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










Publication year range
1.
J Biol Chem ; 300(7): 107358, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38782206

ABSTRACT

Aristolochic acids I and II (AA-I/II) are carcinogenic principles of Aristolochia plants, which have been employed in traditional medicinal practices and discovered as food contaminants. While the deleterious effects of AAs are broadly acknowledged, there is a dearth of information to define the mechanisms underlying their carcinogenicity. Following bioactivation in the liver, N-hydroxyaristolactam and N-sulfonyloxyaristolactam metabolites are transported via circulation and elicit carcinogenic effects by reacting with cellular DNA. In this study, we apply DNA adduct analysis, X-ray crystallography, isothermal titration calorimetry, and fluorescence quenching to investigate the role of human serum albumin (HSA) in modulating AA carcinogenicity. We find that HSA extends the half-life and reactivity of N-sulfonyloxyaristolactam-I with DNA, thereby protecting activated AAs from heterolysis. Applying novel pooled plasma HSA crystallization methods, we report high-resolution structures of myristic acid-enriched HSA (HSAMYR) and its AA complexes (HSAMYR/AA-I and HSAMYR/AA-II) at 1.9 Å resolution. While AA-I is located within HSA subdomain IB, AA-II occupies subdomains IIA and IB. ITC binding profiles reveal two distinct AA sites in both complexes with association constants of 1.5 and 0.5 · 106 M-1 for HSA/AA-I versus 8.4 and 9.0 · 105 M-1 for HSA/AA-II. Fluorescence quenching of the HSA Trp214 suggests variable impacts of fatty acids on ligand binding affinities. Collectively, our structural and thermodynamic characterizations yield significant insights into AA binding, transport, toxicity, and potential allostery, critical determinants for elucidating the mechanistic roles of HSA in modulating AA carcinogenicity.

2.
Carcinogenesis ; 37(7): 647-655, 2016 07.
Article in English | MEDLINE | ID: mdl-27207664

ABSTRACT

Aristolochic acids (AA) are implicated in the development of chronic renal disease and upper urinary tract carcinoma in humans. Using in vitro approaches, we demonstrated that N-hydroxyaristolactams, metabolites derived from partial nitroreduction of AA, require sulfotransferase (SULT)-catalyzed conjugation with a sulfonyl group to form aristolactam-DNA adducts. Following up on this observation, bioactivation of AA-I and N-hydroxyaristolactam I (AL-I-NOH) was studied in human kidney (HK-2) and skin fibroblast (GM00637) cell lines. Pentachlorophenol, a known SULT inhibitor, significantly reduced cell death and aristolactam-DNA adduct levels in HK-2 cells following exposure to AA-I and AL-I-NOH, suggesting a role for Phase II metabolism in AA activation. A gene knockdown, siRNA approach was employed to establish the involvement of selected SULTs and nitroreductases in AA-I bioactivation. Silencing of SULT1A1 and PAPSS2 led to a significant decrease in aristolactam-DNA levels in both cell lines following exposure to AA-I, indicating the critical role for sulfonation in the activation of AA-I in vivo Since HK-2 cells proved relatively resistant to knockdown with siRNAs, gene silencing of xanthine oxidoreductase, cytochrome P450 oxidoreductase and NADPH:quinone oxidoreductase was conducted in GM00637 cells, showing a significant increase, decrease and no effect on aristolactam-DNA levels, respectively. In GM00637 cells exposed to AL-I-NOH, suppressing the SULT pathway led to a significant decrease in aristolactam-DNA formation, mirroring data obtained for AA-I. We conclude from these studies that SULT1A1 is involved in the bioactivation of AA-I through the sulfonation of AL-I-NOH, contributing significantly to the toxicities of AA observed in vivo.


Subject(s)
Aristolochic Acids/metabolism , Arylsulfotransferase/genetics , Multienzyme Complexes/genetics , Sulfate Adenylyltransferase/genetics , Arylsulfotransferase/antagonists & inhibitors , Arylsulfotransferase/metabolism , Carcinogens/metabolism , Carcinogens/toxicity , DNA/genetics , DNA/metabolism , Fibroblasts/metabolism , Gene Knockdown Techniques , Humans , Kidney/metabolism , Kidney/pathology , Multienzyme Complexes/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Pentachlorophenol/pharmacology , RNA, Small Interfering , Sulfate Adenylyltransferase/metabolism , Xanthine Dehydrogenase/metabolism
3.
Biopolymers ; 103(9): 491-508, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25991500

ABSTRACT

The magnitude and nature of lesion-induced energetic perturbations empirically correlate with mutagenicity/cytotoxicity profiles and can be predictive of lesion outcomes during polymerase-mediated replication in vitro. In this study, we assess the sequence and counterbase-dependent energetic impact of the Thymine glycol (Tg) lesion on a family of deoxyoligonucleotide duplexes. Tg damage arises from thymine and methyl-cytosine exposure to oxidizing agents or radiation-generated free-radicals. The Tg lesion blocks polymerase-mediated DNA replication in vitro and the unrepaired site elicits cytotoxic lethal consequences in vivo. Our combined calorimetric and spectroscopic characterization correlates Tg -induced energetic perturbations with biological and structural properties. Specifically, we incorporate a 5R-Tg isomer centered within the tridecanucleotide sequence 5'-GCGTACXCATGCG-3' (X = Tg or T) which is hybridized with the corresponding complementary sequence 5'-CGCATGNGTACGC-3' (N = A, G, T, C) to generate families of Tg -damaged (Tg ·N) and lesion-free (T·N) duplexes. We demonstrate that the magnitude and nature of the Tg destabilizing impact is dependent on counterbase identity (i.e., A ∼ G < T < C). The observation that a Tg lesion is less destabilizing when positioned opposite purines suggests that favorable counterbase stacking interactions may partially compensate lesion-induced perturbations. Moreover, the destabilizing energies of Tg ·N duplexes parallel their respective lesion-free T·N mismatch counterparts (i.e., G < T < C). Elucidation of Tg-induced destabilization relative to the corresponding undamaged mismatch energetics allows resolution of lesion-specific and sequence-dependent impacts. The Tg-induced energetic perturbations are consistent with its replication blocking properties and may serve as differential recognition elements for discrimination by the cellular repair machinery.


Subject(s)
DNA/chemistry , Thymine/analogs & derivatives , Calorimetry, Differential Scanning , Circular Dichroism , DNA Damage/genetics , Nucleic Acid Conformation , Thermodynamics , Thymine/chemistry
4.
Chem Res Toxicol ; 27(7): 1236-42, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-24877584

ABSTRACT

Plants from the Aristolochia genus have been recommended for the treatment of a variety of human ailments since the time of Hippocrates. However, many species produce the highly toxic aristolochic acids (AAs), which are both nephrotoxic and carcinogenic. For the purposes of extensive biological studies, a versatile approach to the synthesis of the AAs and their major metabolites was devised based primarily on a Suzuki-Miyaura coupling reaction. The key to success lies in the preparation of a common ring-A precursor, namely, the tetrahydropyranyl ether of 2-nitromethyl-3-iodo-4,5-methylendioxybenzyl alcohol (27), which was generated in excellent yield by oxidation of the aldoxime precursor 26. Suzuki-Miyaura coupling of 27 with a variety of benzaldehyde 2-boronates was accompanied by an aldol condensation/elimination reaction to give the desired phenanthrene intermediate directly. Deprotection of the benzyl alcohol followed by two sequential oxidation steps gave the desired phenanthrene nitrocarboxylic acids. This approach was used to synthesize AAs I-IV and several other related compounds, including AA I and AA II bearing an aminopropyloxy group at position-6, which were required for further conversion to fluorescent biological probes. Further successful application of the Suzuki-Miyaura coupling reaction to the synthesis of the N-hydroxyaristolactams of AA I and AA II then allowed the synthesis of the putative, but until now elusive, N-acetoxy- and N-sulfonyloxy-aristolactam metabolites.


Subject(s)
Aristolochic Acids/chemical synthesis , Aristolochic Acids/chemistry , DNA Adducts , Lactams/chemical synthesis
5.
Carcinogenesis ; 35(8): 1814-22, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24743514

ABSTRACT

Aristolochic acids are potent human carcinogens; the role of phase II metabolism in their bioactivation is unclear. Accordingly, we tested the ability of the partially reduced metabolites, N-hydroxyaristolactams (AL-NOHs), and their N-O-sulfonated and N-O-acetylated derivatives to react with DNA to form aristolactam-DNA adducts. AL-NOHs displayed little or no activity in this regard while the sulfo- and acetyl compounds readily form DNA adducts, as detected by (32)P-post-labeling analysis. Mouse hepatic and renal cytosols stimulated binding of AL-NOHs to DNA in the presence of adenosine 3'-phosphate 5'-phosphosulfate (PAPS) but not of acetyl-CoA. Using Time of Flight liquid chromatography/mass spectrometry, N-hydroxyaristolactam I formed the sulfated compound in the presence of PAPS and certain human sulfotransferases, SULT1B1 >>> SULT1A2 > SULT1A1 >>> SULT1A3. The same pattern of SULT reactivity was observed when N-hydroxyaristolactam I was incubated with these enzymes and PAPS and the reaction was monitored by formation of aristolactam-DNA adducts. In the presence of human NAD(P)H: quinone oxidoreductase, the ability of aristolochic acid I to bind DNA covalently was increased significantly by addition of PAPS and SULT1B1. We conclude from these studies that AL-NOHs, formed following partial nitroreduction of aristolochic acids, serve as substrates for SULT1B1, producing N-sulfated esters, which, in turn, are converted to highly active species that react with DNA and, potentially, cellular proteins, resulting in the genotoxicity and nephrotoxicity associated with ingestion of aristolochic acids by humans.


Subject(s)
Aristolochic Acids/pharmacology , Carcinogens/pharmacology , DNA Adducts/drug effects , Fibroblasts/drug effects , Animals , Arylsulfotransferase/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Cytosol/metabolism , DNA Adducts/metabolism , Ethanolamines , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C3H , Models, Molecular , Molecular Structure , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidoreductases/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stearic Acids , Sulfotransferases/metabolism
6.
Chem Res Toxicol ; 25(5): 1119-31, 2012 May 21.
Article in English | MEDLINE | ID: mdl-22515372

ABSTRACT

Aristolochic acids (AAs) are a structurally related family of nephrotoxic and carcinogenic nitrophenanthrene compounds found in Aristolochia herbaceous plants, many of which have been used worldwide for medicinal purposes. AAs have been implicated in the etiology of so-called Chinese herbs nephropathy and of Balkan endemic nephropathy. Both of these disease syndromes are associated with carcinomas of the upper urinary tract (UUC). 8-Methoxy-6-nitrophenanthro-[3,4-d]-1,3-dioxolo-5-carboxylic acid (AA-I) is a principal component of Aristolochia herbs. Following metabolic activation, AA-I reacts with DNA to form aristolactam (AL-I)-DNA adducts. We have developed a sensitive analytical method, using ultraperformance liquid chromatography-electrospray ionization/multistage mass spectrometry (UPLC-ESI/MS(n)) with a linear quadrupole ion-trap mass spectrometer, to measure 7-(deoxyadenosin-N(6)-yl) aristolactam I (dA-AL-I) and 7-(deoxyguanosin-N(2)-yl) aristolactam I (dG-AL-I) adducts. Using 10 µg of DNA for measurements, the lower limits of quantitation of dA-AL-I and dG-AL-I are, respectively, 0.3 and 1.0 adducts per 10(8) DNA bases. We have used UPLC-ESI/MS(n) to quantify AL-DNA adducts in tissues of rodents exposed to AA and in the renal cortex of patients with UUC who reside in Taiwan, where the incidence of this uncommon cancer is the highest reported for any country in the world. In human tissues, dA-AL-I was detected at levels ranging from 9 to 338 adducts per 10(8) DNA bases, whereas dG-AL-I was not found. We conclude that UPLC-ESI/MS(n) is a highly sensitive, specific and robust analytical method, positioned to supplant (32)P-postlabeling techniques currently used for biomonitoring of DNA adducts in human tissues. Importantly, UPLC-ESI/MS(n) could be used to document exposure to AA, the toxicant responsible for AA nephropathy and its associated UUC.


Subject(s)
Aristolochic Acids/analysis , DNA Adducts/analysis , Spectrometry, Mass, Electrospray Ionization/methods , Adult , Aged , Animals , Aristolochia/chemistry , Balkan Nephropathy/etiology , Chromatography, Liquid/methods , Female , Humans , Kidney/metabolism , Kidney Diseases/etiology , Limit of Detection , Male , Mice , Mice, Inbred C57BL , Middle Aged
7.
Chem Res Toxicol ; 25(1): 130-9, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22118289

ABSTRACT

Aristolochic acids are natural nitro-compounds found globally in the plant genus Aristolochia that have been implicated in the severe illness in humans termed aristolochic acid nephropathy (AAN). Aristolochic acids undergo nitroreduction, among other metabolic reactions, and active intermediates arise that are carcinogenic. Previous experiments with rats showed that aristolochic acid I (AA-I), after oral administration or injection, is subjected to detoxication reactions to give aristolochic acid Ia, aristolactam Ia, aristolactam I, and their glucuronide and sulfate conjugates that can be found in urine and feces. Results obtained with whole rats do not clearly define the role of liver and kidney in such metabolic transformation. In this study, in order to determine the specific role of the kidney on the renal disposition of AA-I and to study the biotransformations suffered by AA-I in this organ, isolated kidneys of rats were perfused with AA-I. AA-I and metabolite concentrations were determined in perfusates and urine using HPLC procedures. The isolated perfused rat kidney model showed that AA-I distributes rapidly and extensively in kidney tissues by uptake from the peritubular capillaries and the tubules. It was also established that the kidney is able to metabolize AA-I into aristolochic acid Ia, aristolochic acid Ia O-sulfate, aristolactam Ia, aristolactam I, and aristolactam Ia O-glucuronide. Rapid demethylation and sulfation of AA-I in the kidney generate aristolochic acid Ia and its sulfate conjugate that are voided to the urine. Reduction reactions to give the aristolactam metabolites occur to a slower rate. Renal clearances showed that filtered AA-I is reabsorbed at the tubules, whereas the metabolites are secreted. The unconjugated metabolites produced in the renal tissues are transported to both urine and perfusate, whereas the conjugated metabolites are almost exclusively secreted to the urine.


Subject(s)
Aristolochic Acids/metabolism , Carcinogens/metabolism , Kidney/metabolism , Animals , Aristolochic Acids/pharmacokinetics , Aristolochic Acids/urine , Biotransformation , Carcinogens/pharmacokinetics , Chromatography, High Pressure Liquid , Drugs, Chinese Herbal/metabolism , Drugs, Chinese Herbal/pharmacokinetics , In Vitro Techniques , Liver/metabolism , Male , Perfusion , Rats , Rats, Sprague-Dawley , Rats, Wistar , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
8.
Nucleic Acids Res ; 38(1): 339-52, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19854934

ABSTRACT

Aristolochic acids I and II (AA-I, AA-II) are found in all Aristolochia species. Ingestion of these acids either in the form of herbal remedies or as contaminated wheat flour causes a dose-dependent chronic kidney failure characterized by renal tubulointerstitial fibrosis. In approximately 50% of these cases, the condition is accompanied by an upper urinary tract malignancy. The disease is now termed aristolochic acid nephropathy (AAN). AA-I is largely responsible for the nephrotoxicity while both AA-I and AA-II are genotoxic. DNA adducts derived from AA-I and AA-II have been isolated from renal tissues of patients suffering from AAN. We describe the total synthesis, de novo, of the dA and dG adducts derived from AA-II, their incorporation site-specifically into DNA oligomers and the splicing of these modified oligomers into a plasmid construct followed by transfection into mouse embryonic fibroblasts. Analysis of the plasmid progeny revealed that both adducts blocked replication but were still partly processed by DNA polymerase(s). Although the majority of coding events involved insertion of correct nucleotides, substantial misincorporation of bases also was noted. The dA adduct is significantly more mutagenic than the dG adduct; both adducts give rise, almost exclusively, to misincorporation of dA, which leads to AL-II-dA-->T and AL-II-dG-->T transversions.


Subject(s)
Aristolochic Acids/chemical synthesis , DNA Adducts/chemical synthesis , Mutagenesis , Animals , Aristolochic Acids/chemistry , Cells, Cultured , DNA/biosynthesis , DNA Adducts/chemistry , Mice , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides/chemical synthesis , Oligodeoxyribonucleotides/chemistry
9.
Biopolymers ; 93(4): 370-82, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19937758

ABSTRACT

Acrolein is an alpha,beta-unsaturated aldehyde that is a major environmental pollutant, as well as a product of cellular metabolism. DNA bases react with acrolein to form two regioisomeric exocyclic guanine adducts, namely gamma-hydroxy-propanodeoxyguanosine (gamma-OH-PdG) and its positional isomer alpha-hydroxy-propanodeoxyguanosine (alpha-OH-PdG). The gamma-OH-PdG isomer adopts a ring-opened conformation with minimal structural perturbation of the DNA host duplex. Conversely, the alpha-OH-PdG isomer assumes a ring-closed conformation that significantly disrupts Watson-Crick base-pair alignments within the immediate vicinity of the damaged site. We have employed a combination of calorimetric and spectroscopic techniques to characterize the thermodynamic origins of these lesion-induced structural alterations. Specifically, we have assessed the energetic impact of alpha-OH-PdG centered within an 11-mer duplex by hybridizing the adduct-containing oligonucleotide with its complementary strand harboring a central base N [where N = C or A], yielding a pair of duplexes containing the nascent lesion (alpha-OH-PdG.C) or mismatched adduct (alpha-OH-PdG.A), respectively. Our data reveal that the nascent lesion is highly destabilizing, whereas its mismatched counterpart partially ameliorates alpha-OH-PdG-induced destabilization. Collectively, our data provide energetic characterizations of the driving forces that modulate error-free versus error-prone DNA translesion synthesis. The biological implications of our findings are discussed in terms of energetically probing acrolein-mediated mutagenicity versus adduct-induced genotoxicity.


Subject(s)
DNA Adducts/chemistry , DNA Damage , DNA/chemistry , Guanine/analogs & derivatives , Guanine/chemistry , Mutagenesis , Acrolein/chemistry , Acrolein/toxicity , Calorimetry, Differential Scanning , Circular Dichroism , Oligonucleotides/chemistry , Thermodynamics
10.
Chem Res Toxicol ; 19(6): 734-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16780350

ABSTRACT

The 2'-deoxyguanosine adduct of the dietary mutagen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) has been synthesized and incorporated into DNA using solid state synthesis technology. The key step to obtaining the C8-dG adduct is a palladium (Xantphos-chelated)-catalyzed N-arylation (Buchwald-Hartwig reaction) of PhIP by a suitably protected 8-bromo-2'-deoxyguanosine derivative. The reaction proceeded in good yield without complicating side products, and the adduct was converted to the required 5'-O-DMT-3'-O-phosphoramidite by standard methods. This modified deoxynucleoside was used to synthesize three oligodeoxynucleotides in which the C8-PhIP-dG adduct was incorporated at a single site. The oligomers were purified by reverse phase HPLC and characterized by mass spectrometry.


Subject(s)
DNA/chemical synthesis , Deoxyguanosine/chemistry , Imidazoles/chemical synthesis , Chromatography, High Pressure Liquid , DNA/chemistry , Food , Imidazoles/chemistry , Imidazoles/metabolism , Molecular Structure , Mutagens/chemistry , Spectrometry, Mass, Electrospray Ionization
11.
Mol Cell Proteomics ; 5(5): 858-67, 2006 May.
Article in English | MEDLINE | ID: mdl-16474175

ABSTRACT

Apurinic/apyrimidinic (AP) sites, a prominent type of DNA damage, are repaired through the base excision repair mechanism in both prokaryotes and eukaryotes and may interfere with many other cellular processes. A full repertoire of AP site-binding proteins in cells is presently unknown, preventing reliable assessment of harm inflicted by these ubiquitous lesions and of their involvement in the flux of DNA metabolism. We present a proteomics-based strategy for assembling at least a partial catalogue of proteins capable of binding AP sites in DNA. The general scheme relies on the sensitivity of many AP site-bound protein species to NaBH(4) cross-linking. An affinity-tagged substrate is used to facilitate isolation of the cross-linked species, which are then separated and analyzed by mass spectrometry methods. We report identification of seven proteins from Escherichia coli (AroF, DnaK, MutM, PolA, TnaA, TufA, and UvrA) and two proteins from bakers' yeast (ARC1 and Ygl245wp) reactive for AP sites in this system.


Subject(s)
DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , Proteomics , Cell Extracts , Cross-Linking Reagents , DNA-Binding Proteins/chemistry , Escherichia coli , Saccharomyces cerevisiae , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Streptavidin
12.
Anal Chem ; 77(21): 7032-8, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16255606

ABSTRACT

Environmental endocrine disruptors such as estrone (E1) and beta-estradiol (E2) are excreted in human urine primarily as water-soluble glucuronides and sulfates that can dissociate in wastewater treatment systems to the more active free estrogens. Measurement of the distribution and fate of the steroid conjugates and the corresponding free estrogens in treatment plants and receiving waters is critical for understanding the reproductive and developmental effects of these substances on aquatic organisms. A sensitive method to measure steroid estrogen conjugates in matrix-rich sewage influents and effluents (method detection limits ranged from 0.04 to 0.28 ng/L) has been developed using HPLC tandem mass spectrometry with electrospray ionization. The method employs extensive sample purification by selective extraction from an Oasis HLB solid-phase cartridge followed by separation by anion exchange chromatography. This purification scheme, combined with a stable isotope dilution approach, was used to overcome problems of matrix suppression of ionization and permitted selective and sensitive detection of six target conjugates of E1 and E2. Accurate quantitation was highly dependent on the method of sample preservation. Acidification of each sample (pH 2.0) was effective in preventing enzymatic or chemical decomposition of steroid conjugates in all sample types, whereas glucuronide conjugates were hydrolyzed in the presence of mercury and formalin preservatives. Measured concentrations of steroid sulfates in the influent to a sewage treatment plant were approximately 100 times greater than that of the respective steroid glucuronides, suggesting that the preponderance of glucuronides had dissociated prior to reaching the treatment plant. A small percentage of the steroid sulfates persisted through biological treatment of sewage and was measured in the effluent. Steroid conjugates that survive decomposition or bypass biological treatment of municipal wastewater are released into surface waters and may serve as a source of free steroids.


Subject(s)
Chromatography, Liquid/methods , Estrogens/analysis , Sewage/chemistry , Tandem Mass Spectrometry/methods , Estrogens/metabolism , Reproducibility of Results , Sensitivity and Specificity , Water Pollutants, Chemical/analysis
13.
Chem Res Toxicol ; 18(3): 457-65, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15777085

ABSTRACT

In previous work we described an efficient procedure for the synthesis of the respective N2 and N6 adducts of 2'-deoxyguanosine (dG) and 2'-deoxyadenosine (dA) derived from a series of aminoaryl compounds. We now outline methods for the site-specific introduction into oligomeric DNA of the adducts dG-N2-AN (6), dG-N2-AAN (7), dG-N2-AF (8), and dG-N2-AAF (9) derived from 2-aminonaphthalene (2-AN) or 2-aminofluorene (2-AF). For the 2-AN adduct 7, containing an acetylamino group, the 5'-O-4,4'-dimethoxytrityl- (DMT-) 3'-O-phosphoramidite (14) required for automated DNA synthesis was synthesized in high yield via the sequence 10-->11-->14. On the other hand, introduction of the desacetyl adduct 6 into oligomeric DNA was accomplished via the N-trifluoroacetyl-DMT-phosphoramidite derivative 18. This involved a similar sequence (10-->15-->18) except that the order of the reactions was changed to avoid a decomposition that occurred when the silyl-protected amino derivative 11 was treated with trifluoroacetic anhydride. In the 2-AF series the 5'-O-DMT-3'-O-phosphoramidites 27a and 27b, related to 8 and 9, were prepared by similar methods. Again, however, the order of the reactions was changed to avoid the extreme insolubility associated with the N2-[3-(2-acetylaminofluoren-3-yl)]dG (dG-N2-AAF, 9) adduct that we had noted previously. The incorporation into oligomeric DNA of the acetylamino compounds 7 and 9 proceeded smoothly and in high yield (95-100%). By contrast, the trifluoroacetyl analogues led in both the naphthyl and fluorenyl series to a mixture of oligomers containing the desired free amino adduct (6 or 8) accompanied by the N-acetyl adduct (7 or 9, respectively, after the deprotection step), indicating secondary acetylation by the capping agent acetic anhydride.


Subject(s)
2-Naphthylamine/chemistry , DNA Adducts/chemical synthesis , Deoxyguanosine/chemistry , Fluorenes/chemistry , Oligonucleotides/chemistry , DNA Adducts/chemistry , Oligonucleotides/chemical synthesis
14.
J Biol Chem ; 279(40): 41310-8, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15280390

ABSTRACT

In mammalian cells scavenger receptor class B, type I (SR-BI), mediates the selective uptake of high density lipoprotein (HDL) cholesteryl ester into hepatic and steroidogenic cells. In addition, SR-BI has a variety of effects on plasma membrane properties including stimulation of the bidirectional flux of free cholesterol (FC) between cells and HDL and changes in the organization of plasma membrane FC as indicated by increased susceptibility to exogenous cholesterol oxidase. Recent studies in SR-BI-deficient mice and in SR-BI-expressing Sf9 insect cells showed that SR-BI has significant effects on plasma membrane ultrastructure. The present study was designed to test the range of SR-BI effects in Sf9 insect cells that typically have very low cholesterol content and a different phospholipid profile compared with mammalian cells. The results showed that, as in mammalian cells, SR-BI expression increased HDL cholesteryl ester selective uptake, cellular cholesterol mass, FC efflux to HDL, and the sensitivity of membrane FC to cholesterol oxidase. These activities were diminished or absent upon expression of the related scavenger receptor CD36. Thus, SR-BI has fundamental effects on cholesterol flux and membrane properties that occur in cells of evolutionarily divergent origins. Profiling of phospholipid species by electrospray ionization mass spectrometry showed that scavenger receptor expression led to the accumulation of phosphatidylcholine species with longer mono- or polyunsaturated acyl chains. These changes would be expected to decrease phosphatidylcholine/cholesterol interactions and thereby enhance cholesterol desorption from the membrane. Scavenger receptor-mediated changes in membrane phosphatidylcholine may contribute to the increased flux of cholesterol and other lipids elicited by these receptors.


Subject(s)
CD36 Antigens/physiology , Cell Membrane/metabolism , Phosphatidylcholines/analysis , Receptors, Immunologic/physiology , Animals , CD36 Antigens/genetics , Cell Line , Cell Membrane/chemistry , Cholesterol/metabolism , Cholesterol Esters/metabolism , Cholesterol Oxidase/metabolism , Insecta , Lipoproteins, HDL/metabolism , Mass Spectrometry , Mice , Phosphatidylcholines/metabolism , Phospholipids/analysis , Rats , Receptors, Immunologic/genetics , Receptors, Scavenger , Scavenger Receptors, Class B , Transfection
15.
DNA Repair (Amst) ; 3(3): 313-22, 2004 Mar 04.
Article in English | MEDLINE | ID: mdl-15177046

ABSTRACT

Thymine glycols (Tg) are major pyrimidine oxidation products produced by chemical agents and ionizing radiation. Recent improvements in purification procedures gave us the opportunity to examine the incision of DNA duplexes containing a single (5S,6R)- or (5R,6S)-Tg lesion by mouse NTH1 DNA glycosylase and mammalian cell nuclear extracts. Time course experiments and steady state enzyme kinetics indicated that mNTH1 discriminates between the cis-Tg isomers. In addition, a variety of mammalian cell nuclear extracts showed a similar discrimination between the cis-Tg isomers. Trapping of Schiff base intermediates with sodium borohydride demonstrated that a single protein-DNA complex was formed in the presence of the nuclear extracts. The electrophoretic mobility of trapped complexes formed with both Tg isomers was identical to one another and similar to that of the complex formed with recombinant mNTH1. These results suggest that among all Tg-active DNA glycosylases, NTH1 is the major enzyme in mammalian cell nuclear extracts responsible for incision of duplexes containing cis-Tg isomers.


Subject(s)
Cell Extracts/pharmacology , Cell Nucleus/chemistry , DNA Repair/drug effects , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , Thymine/analogs & derivatives , Thymine/metabolism , Animals , Cross-Linking Reagents/metabolism , DNA Glycosylases/metabolism , Deoxyribonuclease (Pyrimidine Dimer)/isolation & purification , Electrophoresis, Polyacrylamide Gel , Kinetics , Mice , Oligonucleotides/metabolism , Schiff Bases/metabolism , Stereoisomerism , Tumor Cells, Cultured
16.
Nucleic Acids Res ; 32(1): 338-45, 2004.
Article in English | MEDLINE | ID: mdl-14726482

ABSTRACT

DNA damage created by reactive oxygen species includes the prototypic oxidized pyrimidine, thymine glycol (Tg), which exists in oxidatively damaged DNA as two diastereoisomeric pairs. In Escherichia coli, Saccharomyces cerevesiae and mice, Tg is preferentially excised by endonuclease III (Endo III) and endonuclease VIII (Endo VIII), yNTG1 and yNTG2, and mNTH and mNEIL1, respectively. We have explored the ability of these DNA glycosylases to discriminate between Tg stereoisomers. Oligonucleotides containing a single, chromatographically pure (5S,6R) or (5R,6S) stereoisomer of Tg were prepared by oxidation with osmium tetroxide. Steady-state kinetic analyses of the excision process revealed that Endo III, Endo VIII, yNTG1, mNTH and mNEIL1, but not yNTG2, excise Tg isomers from DNA in a stereoselective manner, as reflected in the parameter of catalytic efficiency (kcat/Km). When DNA glycosylases occur as complementary pairs, failure of one or both enzymes to excise their cognate Tg stereoisomer from oxidatively damaged DNA could have deleterious consequences for the cell.


Subject(s)
DNA Damage , DNA Glycosylases/metabolism , DNA Repair , DNA/chemistry , DNA/metabolism , Thymine/analogs & derivatives , Thymine/chemistry , Thymine/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , Escherichia coli Proteins/metabolism , Kinetics , N-Glycosyl Hydrolases/metabolism , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Oxidation-Reduction , Oxidative Stress , Saccharomyces cerevisiae Proteins/metabolism , Stereoisomerism , Substrate Specificity
17.
Chem Res Toxicol ; 16(6): 708-14, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12807353

ABSTRACT

Chloroethylene oxide and chloroacetaldehyde, reactive intermediates derived from vinyl chloride, and the epoxy-hydroxy-alkanals, produced endogenously in the metabolism of polyunsaturated fatty acids, react with nucleic acid bases in DNA to form exocyclic etheno derivatives of 2'-deoxyadenosine, 2'-deoxyguanosine, and 2'-deoxycytidine. This paper describes an efficient method for the synthesis of the exocyclic 1,N(2)-etheno adduct of 2'-deoxyguanosine and its incorporation into DNA oligomers using automated synthesis techniques. The synthesis was initiated by a high-yield alkylation of N(2)-protected 2'-deoxyguanosine at the 1-position with 1,2-diacetoxy-3-bromopropane. The product was converted to the 5'-O-dimethoxytrityl-3'-O-phosphoramidite using published techniques and incorporated site specifically into DNA oligomers with 99% coupling efficiency. Ring closure to yield the 6-hydroxyethano derivative was accomplished by oxidation with sodium periodate, and facile dehydration then afforded DNA oligomers containing 1,N(2)-etheno-2'-deoxyguanosine. All oligomers were characterized fully by physicochemical methods.


Subject(s)
DNA Adducts/chemistry , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/chemical synthesis , Oligonucleotides/chemical synthesis , Deoxyguanosine/chemistry , Molecular Structure , Oligonucleotides/chemistry
18.
Bioorg Chem ; 31(2): 136-48, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12729571

ABSTRACT

Acrolein, a known mutagen, undergoes reaction in vitro under physiological conditions with both 2(')-deoxyguanosine and native DNA to give rise to exocyclic adducts of the 5,6,7,8-tetrahydropyrimido[1,2-a]purine-10(3H)-one class having an hydroxy group at either the 6 or the 8 position. Previously we have shown that the 8-hydroxy derivative in a bacterial system has very low mutagenicity probably because in double-stranded DNA this residue exists in the open-chain aldehydic form [N(2)-(3-oxopropyl)-2(')-deoxyguanosine] (3). To continue our investigation in this area, we needed ample supplies of the 6-hydroxy isomers. This current paper describes high-yield simple methods for the synthesis in bulk of the 6-hydroxy adduct 1 and its incorporation into DNA oligomers. The basic methods for the synthesis of the adduct 1, involve 1-substitution of dG derivatives with a 3-butenyl group, dihydroxylation of the olefin with osmium tetroxide and N-methylmorpholine N-oxide, then diol cleavage with periodate ion after incorporation of the 1-(3,4-diacetoxybutyl)-2(')-deoxyguanosine into oligomeric DNA.


Subject(s)
Acrolein/chemical synthesis , DNA Adducts/chemistry , Deoxyguanosine/chemical synthesis , Oligonucleotides/chemical synthesis , Acrolein/chemistry , Chromatography, High Pressure Liquid , Deoxyguanosine/chemistry , Oligonucleotides/chemistry
19.
Chem Res Toxicol ; 15(12): 1489-94, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12482230

ABSTRACT

A new and efficient method is described for the synthesis in gram quantities of the benzo[a]pyrene (B[a]P) metabolic adducts of 2'-deoxyguanosine (dG) and 2'-deoxyadenosine (dA) substituted, respectively, at the N(2)- and N(6)- positions. When the racemic form of the tris(benzoyloxy)amine 5 (related to the notoriously carcinogenic epoxydiol 2) is coupled with the bromoinosine derivative 6 by means of a Buchwald-Hartwig reaction, the expected pair of diastereomers, 7 and 8, is obtained in high (combined) yield. Selective deblocking of this mixture then gave cleanly the pair of diastereomers 9. These were used in the synthesis of a series of DNA oligomers via their 5'-O-DMT-3'-O-phosphoramidites (10) using standard automated methods. Coupling efficiencies were 94-98% at the point of introduction of the xeno-2'-deoxynucleoside, and in all cases the mixtures of the two diastereomeric oligomers (DMT-off stage) were easily separated by HPLC. By a similar sequence of reactions beginning with 5 and the protected 6-bromopurine 2'-deoxynucleoside 11, it was possible with equal efficiency to introduce the N(6)-modified diastereomers (16) of dA into oligomeric DNA. Circular dichroism measurements were used to establish the fundamental configurations at the xeno-2'-deoxynucleoside site for each of the oligomers. Mass spectral data in both the dG and the dA series confirmed the presence of the xeno-2'-deoxynucleoside in the oligomers. This was complemented by enzymatic degradation of one of the oligomers from each of the series. In both of these cases, after HPLC separation, circular dichroism measurements on the reisolated xenonucleoside also confirmed its presence in the oligomer.


Subject(s)
Benzo(a)pyrene/chemistry , DNA Adducts/chemical synthesis , DNA/chemistry , Deoxyadenosines/chemistry , Deoxyguanosine/chemistry , Oligodeoxyribonucleotides/chemistry , Alkaline Phosphatase/metabolism , Animals , Base Sequence , Chromatography, High Pressure Liquid/methods , Circular Dichroism , DNA/chemical synthesis , DNA/metabolism , DNA Adducts/chemistry , Mass Spectrometry/methods , Oligodeoxyribonucleotides/chemical synthesis , Oligodeoxyribonucleotides/metabolism , Phosphodiesterase I , Phosphoric Diester Hydrolases/metabolism , Snakes , Stereoisomerism , Venoms
20.
Circulation ; 105(9): 1037-43, 2002 Mar 05.
Article in English | MEDLINE | ID: mdl-11877351

ABSTRACT

BACKGROUND: It has been established that hyperhomocyst(e)inemia (HHCy) is an independent and graded risk factor for atherosclerosis, although the molecular link to the atherosclerotic process remains obscure. METHODS AND RESULTS: Screening human umbilical vein endothelial cells (HUVECs) with complementary DNA microarray for the gene expression modified by homocysteine (Hcy) revealed that 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) was upregulated. This effect was confirmed using quantitative reverse transcriptase-polymerase chain reaction. Actinomycin D studies revealed that Hcy stabilized HMGCR mRNA (tau(1/2), 9.5 +/- 1.0 versus 5.0 +/- 0.2 hours). Expression of immunodetectable HMGCR in both HUVECs and renal microvascular endothelial cells was increased in Hcy-treated cells in association with the increased abundance of caveolin. Application of a cell-permeable superoxide dismutase mimetic, Mn-TBAP, reversed the Hcy-induced expression of HMGCR. Additional biochemical analysis of the abundance of total cellular cholesterol showed that 0, 20, 50, and 100 micromol/L Hcy resulted in 22.2 +/- 7.3%, 39.5 +/- 1.2%, and 50.4 +/- 6.8% increase, respectively. Gas chromatography mass spectrometry analysis of extracted cholesterol from Hcy-treated HUVECs and from the culture medium showed 17.8 +/- 5.2% and 24.0 +/- 14.5% increases, respectively. Application of simvastatin to Hcy-treated cells reduced cellular cholesterol and prevented Hcy-induced suppression of NO production by HUVECs in a dose-dependent manner. CONCLUSIONS: Using a cDNA microarray, the data disclosed an unexpected link between Hcy and cholesterol dysregulation based on the finding of increased abundance of HMGCR mRNA and protein in endothelial cells, demonstrated the possible role of Hcy-induced oxidative stress in this response, and revealed the improvement of endothelial NO production in Hcy-treated HUVECs by statins. Collectively, these findings may provide a solid explanation for the observed proatherogenic effect of HHcy.


Subject(s)
Arteriosclerosis/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Homocysteine/pharmacology , Hydroxymethylglutaryl CoA Reductases/metabolism , Arteriosclerosis/etiology , Caveolin 1 , Caveolins/metabolism , Cells, Cultured , Cholesterol/metabolism , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Enzyme Induction/drug effects , Free Radical Scavengers/pharmacology , Gene Expression/drug effects , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kidney/blood supply , Kidney/cytology , Kidney/enzymology , Metalloporphyrins/pharmacology , Nitric Oxide/metabolism , Oligonucleotide Array Sequence Analysis , RNA Stability/drug effects , RNA, Messenger/metabolism , Risk Factors , Simvastatin/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...