Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
J R Coll Physicians Edinb ; 44(3): 196-200, 2014.
Article in English | MEDLINE | ID: mdl-25318394

ABSTRACT

Targeting of microRNAs that are overexpressed or replacement of microRNAs whose expression is lost are two distinct and novel approaches to treat disease(s) driven by microRNA dysregulation. This can be achieved by chemical modification of either a single stranded oligonucleotide called an antimiR or a double stranded nucleic acid molecule termed a microRNA mimic.With hundreds of microRNAs identified and knowledge of their role in disease becoming clearer there is the prospect, over the coming years, to harness engineered microRNA therapeutics to revolutionise the way diseases are treated.Both types of engineered microRNA therapeutics have advanced into clinical development with human proof of concept achieved with an anti-miR targeting miR-122 (one of the most abundant microRNAs in human hepatocytes that is utilised by the hepatitis C virus to enable its function and replication). Rather than targeting individual proteins or enzymes involved in human disease, an opportunity now exists to modulate multiple different proteins/enzymes which act in concert in the progression of disease.


Subject(s)
MicroRNAs/antagonists & inhibitors , Oligonucleotides/therapeutic use , Animals , Biomedical Engineering , Hepacivirus/genetics , Hepatitis C/therapy , Hepatitis C/virology , Humans , Kidney/pathology , MicroRNAs/metabolism , Molecular Mimicry
2.
Br J Cancer ; 75(1): 69-75, 1997.
Article in English | MEDLINE | ID: mdl-9000600

ABSTRACT

NAD(P)H:quinone oxidoreductase (NQO1, EC 1.6.99.2) is an obligate two-electron reductase that can either bioactivate or detoxify quinones and has been proposed to play an important role in chemoprevention. We have previously characterized a homozygous point mutation in the BE human colon carcinoma cell line that leads to a loss of NQO1 activity. Sequence analysis showed that this mutation was at position 609 of the NQO1 cDNA, conferring a proline to serine substitution at position 187 of the NQO1 enzyme. Using polymerase chain reaction (PCR) analysis, we have found that the H596 human non-small-cell lung cancer (NSCLC) cell line has elevated NQO1 mRNA, but no detectable enzyme activity. Sequencing of the coding region of NQO1 from the H596 cells showed the presence of the identical homozygous point mutation present in the BE cell line. Expression and purification of recombinant wild-type and mutant protein from E. coli showed that mutant protein could be detected using immunoblot analysis and had 2% of the enzymatic activity of the wild-type protein. PCR and Northern blot analysis showed moderate to low levels of expression of the correctly sized transcript in the mutant cells. Immunoblot analysis also revealed that recombinant mutant protein was immunoreactive; however, the mutant protein was not detected in the cytosol of either BE or H596 cells, suggesting that the mutant proteins were either not translated or were rapidly degraded. The absence of any detectable, active protein, therefore, appears to be responsible for the lack of NQO1 activity in cells homozygous for the mutation. A polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis for the mutation at position 609 conducted on 90 human lung tissue samples (45 matched sets of tumour and uninvolved tissue) revealed a 7% incidence of individuals homozygous for the mutation, and 42% heterozygous for the mutation. These data suggest that the mutation at position 609 represents a polymorphism in an important xenobiotic metabolizing enzyme, which has implications for cancer therapy, chemoprevention and chemoprotection.


Subject(s)
NAD(P)H Dehydrogenase (Quinone)/genetics , Polymorphism, Genetic/genetics , Adult , Aged , Aged, 80 and over , Blotting, Northern , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Small Cell/enzymology , Carcinoma, Small Cell/genetics , DNA, Neoplasm/chemistry , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Male , Middle Aged , Point Mutation , Recombinant Proteins/metabolism , Tumor Cells, Cultured
3.
Cell Growth Differ ; 8(11): 1199-210, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9372243

ABSTRACT

Dysregulated expression of basic fibroblast growth factor [fibroblast growth factor 2 (FGF-2)] mediates autocrine growth of melanoma cells. The presence of a consensus Myb binding site in the human FGF-2 promoter prompted us to investigate whether this transcription factor could regulate FGF-2 expression in melanomas. We report that c-MYB mRNA is overexpressed in melanoma cell lines compared to normal melanocytes and that ectopic expression of murine c-Myb in SK-MEL-2 human melanoma cells resulted in increased expression of FGF-2 mRNA and FGF-2 protein. Furthermore, murine c-Myb transactivated a reporter plasmid containing the human FGF-2 promoter region in contransfected SK-MEL-2 human melanoma cells. Although a functional DNA-binding domain was required for transactivation, responsiveness to c-Myb was independent of the putative Myb binding site and mapped to two regions of the FGF-2 promoter that did not bind c-Myb in vitro. We suggest that c-MYB contributes to FGF-2-mediated autocrine growth of melanomas by indirectly regulating the FGF-2 promoter.


Subject(s)
DNA-Binding Proteins/metabolism , Fibroblast Growth Factor 2/biosynthesis , Melanoma/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Binding Sites , DNA-Binding Proteins/genetics , Fibroblast Growth Factor 2/genetics , Humans , Mice , Promoter Regions, Genetic , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myb , RNA, Messenger/metabolism , Trans-Activators/genetics , Transcriptional Activation
4.
Biochem Pharmacol ; 51(5): 645-52, 1996 Mar 08.
Article in English | MEDLINE | ID: mdl-8615901

ABSTRACT

The metabolism, cytotoxicity, and genotoxicity of streptonigrin (SN) w ere determined in two human colon carcinoma cell lines: HT-29 with high NAD(P)H:quinone oxidoreductase (EC 1.6.99.2, DTD) activity and BE with undetectable DTD activity. Dicumarol-sensitive oxidation of NADH was observed with HT-29 cytosol, but not with BE cytosol. Oxygen consumption was also observed using HT-29 cytosol, but was absent with BE cytosol. Dicumarol inhibited oxygen consumption with HT-29 cytosol, but deferoxamine had no effect, suggesting that divalent metal cations were not necessary for efficient auto-oxidation of SN hydroquinone. In cytotoxicity studies, SN was much more toxic to the DTD-rich HT-29 cells than to the DTD-deficient BE cells. Deferoxamine decreased toxicity in both cell lines, implicating hydroxyl radicals produced during Fenton-type reactions as the toxic species. In the genotoxicity assay, SN induced a much higher incidence of DNA strand breaks in HT-29 cells than in BE cells, and deferoxamine protected against DNA strand breaks in both cell lines. Some evidence of DNA repair was also observed in the two cell lines. These results support an important role for DTD in the cytotoxicity of SN in the high DTD HT-29 colon carcinoma cell line.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA Damage , DNA/drug effects , NAD(P)H Dehydrogenase (Quinone)/physiology , Streptonigrin/pharmacology , Cell Survival/drug effects , Humans , Oxidation-Reduction , Streptonigrin/metabolism , Tumor Cells, Cultured
5.
Arch Biochem Biophys ; 320(1): 51-8, 1995 Jun 20.
Article in English | MEDLINE | ID: mdl-7793984

ABSTRACT

Exposure of renal proximal tubular epithelial cells (LLC-PK1) to the nephrotoxicants 2-bromo-6-(glutathion-S-yl)hydroquinone, 2-bromo-3-(glutathion-S-yl)-hydroquinone, and 2-bromo-(diglutathion-S-yl)hydroquinone caused DNA fragmentation and cytotoxicity. Viability measured by lysosomal neutral red accumulation was the most sensitive parameter of cytotoxicity, and preceded toxicity determined by either the mitochondrial MTT assay or by measuring intracellular lactate dehydrogenase activity. DNA fragmentation was detected as early as 15 min after exposure to 2-bromo-6-(glutathion-S-yl)hydroquinone (100 microM), 2-bromo-3-(glutathion-S-yl)hydroquinone (200 microM), and 2-bromo-(diglutathion-S-yl)hydroquinone (400 microM) and prior to other indices of toxicity. The ability of the cells to repair DNA damage was evident by the decrease in the extent of single strand breaks following removal of 2-bromo-3-(glutathion-S-yl)hydroquinone from the incubation medium. Moreover, inhibition of poly(ADP-ribose)polymerase with 3-amino-benzamide (10 mM), following exposure of LLC-PK1 cells to 0.5 mM 2-bromo-6-(glutathion-S-yl)hydroquinone or 2-bromo-(diglutathion-S-yl)hydroquinone, decreased cytotoxicity, indicating that DNA repair processes, activated in response to DNA damage, exacerbate toxicity. Treatment with the endonuclease inhibitor, aurintricarboxylic acid did not decrease cytotoxicity. A decrease in the cytotoxicity caused by 2-bromo-6-(glutathion-S-yl)hydroquinone and 2-bromo-(diglutathion-S-yl)hydroquinone was observed when cells were incubated with catalase or pretreated with deferoxamine (10 mM). The data suggest a mechanism whereby the conjugates generate hydrogen peroxide, and the subsequent iron-catalyzed generation of hydroxyl radicals causes DNA fragmentation and cytotoxicity.


Subject(s)
DNA Damage , Glutathione/analogs & derivatives , Hydroquinones/toxicity , Reactive Oxygen Species/metabolism , Animals , Cell Survival/drug effects , DNA Repair , DNA, Single-Stranded/drug effects , DNA, Single-Stranded/metabolism , Endonucleases/metabolism , Glutathione/toxicity , Iron/metabolism , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Kinetics , LLC-PK1 Cells , Poly Adenosine Diphosphate Ribose/metabolism , Swine
6.
Cancer Res ; 54(14): 3766-71, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-8033096

ABSTRACT

NAD(P)H:quinone acceptor oxidoreductase (NQO1, EC 1.6.99.2) is an enzyme that is believed to play a central role in the bioreductive activation of several compounds, particularly quinones. The results of this study demonstrate that the activity of NQO1 is significantly elevated (2.5-fold) in HT-29 human colon cells that are in the plateau phase of the growth curve as opposed to cells in the exponential phase. Analysis of gene expression using semiquantitative reverse transcription-polymerase chain reaction and Northern blot analysis demonstrates that the increased enzyme activity is associated with increased NQO1 mRNA levels. Sequential trypsinization of layers of cells from HT-29 multicellular spheroids and analysis of gene expression by reverse transcription-polymerase chain reaction demonstrate that NQO1 expression is elevated in cells close to the necrotic center. Maximum expression occurs at a depth of 90-110 microns, with reduced expression as the distance toward both the surface and the necrotic center decreases. HT-29 spheroids were significantly more responsive than monolayers (concentration producing 50% inhibition, 124.6 and 364 nM, respectively) to the experimental drug, 2,5-dimethyl-3,6 diaziridinyl-1,4 benzoquinone. While the environmental stimulus responsible for causing elevated NQO1 expression has not been identified, the fact that NQO1 expression is influenced by microenvironmental conditions will have important implications for those drugs that are activated by NQO1.


Subject(s)
Colonic Neoplasms/enzymology , Quinone Reductases/biosynthesis , Aziridines/pharmacology , Base Sequence , Benzoquinones/pharmacology , Cell Division , Colonic Neoplasms/pathology , Gene Expression , Humans , Molecular Sequence Data , Quinone Reductases/genetics , Tumor Cells, Cultured
7.
Cancer Res ; 54(12): 3196-201, 1994 Jun 15.
Article in English | MEDLINE | ID: mdl-8205540

ABSTRACT

The metabolisms of two standard electron acceptors and a series of bioreductive antitumor compounds by purified rat and human DT-diaphorases (DTD) were compared. DTD was purified from rat liver cytosol and from Escherichia coli in which rat liver or human lung tumor DTD complementary DNA was expressed. Km and kcat values for menadione and 2,6-dichlorophenolindophenol reduction were similar for the three enzyme preparations except that rat E. coli DTD had 2-3-fold higher kcat values for both menadione and 2,6-dichlorophenolindophenol and a 2-3-fold higher Km for menadione than either rat liver or human E. coli DTD. Reduction of the antitumor compounds was 1.9-4.9 times faster with rat E. coli DTD than with human E. coli DTD. The antitumor compounds were reduced in the following order by rat E. coli DTD: 2,5-dimethyl-3,6-diaziridinyl-1,4-benzoquinone > streptonigrin > mitomycin A > diaziquone > mitomycin C (MC) > 5-(aziridin-1-yl)-2,4-dinitrobenzamide. The order was the same for human E. coli DTD with one exception; diaziquone was reduced slightly faster than mitomycin A. Metabolism of doxorubicin could not be detected using rat or human E. coli DTD. MC-induced DNA cross-linking was also more efficient using rat E. coli DTD relative to human E. coli DTD. Metabolism of MC by rat and human E. coli DTD was also compared under aerobic and hypoxic conditions. Rates of reduction of MC and metabolite formation were similar under aerobic and hypoxic conditions, and the toxicity of MC to DTD-rich HT-29 cells was also similar in aerobic and hypoxic conditions. In contrast, the toxicity of MC to DTD-deficient BE cells was potentiated markedly under hypoxia. These data show that although small catalytic differences between rat and human E. coli DTD can be observed, compounds such as 2,5-dimethyl-3,6-diaziridinyl-1,4-benzoquinone and streptonigrin are still excellent substrates for the human enzyme and may be useful in the therapy of tumors high in DTD activity. In addition, metabolism of MC by rat and human E. coli DTD was similar in aerobic and hypoxic conditions; in agreement with these data, cytotoxicity of MC to a DTD-rich cell line was oxygen independent. Increased MC cytotoxicity under hypoxia appears to be mediated by enzymes other than DTD.


Subject(s)
Antineoplastic Agents/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Aerobiosis , Animals , Antineoplastic Agents/pharmacokinetics , Biotransformation , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Hypoxia/physiology , Chromatography, High Pressure Liquid , DNA Damage , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Humans , Kinetics , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Mitomycin/metabolism , Mitomycin/pharmacokinetics , Mitomycin/toxicity , NAD/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Transfection , Tumor Cells, Cultured
8.
Biochemistry ; 33(19): 6024-30, 1994 May 17.
Article in English | MEDLINE | ID: mdl-8180230

ABSTRACT

In this study, we have mapped the intracellular alkylation sites of adozelesin and bizelesin, two potent analogs of CC-1065, in individual genes at the single-nucleotide level. Human colon carcinoma cells were treated with adozelesin and bizelesin, and the position of adducts were mapped within the PGK-1 and p53 genes by means of ligation-mediated polymerase chain reaction. The monofunctional alkylating agent adozelesin was found to alkylate genomic DNA predominantly within 5'-(A/T)(A/T)A* sequences. Additional sites of alkylation were observed within 5'-(A/T)(G/C)(A/T)A* sequences; however, these were considered to represent sites of medium to low preference. Bizelesin, a bifunctional analog capable of both DNA monofunctional alkylation and DNA interstrand cross-link formation, was also found to alkylate 5'-(A/T)(A/T)A* sequences. Putative bizelesin DNA interstrand cross-link sites indicated that AT-rich sequences are preferred in the intervening sequence between the two cross-linked adenines. Both six- and seven-nucleotide regions were identified as putative sites of DNA interstrand cross-link formation with 5'-TTTTTTA*, 5'-TTTATCA* and 5'-GTACTAA* sequences being preferred. Non-adenine bases are not observed as potential intracellular sites of either DNA interstrand cross-linking formation or monofunctional alkylation. Thus, the patterns of alkylation induced by adozelesin and bizelesin in genomic DNA are similar but not identical to that observed in purified cell-free DNA.


Subject(s)
Alkylating Agents/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , DNA/chemistry , Indoles/pharmacology , Urea/analogs & derivatives , Base Sequence , Benzofurans , Cyclohexenes , DNA/drug effects , Duocarmycins , Genes, p53 , Humans , Molecular Sequence Data , Phosphoglycerate Kinase/genetics , Polymerase Chain Reaction/methods , Tumor Cells, Cultured , Urea/pharmacology
9.
Cancer Res ; 54(7): 1622-6, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-8137269

ABSTRACT

Diaziridinylbenzoquinones such as 3,6-diaziridinyl-1,4-benzoquinone (DZQ) and its 2,5-methyl analog (MeDZQ) require bioreductive activation in order to elicit their cytotoxic activities. In this study, we have mapped the intracellular alkylation sites induced by DZQ and MeDZQ in a single copy gene at the nucleotide level using ligation-mediated polymerase chain reaction. We have performed this analysis in two human colon carcinoma cells, one proficient (HT-29) and one deficient (BE) in DT-diaphorase (DTD) activity. In the DTD-proficient HT-29 cell line, DZQ and MeDZQ were found to alkylate both 5'-(A/T)G(C)-3' and 5'-(A/T)A-3' sequences. This is consistent with the nucleotide preferences observed when DZQ and MeDZQ are activated by purified DTD to reactive metabolites capable of alkylating DNA in vitro (C-S. Lee, J. A. Hartley, M. D. Berardini, J. Butler, D. Siegel, D. Ross, and N. W. Gibson. Biochemistry, 31: 3019-3025, 1992). Surprisingly in the DTD-deficient BE cell line a pattern of alkylation induced by DZQ and MeDZQ similar to that observed in the DTD-proficient HT-29 cells was observed. This suggests that reductive enzymes other than DTD can be involved in activating DZQ and MeDZQ to DNA-reactive species in vivo.


Subject(s)
Antineoplastic Agents/metabolism , Aziridines/metabolism , Benzoquinones/metabolism , DNA, Neoplasm/chemistry , DNA/chemistry , DNA/metabolism , Genes, p53 , Alkylation , Base Sequence , Binding Sites , Cell Line , Colonic Neoplasms , DNA Primers , DNA, Neoplasm/metabolism , Exons , Humans , Molecular Sequence Data , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Phosphoglycerate Kinase/genetics , Polymerase Chain Reaction/methods , Tumor Cells, Cultured
10.
Oncol Res ; 6(10-11): 493-500, 1994.
Article in English | MEDLINE | ID: mdl-7620217

ABSTRACT

Because of the elevated DT-diaphorase (DTD) activity in certain tumors such as human nonsmall cell lung cancer (NCSLC), DTD is a potential target on which to base the development of new antitumor compounds. Mitomycin C is the most effective single agent used for the therapy of NSCLC and is metabolized and bioactivated by DTD. Mitomycin C is a poor substrate for DTD, however, and its metabolism is pH-dependent. We have therefore focused on identifying more efficient substrates for DTD. We have developed a metabolic and cytotoxicity screen that identifies compounds which are efficiently bioactivated by DTD. This screen utilizes both aerobic and hypoxic conditions and cell lines with both elevated and deficient DTD activity as an index of selectivity. Using the screen described above, we have identified [3-hydroxy-5-aziridinyl-1-methyl-2-(1H-indole-4,7-indione)-prop-be ta-en- alpha-ol] (E09), 2,5-diaziridinyl-1,4-benzoquinone (MeDZQ), and streptonigrin as compounds that are most efficiently bioactivated by DTD and exert selective cytotoxicity. Although certain tumors such as NSCLC have elevated DTD activity, we have characterized a point mutation at position 609 in the DTD cDNA, which codes for a proline to serine change in the protein and leads to a loss of enzyme activity. We have characterized this mutation in both BE human colon carcinoma cells and H596 human NSCLC cells. This mutation and resulting lack of DTD activity complicates the use of agents designed to target DTD in tumors.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antineoplastic Agents/pharmacokinetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Quinones/pharmacokinetics , Animals , Biotransformation , Gene Expression , Humans , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/enzymology , Oxidation-Reduction
12.
Mol Pharmacol ; 44(6): 1128-34, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8264549

ABSTRACT

Mitomycin C and porfiromycin were found to inactivate rat hepatic DT-diaphorase. Inactivation was pH dependent; little inactivation was detected at pH 5.8, but inactivation increased as the pH was raised to 7.8. Inactivation was concentration and time dependent and displayed pseudo-first-order kinetics. Inactivation was NADH dependent, indicating that reductive metabolism was necessary for inhibition. [3H]Mitomycin C was covalently bound to DT-diaphorase during inhibition, and the stoichiometry for inactivation of DT-diaphorase by mitomycin C was approximately 0.8 nmol of mitomycin C bound/nmol of enzyme. A higher molecular mass product (60 kDa) was detected by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and Western blot analysis of DT-diaphorase preincubated with NADH and mitomycin C at pH 7.8, suggesting that mitomycin C is capable of cross-linking DT-diaphorase. The kinetics of inhibition, requirement for NADH for inhibition, covalent binding of [3H] mitomycin C to DT-diaphorase, and approximate 1:1 stoichiometry suggest that this inactivation process may be mechanism based. Inhibition of DT-diaphorase by mitomycin C and porfiromycin is not limited to a cell-free system and could also be observed in HT-29 cells in culture at pH 7.2. Bioactivation of mitomycin C or porfiromycin by DT-diaphorase is favored at lower pH, whereas at higher pH values enzyme alkylation and inactivation of DT-diaphorase occur. These data suggest that the success of attempts to exploit the elevated DT-diaphorase content of certain human tumors for improved chemotherapeutic response using mitomycin C or porfiromycin will depend on intracellular pH.


Subject(s)
Mitomycin/pharmacology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Porfiromycin/pharmacology , Animals , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Humans , Hydrogen-Ion Concentration , Kinetics , NAD/metabolism , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
13.
Biochemistry ; 32(47): 12857-63, 1993 Nov 30.
Article in English | MEDLINE | ID: mdl-8251508

ABSTRACT

Two-electron reduction of the structurally related aziridinylbenzoquinones DZQ and MeDZQ to their hydroquinone forms, DZHQ and MeDZHQ, respectively, generates species which interact and cross-link DNA at distinct nucleotide sequences. Within single target site duplex oligonucleotides, DZHQ was found to cross-link DNA at 5'-GC-3' and 5'-GNNC-3' sequences, whereas MeDZHQ was found to cross-link predominantly at 5'-GNC-3' within a 5'-GTCA-3' sequence. In a multitarget site duplex oligonucleotide, which contains either the target sequence 5'-TGCAC-3' or 5'-TGCTC-3', DZHQ was found to cross-link at both a 5'-GC-3' (a 1,2 cross-link) and a 5'-GNNC-3' (a 1,4 cross-link) site with approximately equal efficiency. Molecular dynamics simulations were able to accurately reproduce the experimental results and provide a structural basis for the alkylation preferences. Calculations were performed to determine the mobility of the hydroquinone species following guanine N7 (G) alkylation at 5'-TGCAC-3' and 5'-TGTCA-3' sequences. Conformations consistent with the formation of both 1,2 and 1,4 cross-links were observed when DZHQ was placed within a 5-TGCAC-3' sequence. The 1,2 cross-link orientation was more stable and thermodynamically favored. For MeDZHQ at the same site the ligand was unable to form stable 1,2 or 1,4 cross-linking conformations, primarily due to clashes with thymine methyl groups. In contrast, the MeDZHQ monoadduct with a 5'-TGTCA-3' sequence adopted a very stable conformation consistent with formation of a 1,3 cross-link.


Subject(s)
Aziridines/chemistry , Benzoquinones/chemistry , Cross-Linking Reagents/chemistry , Hydroquinones/chemistry , Oligodeoxyribonucleotides/chemistry , Alkylation , Animals , Base Sequence , Computer Simulation , DNA Damage , Dihydrolipoamide Dehydrogenase/metabolism , Liver/enzymology , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Rats
14.
Biochemistry ; 32(35): 9108-14, 1993 Sep 07.
Article in English | MEDLINE | ID: mdl-8369282

ABSTRACT

Bizelesin, a cyclopropylpyrroloindole (CPI) antitumor agent, has been shown to alkylate and cross-link DNA within A/T-rich tracts. Previous studies have shown that covalent reaction of the CPI adozelesin with DNA was reversible [Warpehoski, M. A., Harper, D. E., Mitchell, M. A., & Monroe, T. J. (1992) Biochemistry 31, 2502-2508]. That is, the monofunctional adduct could be lost from DNA, thus restoring the fidelity of DNA. In this study, we demonstrate that covalent DNA adducts induced by bizelesin at the adenine N3 position undergo two subsequent competing reactions: one which causes DNA strand cleavage, via depurination, and one which proceeds through loss of the DNA adduct (adduct reversal with restoration of DNA integrity). Our results were obtained by studying the chemical stability of synthetic DNA oligonucleotides which contained either a distinct DNA monofunctional adduct or DNA interstrand cross-links. Quantification of adduct reversal was performed on the basis that drug-modified DNA, upon exposure to heat followed by hot piperidine treatment, was resistant to strand cleavage at the site of alkylation. The rate of adduct reversal was found to increase with increasing temperature and was found to be maximum at 70-80 degrees C. The rate of adduct reversal was also found to increase with increasing pH and ionic strength. In contrast, the rate of depurination and subsequent DNA strand cleavage decreased as pH and ionic strength were increased. Adduct reversal was favored in DNA containing interstrand cross-links, whereas rapid depurination occurred preferentially within monofunctionally alkylated DNA.


Subject(s)
Antineoplastic Agents/chemistry , Cross-Linking Reagents/chemistry , DNA Damage , Indoles/chemistry , Oligodeoxyribonucleotides/chemistry , Urea/analogs & derivatives , Base Sequence , Duocarmycins , Hydrogen-Ion Concentration , Models, Chemical , Molecular Sequence Data , Sodium Hydroxide , Urea/chemistry
15.
Biochemistry ; 32(21): 5518-25, 1993 Jun 01.
Article in English | MEDLINE | ID: mdl-8504072

ABSTRACT

Aerobic reduction of MMC by DTD, an obligate two-electron reductase, or chemical reduction by sodium borohydride results predominantly in monoalkylation of DNA at the guanine N7 position within 5'-GG-3' and 5'-GTC-3' sequences. The level of guanine N7 alkylation after DTD reduction increased as the pH was decreased from 7.8 and was optimal at pH 6.6. A similar profile of alkylation was obtained when the major metabolite of DTD-mediated MMC metabolism, 2,7-diaminomitosene, was further reduced by DTD. The sequence preference for DNA interstrand cross-linking (ISC) was also determined using singly end-labeled oligonucleotide duplexes. Reduction of MMC by DTD induced DNA cross-links which were resistant to piperidine cleavage. Exposure of cross-linked DNA to dimethyl sulfate or formic acid and subsequent piperidine cleavage displayed a discontinuity in band pattern which suggested a 5'-CG-3' preference for DNA ISC. Major groove alkylation is proposed to occur via generation, and subsequent metabolism by DTD, of 2,7-diaminomitosene. Cross-linking of DNA, at 5'-CG-3' sequences, is proposed to require the formation of either the protonated leucomitomycin C or the leucoaziridinomitosene during DTD-mediated metabolism of MMC.


Subject(s)
Cross-Linking Reagents/pharmacology , Mitomycin/metabolism , Mitomycin/pharmacology , NAD(P)H Dehydrogenase (Quinone)/metabolism , Aerobiosis , Alkylation , Base Sequence , Cross-Linking Reagents/metabolism , DNA/metabolism , Molecular Sequence Data , Plasmids , Restriction Mapping
16.
Cancer Metastasis Rev ; 12(2): 83-101, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8375023

ABSTRACT

A role of DTD in the bioreductive activation of mitomycin C was supported by indirect evidence utilizing enzyme inhibitors in cellular systems. Using a cell-free system, we have confirmed that DTD can bioactivate mitomycin C using both purified rat and human DTD. Metabolism and bioactivation of mitomycin C by DTD is pH-dependent. At pH 7.8 alkylation of DTD leading to enzyme inhibition and DTD crosslinking occurs whereas at pH values of 7.4 and below metabolite formation, preservation of catalytic activity of DTD and sequence-selective DNA crosslinking occurs. Bioactivation of mitomycin C by DTD and the cytotoxicity of this drug in DTD-rich cell lines is oxygen-independent. Mitomycin C induces greater DNA crosslinking, even after chemical reduction, at lower pH values. This suggests that if mitomycin C is used in tumors with elevated DTD activity, greater therapeutic activity may be obtained by lowering intratumoral pH. Human NSCLC has elevated DTD activity relative to SCLC and normal lung and may be a target for the development of drugs which can be efficiently bioactivated by DTD. Because of the pH-dependent inactivation of DTD by mitomycin C, however, other drugs which are efficiently metabolized and bioactivated by DTD may be better candidates for the therapy of tumors high in DTD such as NSCLC.


Subject(s)
Mitomycin/pharmacology , Mitomycin/pharmacokinetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Quinones/pharmacology , Quinones/pharmacokinetics , Animals , Cell Line , Humans , Inactivation, Metabolic , Mitomycin/toxicity , NADPH-Ferrihemoprotein Reductase/metabolism , Quinones/toxicity , Tumor Cells, Cultured
17.
Biochemistry ; 32(13): 3306-12, 1993 Apr 06.
Article in English | MEDLINE | ID: mdl-8461296

ABSTRACT

The nucleotide sequence preferences for the formation of interstrand cross-links induced in DNA by 2,5-diaziridinyl-1,4-benzoquinone (DZQ) and 3,6-dimethyl-2,5-diaziridinyl-1,4-benzoquinone (MeDZQ) were studied using synthetic duplex oligonucleotides and denaturing polyacrylamide gel electrophoresis (PAGE). Reaction of these bifunctional alkylating agents with a DNA duplex containing several potential cross-linking sites resulted in the formation of cross-linked DNAs with different electrophoretic mobilities. Analysis of the principal cross-linked products by piperidine fragmentation revealed that the preferential site of cross-linking was altered from a 5'-GNC to a 5'-GC sequence upon reduction of DZQ to the hydroquinone form by the enzyme DT-diaphorase. In contrast, the reduced form of MeDZQ was found to preferentially cross-link at 5'-GNC sites within the same sequence. These preferences were confirmed in duplex oligonucleotides containing single potential cross-linking sites. Additional minor cross-linked products were characterized and revealed that DZQ and MeDZQ are both capable of cross-linking across four base pairs in a 5'-GNNC sequence.


Subject(s)
Aziridines/chemistry , Benzoquinones/chemistry , Cross-Linking Reagents/chemistry , DNA/chemistry , Alkylation , Base Sequence , Guanine/chemistry , Molecular Sequence Data , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oligodeoxyribonucleotides/chemistry , Oxidation-Reduction
19.
Biochemistry ; 32(10): 2592-600, 1993 Mar 16.
Article in English | MEDLINE | ID: mdl-8448117

ABSTRACT

Using a 21-base-pair duplex oligonucleotide containing a centrally located defined cross-linkable site, we have separated by gel electrophoresis DNA interstrand cross-links (ISC) from monofunctionally alkylated DNA (MA) and investigated the sequence selectivity for DNA ISC induced by the CC-1065 analogue U-77,779 (U-77). Sequencing gel analysis shows that U-77 induces two distinct types of DNA ISC. The first distinct form of DNA ISC spans six nucleotides and links two adenine N3 positions within an A/T-rich sequence. The second distinct DNA ISC spans seven nucleotides, also linking two adenine N3 positions, with a preference for contiguous runs of adenines. Three major 6-nucleotide DNA ISC's were identified and found to occur within 5'-TAATTA-3', 5'-TAAATA-3', and 5'-TAAAAA-3' sequences. The major 7-nucleotide DNA ISC was found to occur within 5'-TAAAAAA-3' sequences. Within this sequence, the formation of the 7-nucleotide DNA ISC was preferred over the 6-nucleotide DNA ISC by a ratio of approximately 2:1. DNA ISC formation within adenine tracts eliminated the inherent DNA bending associated with such sequences. Further, chemical probing of each isolated DNA ISC with diethyl pyrocarbonate (A-specific) and potassium permanganate (T-specific) shows that the major DNA conformational changes, such as helical distortion, were localized within the cross-linked sequence. These results suggest that a significant degree of DNA distortion may occur as a consequence of interstrand cross-linking.


Subject(s)
Antineoplastic Agents , Cross-Linking Reagents , DNA/chemistry , Indoles , Intercalating Agents , Oligodeoxyribonucleotides/chemistry , Urea/analogs & derivatives , Alkylation , Base Sequence , Duocarmycins , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data , Nucleic Acid Conformation , Nucleic Acid Denaturation , Oligodeoxyribonucleotides/chemical synthesis , Structure-Activity Relationship
20.
Article in English | MEDLINE | ID: mdl-8312113

ABSTRACT

In conclusion, recent work has highlighted the fact that NSCLCs have elevated levels of NQO1 activity and that such an increase represents an excellent target for therapeutic exploitation. The 5-year survival rate seen with lung cancer is dismal and there is a high number of cancer deaths associated with this disease each year. This renders the design of molecules that can be activated by NQO1 (such as MeDZQ or CB 10-200) an extremely important and urgent issue.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/enzymology , Mitomycin/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Drug Design , Humans , Oxidation-Reduction , Quinones/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...