865

ABSTRACT

We have discovered a novel chemical compound, (E)-3-(furan-2-yl)-N-(4-sulfamoylphenyl) acrylamide, that suppresses the enzymatic activities of SARS coronavirus helicase. To determine the inhibitory effect, ATP hydrolysis and double-stranded DNA unwinding assays were performed in the presence of various concentrations of the compound. Through these assays, we obtained IC50 values of 2.09 ± 0.30 µM (ATP hydrolysis) and 13.2 ± 0.9 µM (DNA unwinding), respectively. Moreover, we found that the compound did not have any significant cytotoxicity when 40 µM of it was used. Our results showed that the compound might be useful to be developed as an inhibitor against SARS coronavirus.


Subject(s)
Antiviral Agents/antagonists & inhibitors , DNA Helicases/drug effects , Enzyme Inhibitors/pharmacology , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe acute respiratory syndrome-related coronavirus/enzymology , Adenosine Triphosphate , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Cell Line/drug effects , Cell Survival/drug effects , DNA/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Humans , Hydrolysis , Inhibitory Concentration 50
8.
J Med Chem ; 59(1): 238-52, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26649766

ABSTRACT

Novel topoisomerase II (Topo II) inhibitors have gained considerable interest for the development of anticancer agents. In this study, a series of 1,3-benzoazolyl-substituted pyrrolo[2,3-b]pyrazine derivatives were designed, synthesized, and evaluated as potential Topo II catalytic inhibitors. It was found that some of derivatives had good antiproliferative activity on seven cancer cell lines, especially on HL-60/MX2, a cancer cell line derivative from HL-60 that is resistant to Topo II poison. Topo II mediated DNA relaxation assay results showed that derivatives could significantly inhibit the activity of Topo II, and the structure-activity relationship studies indicated the importance of the alkylamino side chain and the benzoazolyl group. Further mechanism studies revealed that derivatives function as Topo II nonintercalative catalytic inhibitors and may block the ATP binding site of Topo II. Moreover, flow cytometric analysis showed that this class of compounds could induce apoptosis of HL-60 cells.


Subject(s)
Pyrazines/chemical synthesis , Pyrazines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Catalysis , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/drug effects , DNA Helicases/drug effects , DNA Topoisomerases, Type II/chemistry , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Docking Simulation , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/metabolism , Structure-Activity Relationship
9.
Hepatology ; 60(4): 1241-50, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24824848

ABSTRACT

UNLABELLED: The far upstream element binding protein (FBP) and the FBP-interacting repressor (FIR) represent molecular tools for transcriptional fine tuning of target genes. Strong overexpression of FBP in human hepatocellular carcinoma (HCC) supports tumor growth and correlates with poor patient prognosis. However, the role of the transcriptional repressor FIR in hepatocarcinogenesis remains poorly delineated. We show that overexpression of FIR correlates with tumor dedifferentiation and tumor cell proliferation in about 60% of primary HCCs. Elevated FIR levels are associated with genomic gains of the FIR gene locus at chromosome 8q24.3 in human HCC specimens. In vitro, nuclear enrichment of FIR supports HCC cell proliferation and migration. Expression profiling of HCC cells after small interfering RNA (siRNA)-mediated silencing of FIR identified the transcription factor DP-1 (TFDP1) as a transcriptional target of FIR. Surprisingly, FIR stimulates the expression of FBP in a TFDP1/E2F1-dependent manner. FIR splice variants lacking or containing exon 2 and/or exon 5 are expressed in the majority of HCCs but not in normal hepatocytes. Specific inhibition of FIR isoforms with and without exon 2 revealed that both groups of FIR splice variants facilitate tumor-supporting effects. This finding was confirmed in xenograft transplantation experiments with lentiviral-infected short hairpin RNA (shRNA) targeting all FIR variants as well as FIR with and without exon 2. CONCLUSION: High-level nuclear FIR does not facilitate repressor properties but supports tumor growth in HCC cells. Thus, the pharmacological inhibition of FIR might represent a promising therapeutic strategy for HCC patients with elevated FIR expression.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation/physiology , DNA Helicases/physiology , DNA-Binding Proteins/physiology , Liver Neoplasms/physiopathology , RNA-Binding Proteins/physiology , Repressor Proteins/physiology , Animals , Carcinoma, Hepatocellular/pathology , DNA Helicases/drug effects , DNA Helicases/genetics , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Exons/genetics , Humans , In Vitro Techniques , Liver Neoplasms/pathology , Mice, SCID , Mice, Transgenic , Protein Isoforms/genetics , RNA Splicing Factors , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/genetics , Repressor Proteins/drug effects , Repressor Proteins/genetics , Transcription Factor DP1/physiology , Transplantation, Heterologous
10.
J Hepatol ; 52(5): 681-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20346530

ABSTRACT

BACKGROUND & AIMS: We previously showed that Reptin is overexpressed in hepatocellular carcinoma (HCC), and that in vitro depletion of Reptin with siRNAs led to HCC cell growth arrest and apoptosis. Here, we asked whether in vivo targeting of Reptin in established tumours had a therapeutic effect. METHODS: We used lentiviral vectors to construct HuH7 and Hep3B cell lines with doxycycline (Dox)-dependent expression of Reptin (R2) or control shRNA (GL2). Cells were injected subcutaneously into immunodeficient mice, and Dox was given when tumours reached a volume of 250 mm(3). RESULTS: In vitro, the growth of GL2-Dox, GL2+Dox, and R2-Dox cells was undistinguishable whereas that of R2+Dox cells stopped 4 days after Dox treatment. The growth decrease was associated with increased apoptosis, and evidence of replicative senescence, as shown by staining for acid beta-galactosidase and the presence of senescence-associated heterochromatin foci. In xenografted mice, R2+Dox tumour growth stagnated or even regressed with prolonged treatment in contrast with the GL2-Dox, GL2+Dox, and R2-Dox tumours that progressed steadily. The blockage of tumour progression was associated with the induction of senescence and reduced cell proliferation. CONCLUSIONS: In vivo Reptin depletion leads to tumour growth arrest. Reptin may prove a valuable target in HCC.


Subject(s)
Carrier Proteins/genetics , DNA Helicases/genetics , Gene Silencing , Liver Neoplasms/prevention & control , ATPases Associated with Diverse Cellular Activities , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Carrier Proteins/drug effects , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor , Cellular Senescence/drug effects , DNA Helicases/drug effects , DNA Primers , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter/drug effects , Humans , Liver Neoplasms/pathology , Luciferases/genetics , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
11.
PLoS Pathog ; 5(11): e1000663, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19936046

ABSTRACT

The Vibrionaceae is comprised of numerous aquatic species and includes several human pathogens, such as Vibrio cholerae, the cause of cholera. All organisms in this family have two chromosomes, and replication of the smaller one depends on rctB, a gene that is restricted to the Vibrionaceae. Given the increasing prevalence of multi-drug resistance in pathogenic vibrios, there is a need for new targets and drugs to combat these pathogens. Here, we carried out a high throughput cell-based screen to find small molecule inhibitors of RctB. We identified a compound that blocked growth of an E. coli strain bearing an rctB-dependent plasmid but did not influence growth of E. coli lacking this plasmid. This compound, designated vibrepin, had potent cidal activity against V. cholerae and inhibited the growth of all vibrio species tested. Vibrepin blocked RctB oriCII unwinding, apparently by promoting formation of large non-functional RctB complexes. Although vibrepin also appears to have targets other than RctB, our findings suggest that RctB is an attractive target for generation of novel antibiotics that only block growth of vibrios. Vibrio-specific agents, unlike antibiotics currently used in clinical practice, will not engender resistance in the normal human flora or in non-vibrio environmental microorganisms.


Subject(s)
Anti-Infective Agents/pharmacology , Bacterial Proteins/drug effects , Chromosomes, Bacterial/genetics , DNA Helicases/drug effects , Trans-Activators/drug effects , Vibrionaceae/genetics , Drug Resistance, Bacterial/drug effects
12.
DNA Repair (Amst) ; 8(3): 354-9, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19100865

ABSTRACT

5-Azacytidine induces CG-to-GC transversion mutations in Escherichia coli. The results presented in this paper provide evidence that repair of the drug-induced lesions that produce these mutations involves components of both the mismatch repair and nucleotide excision repair systems. Strains deficient in mutL, mutS, uvrA, uvrB or uvrC all showed an increase in mutation in response to 5-azacytidine. Using a bacterial two-hybrid assay, we showed that UvrB interacts with MutL and MutS in a drug-dependent manner, while UvrC interacts with MutL independent of drug. We suggest that 5-azacytidine-induced mismatches recruit MutS and MutL, but are poorly processed by mismatch repair. Instead, the stalled MutS-MutL complex recruits the Uvr proteins to complete repair.


Subject(s)
Azacitidine/administration & dosage , Base Pair Mismatch/drug effects , DNA Mismatch Repair/physiology , DNA Repair/physiology , DNA, Bacterial/drug effects , DNA, Bacterial/metabolism , Escherichia coli/metabolism , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/drug effects , DNA Helicases/deficiency , DNA Helicases/drug effects , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/drug effects , DNA, Bacterial/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/drug effects , DNA-Cytosine Methylases/metabolism , Dose-Response Relationship, Drug , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/drug effects , Enzyme Inhibitors/administration & dosage , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/drug effects , Frameshift Mutation/drug effects , MutL Proteins , MutS DNA Mismatch-Binding Protein/deficiency , MutS DNA Mismatch-Binding Protein/drug effects , Two-Hybrid System Techniques
13.
Nucleic Acids Res ; 36(7): 2249-56, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18285361

ABSTRACT

The quest for inducers and inhibitors of protein amyloidogenesis is of utmost interest, since they are key tools to understand the molecular bases of proteinopathies such as Alzheimer, Parkinson, Huntington and Creutzfeldt-Jakob diseases. It is also expected that such molecules could lead to valid therapeutic agents. In common with the mammalian prion protein (PrP), the N-terminal Winged-Helix (WH1) domain of the pPS10 plasmid replication protein (RepA) assembles in vitro into a variety of amyloid nanostructures upon binding to different specific dsDNA sequences. Here we show that di- (S2) and tetra-sulphonated (S4) derivatives of indigo stain dock at the DNA recognition interface in the RepA-WH1 dimer. They compete binding of RepA to its natural target dsDNA repeats, found at the repA operator and at the origin of replication of the plasmid. Calorimetry points to the existence of a major site, with micromolar affinity, for S4-indigo in RepA-WH1 dimers. As revealed by electron microscopy, in the presence of inducer dsDNA, both S2/S4 stains inhibit the assembly of RepA-WH1 into fibres. These results validate the concept that DNA can promote protein assembly into amyloids and reveal that the binding sites of effector molecules can be targeted to inhibit amyloidogenesis.


Subject(s)
Amyloid/chemistry , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , DNA/antagonists & inhibitors , Indigo Carmine/chemistry , Indigo Carmine/pharmacology , Indoles/chemistry , Indoles/pharmacology , Trans-Activators/chemistry , Amyloid/ultrastructure , Binding Sites , Binding, Competitive , DNA/chemistry , DNA/metabolism , DNA Helicases/drug effects , DNA Helicases/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Models, Molecular , Protein Structure, Tertiary , Thermodynamics , Trans-Activators/drug effects , Trans-Activators/metabolism
14.
Glycoconj J ; 23(3-4): 175-87, 2006 May.
Article in English | MEDLINE | ID: mdl-16691501

ABSTRACT

Apoptosis of human breast carcinoma cells (SKBR-3, MCF-7, and MDA-468) has been observed after treatment of these cells with anti-cancer drug cis-platin and glycosphingolipid biosynthesis inhibitor L- and D-PPMP, respectively. These drugs initiated apoptosis in a dose-dependent manner as measured by phenotypic morphological changes, by binding of a fluorescent phophatidyl serine-specific dye (PSS-380) onto the outer leaflet of the cell membranes, and by activation of caspases, -3, -8, and -9. It was observed that in two hours very little apoptotic process had started but predominant biochemical changes occurred after 6 h. DNA degradation started after 24 hours of drug treatment. However, very little is known about the stability of the ';Replication Complexes'' during the apoptotic process. DNA helicases are motor proteins that catalyze the melting of genomic DNA during its replication, repair, and recombination processes. Previously, DNA helicase-III was characterized as a component of the replication complexes isolated from embryonic chicken brains as well as breast and colon carcinoma cells. Helicase activities were measured by a novel method (ROME assay), and DNA polymerase-alpha activities were determined by regular chain extension of the nicked ACT-DNA, by determining values obtained from +/- aphidicolin-treated incubation mixtures. In all three breast carcinoma cell lines, a common trend was observed: a decrease of activities of DNA polymerase-alpha and Helicase III. A sharp decrease of activities of the glycolipid sialyltransferases: SAT-2 (CMP-NeuAc; GD3 alpha2-8 sialyltransferase) and SAT-4 (CMP-NeuAc: GM1a alpha2-3 sialyltransferase) was observed in the apoptotic carcinoma cells treated with L-PPMP compared with cis-platin.


Subject(s)
Breast Neoplasms/pathology , Cisplatin/pharmacology , DNA Replication/drug effects , Glycolipids/metabolism , Morpholines/pharmacology , Sphingolipids/pharmacology , Acetyltransferases/drug effects , Acetyltransferases/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Breast Neoplasms/drug therapy , Carbohydrate Sequence , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , DNA Helicases/drug effects , DNA Helicases/metabolism , DNA Polymerase I/drug effects , DNA Polymerase I/metabolism , DNA Replication/physiology , Dose-Response Relationship, Drug , Female , Glycosyltransferases/drug effects , Glycosyltransferases/metabolism , Humans , Molecular Sequence Data , Phosphatidylserines/metabolism , Sialyltransferases/drug effects , Sialyltransferases/metabolism
15.
J Biochem ; 139(2): 177-88, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16452305

ABSTRACT

The chromatin remodeling complex SWI/SNF is known to regulate the transcription of several genes by controlling chromatin structure in an ATP-dependent manner. SWI/SNF contains the Swi2p/Snf2p like ATPases BRG1 or BRM exclusively. We found that the expression of BRM gradually increases and that of BRG1 decreases as liver cells differentiate. Chromatin immunoprecipitation assays revealed that the ATPase subunits of SWI/SNF and tumor suppressor retinoblastoma (RB) family proteins bind to the promoter region of the albumin gene in hepatocytes, and that the replacement of BRG1 with BRM and pRB with p130 at this site occurs over the course of differentiation. Small interfering RNA experiments showed that blocking the expression of BRG1 and BRM in fetal and adult hepatocytes, respectively, causes a reduction in albumin expression. In luciferase reporter assays with a pREP4-based reporter plasmid that forms a chromatin structure, BRG1 showed activity stimulating the expression of the albumin promoter mediated by CCAAT/enhancer-binding protein alpha (C/EBPalpha). This enhancement was facilitated by the RB family members pRB and p130. ATPase assays showed that both pRB and C/EBPalpha proteins directly stimulate the ATPase activity of BRG1. Our findings suggest that the mechanism by which the activity of transcription factors is enhanced by RB family members and SWI/SNF includes an increase in the ATPase activity of the chromatin remodeling complex.


Subject(s)
Albumins/genetics , Chromosomal Proteins, Non-Histone/metabolism , Liver/growth & development , Transcription Factors/metabolism , Animals , Blotting, Western , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation/physiology , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/drug effects , DNA Helicases/genetics , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/cytology , Liver/metabolism , Mice , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/drug effects , Transcription Factors/genetics
16.
Recent Pat Anticancer Drug Discov ; 1(2): 185-200, 2006 Jun.
Article in English | MEDLINE | ID: mdl-18221036

ABSTRACT

Increasing evidence suggests that four-stranded tetraplex structures spontaneously form under physiological conditions and these alternate DNA structures are likely to form in vivo. Alternate G-quadruplex DNA structures that may form in regulatory elements of oncogenes or G-rich telomere sequences are potential targets for cancer therapy since these sequence-specific structures are proposed to affect gene expression and telomerase activation, respectively. Small molecule compounds that specifically bind tetraplexes may be used to regulate cell cycle progression by modulating promotor activation or disrupting telomere maintenance, important processes of cellular transformation. In this review, we summarize the current research developments and associated patents that bear relevance to understanding the mechanism and clinical application of tetraplex binding molecules as anti-cancer agents.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , DNA, Neoplasm/chemistry , DNA, Neoplasm/drug effects , Animals , DNA Helicases/drug effects , DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , Humans , Ligands , Nucleic Acid Conformation , Perylene/analogs & derivatives , Perylene/pharmacology , Telomerase/antagonists & inhibitors , Telomere/drug effects , Transcriptional Activation/drug effects
17.
J Biol Chem ; 279(20): 21169-76, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15026416

ABSTRACT

The Werner syndrome and the Nijmegen breakage syndrome are recessive genetic disorders that show increased genomic instability, cancer predisposition, hypersensitivity to mitomycin C and gamma-irradiation, shortened telomeres, and cell cycle defects. The protein mutated in the premature aging disease known as the Werner syndrome is designated WRN and is a member of the RecQ helicase family. The Nbs1 protein is mutated in Nijmegen breakage syndrome individuals and is part of the mammalian Mre11 complex together with the Mre11 and Rad50 proteins. Here, we show that WRN associates with the Mre11 complex via binding to Nbs1 in vitro and in vivo. In response to gamma-irradiation or mitomycin C, WRN leaves the nucleoli and co-localizes with the Mre11 complex in the nucleoplasm. We detect an increased association between WRN and the Mre11 complex after cellular exposure to gamma-irradiation. Small interfering RNA and complementation experiments demonstrated convergence of WRN and Nbs1 in response to gamma-irradiation or mitomycin C. Nbs1 is required for the Mre11 complex promotion of WRN helicase activity. Taken together, these results demonstrate a functional link between the two genetic diseases with partially overlapping phenotypes in a pathway that responds to DNA double strand breaks and interstrand cross-links.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Binding Sites , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/radiation effects , Cell Line , DNA Helicases/drug effects , DNA Helicases/genetics , DNA Helicases/radiation effects , DNA-Binding Proteins/genetics , Exodeoxyribonucleases , Gamma Rays , HeLa Cells , Humans , MRE11 Homologue Protein , Mitomycin/pharmacology , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/radiation effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/radiation effects , RecQ Helicases , Recombinant Proteins/metabolism , Reference Values , Transfection , Werner Syndrome Helicase
18.
Mol Cell Biol ; 23(18): 6385-95, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12944467

ABSTRACT

The Werner syndrome protein (WRN) is a caretaker of the human genome, and the Abl kinase is a regulator of the DNA damage response. Aberrant DNA repair has been linked to the development of cancer. Here, we have identified a direct binding between WRN and c-Abl in vitro via the N-terminal and central regions of WRN and the Src homology domain 3 of c-Abl. After bleomycin treatment in culture, WRN and c-Abl are dissociated and followed by an Abl kinase-dependent WRN relocalization to the nucleoplasm. WRN is a substrate of c-Abl in vitro and in vivo. WRN is tyrosine phosphorylated either transiently by treatment of HeLa cells with bleomycin or constitutively in cells from chronic myeloid leukemia (CML) patients, and these phosphorylations are prevented by treatment with the Abl kinase inhibitor STI-571. Tyrosine phosphorylation of WRN results in inhibition of both WRN exonuclease and helicase activities. Furthermore, anti-WRN immunoprecipitates from CML cells treated with STI-571 show increased 3'-->5' exonuclease activity. These findings suggest a novel signaling pathway by which c-Abl mediates WRN nuclear localization and catalytic activities in response to DNA damage.


Subject(s)
DNA Helicases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Animals , Benzamides , Binding Sites , Bleomycin/pharmacology , DNA Damage/physiology , DNA Helicases/drug effects , DNA Helicases/genetics , Enzyme Inhibitors/pharmacology , Exodeoxyribonucleases , HeLa Cells , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Phosphorylation , Piperazines/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/genetics , Pyrimidines/pharmacology , RecQ Helicases , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tumor Cells, Cultured , Tyrosine/metabolism , Werner Syndrome Helicase , src Homology Domains
19.
J Pineal Res ; 33(1): 48-56, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12121485

ABSTRACT

In the present study, double fluorescence staining combined with confocal laser scanning microscopy analysis were used to examine the effects of melatonin on ischemia-induced neuronal DNA strand breaks and its possible mechanisms in a transient middle cerebral artery (MCA) occlusion model. Results showed that melatonin dose-dependently reduced infarct areas and decreased both DNA double and single strand breaks (DSB and SSB) and enhanced cell viability in the peri-ischemic brain regions. Furthermore, Bcl-2 induction in the ischemic brain was further enhanced by melatonin treatment. Double staining analysis indicated that the cells costained for Bcl-2 and TdT-mediated-deoxyuridine triphosphate (dUTP) nick-end labeling (TUNEL), a DSB marker, displayed a relative regular morphology compared with the cells only stained with TUNEL. Transient ischemia induced an expression of excision repair cross-complementing factor 6 (ERCC6) mRNA, a gene essential for the preferential repair of nuclear excision repair, in the injured neurons. Double labeling showed that ERCC6 only co-localized with proliferating cell nuclear antigen (PCNA), a member of the nuclear excision repair complex, but not with TUNEL. Melatonin further and statistical significantly up-regulated ERCC6 mRNA expression in the peri-ischemic region of rat brains. The results suggest that neuroprotection by melatonin against ischemic injury may be related to modulation of apoptosis and DNA repair capacity.


Subject(s)
Brain Ischemia/prevention & control , DNA Damage/drug effects , DNA Repair/drug effects , Melatonin/pharmacology , Neurons/pathology , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain Ischemia/pathology , Cerebral Arteries , DNA/drug effects , DNA Helicases/drug effects , DNA Helicases/genetics , DNA Helicases/metabolism , DNA, Single-Stranded/drug effects , Disease Models, Animal , In Situ Nick-End Labeling , Male , Microscopy, Confocal , Neurons/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley
20.
Biochem Biophys Res Commun ; 294(2): 334-9, 2002 Jun 07.
Article in English | MEDLINE | ID: mdl-12051716

ABSTRACT

Pea DNA helicase 45 (PDH45) is an ATP-dependent DNA unwinding enzyme, with intrinsic DNA-dependent ATPase activity [Plant J. 24 (2000) 219]. We have determined the effect of various DNA-binding agents, such as daunorubicin, ethidium bromide, ellipticine, cisplatin, nogalamycin, actinomycin C1, and camptothecin on the DNA unwinding and ATPase activities of the plant nuclear DNA helicase PDH45. The results show that all the agents except actinomycin C1, and camptothecin inhibited the helicase (apparent K(i) values ranging from 1.5 to 7.0 microM) and ATPase (apparent K(i) values ranging from 2.5 to 11.9 microM) activities. This is the first study to show the effect of various DNA-binding agents on the plant nuclear helicase and also first to demonstrate inhibition of any helicase by cisplatin. Another striking finding that the actinomycin C1 and ellipticine act differentially on PDH45 as compared to pea chloroplast helicase suggests that the mechanism of DNA unwinding could be different in nucleus and chloroplast. These results suggest that the intercalation of the inhibitors into duplex DNA generates a complex that impedes translocation of PDH45, resulting in both the inhibitions of unwinding activity and ATP hydrolysis. This study would be useful to obtain a better understanding of the mechanism of plant nuclear DNA helicase unwinding and the mechanism by which these agents can disturb genome integrity.


Subject(s)
Adenosine Triphosphatases/drug effects , DNA Helicases/chemistry , DNA/chemistry , Enzyme Inhibitors/chemistry , Intercalating Agents/chemistry , Plant Proteins/chemistry , Adenosine Triphosphatases/chemistry , Adenosine Triphosphate/chemistry , Animals , Cell Nucleus/enzymology , Chloroplasts/enzymology , Cisplatin/chemistry , Cisplatin/pharmacology , DNA/metabolism , DNA Helicases/drug effects , Daunorubicin/chemistry , Daunorubicin/pharmacology , Dose-Response Relationship, Drug , Ellipticines/chemistry , Ellipticines/pharmacology , Enzyme Inhibitors/pharmacology , Ethidium/chemistry , Ethidium/pharmacology , Intercalating Agents/pharmacology , Nogalamycin/chemistry , Nogalamycin/pharmacology , Nucleic Acid Synthesis Inhibitors/chemistry , Nucleic Acid Synthesis Inhibitors/pharmacology , Pisum sativum , Plant Proteins/drug effects , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...