Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 313
Filter
1.
Biochemistry ; 53(35): 5683-91, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25148552

ABSTRACT

DNA lesion bypass polymerases process different lesions with varying fidelities, but the structural, dynamic, and mechanistic origins of this phenomenon remain poorly understood. Human DNA polymerase κ (Polκ), a member of the Y family of lesion bypass polymerases, is specialized to bypass bulky DNA minor groove lesions in a predominantly error-free manner, by housing them in its unique gap. We have investigated the role of the unique Polκ gap and N-clasp structural features in the fidelity of minor groove lesion processing with extensive molecular modeling and molecular dynamics simulations to pinpoint their functioning in lesion bypass. Here we consider the N(2)-dG covalent adduct derived from the carcinogenic aromatic amine, 2-acetylaminofluorene (dG-N(2)-AAF), that is produced via the combustion of kerosene and diesel fuel. Our simulations reveal how the spacious gap directionally accommodates the lesion aromatic ring system as it transits through the stages of incorporation of the predominant correct partner dCTP opposite the damaged guanine, with preservation of local active site organization for nucleotidyl transfer. Furthermore, flexibility in Polκ's N-clasp facilitates the significant misincorporation of dTTP opposite dG-N(2)-AAF via wobble pairing. Notably, we show that N-clasp flexibility depends on lesion topology, being markedly reduced in the case of the benzo[a]pyrene-derived major adduct to N(2)-dG, whose bypass by Polκ is nearly error-free. Thus, our studies reveal how Polκ's unique structural and dynamic properties can regulate its bypass fidelity of polycyclic aromatic lesions and how the fidelity is impacted by lesion structures.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , 2-Acetylaminofluorene/analogs & derivatives , 2-Acetylaminofluorene/chemistry , 2-Acetylaminofluorene/metabolism , Base Pair Mismatch , Catalytic Domain , DNA Adducts/chemistry , DNA Adducts/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/chemistry , Deoxyguanosine/metabolism , Humans , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation , Substrate Specificity
2.
Nucleic Acids Res ; 41(16): 7843-53, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23814187

ABSTRACT

DNA polymerases must accurately replicate DNA to maintain genome integrity. Carcinogenic adducts, such as 2-aminofluorene (AF) and N-acetyl-2-aminofluorene (AAF), covalently bind DNA bases and promote mutagenesis near the adduct site. The mechanism by which carcinogenic adducts inhibit DNA synthesis and cause mutagenesis remains unclear. Here, we measure interactions between a DNA polymerase and carcinogenic DNA adducts in real-time by single-molecule fluorescence. We find the degree to which an adduct affects polymerase binding to the DNA depends on the adduct location with respect to the primer terminus, the adduct structure and the nucleotides present in the solution. Not only do the adducts influence the polymerase dwell time on the DNA but also its binding position and orientation. Finally, we have directly observed an adduct- and mismatch-induced intermediate state, which may be an obligatory step in the DNA polymerase proofreading mechanism.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , Carcinogens/chemistry , DNA Adducts/chemistry , DNA Polymerase I/metabolism , Deoxyguanosine/analogs & derivatives , Fluorenes/chemistry , 2-Acetylaminofluorene/chemistry , DNA/biosynthesis , DNA/chemistry , DNA Polymerase I/chemistry , Deoxyguanosine/chemistry , Fluorometry/methods , Protein Binding
3.
Exp Toxicol Pathol ; 65(4): 451-6, 2013 May.
Article in English | MEDLINE | ID: mdl-22459205

ABSTRACT

Human liver cancer is in part associated with obesity and related metabolic diseases. The present study was undertaken in a mouse model of diet-induced obesity (DIO) and hepatic steatosis, conditions which can be associated with hepatic neoplasia, to determine whether the rates of cell proliferation or hepatocarcinogen bioactivation were altered in ways which could facilitate hepatocarcinogenesis. DIO mice were generated by feeding C57BL/6 (B6) male mice a high-fat diet beginning at 4 weeks of age; age-matched conventional lean (LEAN) B6 mice fed a low fat diet (10% Kcal from fat) were used for comparison. Groups of 28 week old DIO and LEAN mice were dosed with the bioactivation-dependent DNA-reactive hepatocarcinogen 2-acetylaminofluorene (AAF), at 2.24 or 22.4 mg/kg, given by gavage 3 times per week for 31 days, or received no treatment (DIO and LEAN control groups). Compared with the LEAN control group, the DIO control group had a higher mean body weight (16.5 g), higher mean absolute (1.4 g) and mean relative (25.5%) liver weights, higher (394%) liver triglyceride concentrations, and an increased incidence and severity of hepatocellular steatosis at the end of the dosing phase. The DIO control group also had a higher mean hepatocellular replicating fraction (31% increase, determined by proliferating cell nuclear antigen immunohistochemistry). Hepatocarcinogen bioactivation, based on formation of AAF DNA adducts as measured by nucleotide (32)P-postlabeling, was similar in both DIO and LEAN AAF-dosed groups. Thus, hepatocellular proliferation, but not hepatocarcinogen bioactivation, was identified as an alteration in livers of DIO mice which could contribute to their susceptibility to hepatocarcinogenesis.


Subject(s)
Cell Proliferation/drug effects , Fatty Liver/physiopathology , Hepatocytes/drug effects , Obesity/complications , 2-Acetylaminofluorene/analogs & derivatives , 2-Acetylaminofluorene/toxicity , Animal Feed , Animals , Carcinogens/toxicity , DNA Adducts/analysis , DNA Adducts/biosynthesis , Diet, Fat-Restricted , Diet, High-Fat/adverse effects , Disease Models, Animal , Mice , Mice, Inbred C57BL , Obesity/physiopathology
4.
Nucleic Acids Res ; 40(19): 9675-90, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22904073

ABSTRACT

Nucleotide excision repair (NER) efficiencies of DNA lesions can vary by orders of magnitude, for reasons that remain unclear. An example is the pair of N-(2'-deoxyguanosin-8-yl)-2-aminofluorene (dG-C8-AF) and N-(2'-deoxyguanosin-8-yl)-2-acetylaminofluorene (dG-C8-AAF) adducts that differ by a single acetyl group. The NER efficiencies in human HeLa cell extracts of these lesions are significantly different when placed at G(1), G(2) or G(3) in the duplex sequence (5'-CTCG(1)G(2)CG(3)CCATC-3') containing the NarI mutational hot spot. Furthermore, the dG-C8-AAF adduct is a better substrate of NER than dG-C8-AF in all three NarI sequence contexts. The conformations of each of these adducts were investigated by Molecular dynamics (MD) simulation methods. In the base-displaced conformational family, the greater repair susceptibility of dG-C8-AAF in all sequences stems from steric hindrance effects of the acetyl group which significantly diminish the adduct-base stabilizing van der Waals stacking interactions relative to the dG-C8-AF case. Base sequence context effects for each adduct are caused by differences in helix untwisting and minor groove opening that are derived from the differences in stacking patterns. Overall, the greater NER efficiencies are correlated with greater extents of base sequence-dependent local untwisting and minor groove opening together with weaker stacking interactions.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , DNA Adducts/chemistry , DNA Repair , Deoxyguanosine/analogs & derivatives , Fluorenes/chemistry , 2-Acetylaminofluorene/chemistry , 2-Acetylaminofluorene/metabolism , Base Sequence , DNA Adducts/metabolism , Deoxyguanosine/chemistry , Deoxyguanosine/metabolism , Fluorenes/metabolism , HeLa Cells , Humans , Molecular Dynamics Simulation , Nucleic Acid Conformation
5.
J Am Chem Soc ; 133(32): 12600-7, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21751809

ABSTRACT

Water-soluble conjugated polymers with controlled molecular weight characteristics, absence of ionic groups, high emission quantum yields, and end groups capable of selective reactions of wide scope are desirable for improving their performance in various applications and, in particular, fluorescent biosensor schemes. The synthesis of such a structure is described herein. 2-Bromo-7-iodofluorene with octakis(ethylene glycol) monomethyl ether chains at the 9,9'-positions, i.e., compound 4, was prepared as the reactive premonomer. A high-yielding synthesis of the organometallic initiator (dppe)Ni(Ph)Br (dppe = 1,2-bis(diphenylphosphino)ethane) was designed and implemented, and the resulting product was characterized by single-crystal X-ray diffraction techniques. Polymerization of 4 by (dppe)Ni(Ph)Br can be carried out in less than 30 s, affording excellent control over the average molecular weight and polydispersity of the product. Quenching of the polymerization with [2-(trimethylsilyl)ethynyl]magnesium bromide yields silylacetylene-terminated water-soluble poly(fluorene) with a photoluminescence quantum efficiency of 80%. Desilylation, followed by copper-catalyzed azide-alkyne cycloaddition reaction, yields a straightforward route to introduce a wide range of specific end group functionalities. Biotin was used as an example. The resulting biotinylated conjugated polymer binds to streptavidin and acts as a light-harvesting chromophore to optically amplify the emission of Alexa Fluor-488 chromophores bound onto the streptavidin. Furthermore, the biotin end group makes it possible to bind the polymer onto streptavidin-functionalized cross-linked agarose beads and thereby incorporate a large number of optically active segments.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , Biosensing Techniques/methods , Fluorescent Dyes/chemistry , Polymers/chemistry , 2-Acetylaminofluorene/chemical synthesis , 2-Acetylaminofluorene/chemistry , 2-Acetylaminofluorene/metabolism , Biotinylation , Click Chemistry , Ethylene Glycol/chemical synthesis , Ethylene Glycol/chemistry , Ethylene Glycol/metabolism , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Luminescence , Models, Molecular , Polymerization , Polymers/chemical synthesis , Polymers/metabolism , Solubility , Streptavidin/metabolism , Water/chemistry
7.
Biochemistry ; 48(1): 7-18, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19072536

ABSTRACT

Bypass across DNA lesions by specialized polymerases is essential for maintenance of genomic stability. Human DNA polymerase iota (poliota) is a bypass polymerase of the Y family. Crystal structures of poliota suggest that Hoogsteen base pairing is employed to bypass minor groove DNA lesions, placing them on the spacious major groove side of the enzyme. Primer extension studies have shown that poliota is also capable of error-free nucleotide incorporation opposite the bulky major groove adduct N-(deoxyguanosin-8-yl)-2-acetylaminofluorene (dG-AAF). We present molecular dynamics simulations and free energy calculations suggesting that Watson-Crick base pairing could be employed in poliota for bypass of dG-AAF. In poliota with Hoogsteen-paired dG-AAF the bulky AAF moiety would reside on the cramped minor groove side of the template. The Hoogsteen-capable conformation distorts the active site, disrupting interactions necessary for error-free incorporation of dC opposite the lesion. Watson-Crick pairing places the AAF rings on the spacious major groove side, similar to the position of minor groove adducts observed with Hoogsteen pairing. Watson-Crick-paired structures show a well-ordered active site, with a near reaction-ready ternary complex. Thus our results suggest that poliota would utilize the same spacious region for lesion bypass of both major and minor groove adducts. Therefore, purine adducts with bulk on the minor groove side would use Hoogsteen pairing, while adducts with the bulky lesion on the major groove side would utilize Watson-Crick base pairing as indicated by our MD simulations for dG-AAF. This suggests the possibility of an expanded role for poliota in lesion bypass.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , Base Pairing , DNA Adducts/chemistry , DNA-Directed DNA Polymerase/chemistry , Deoxyguanosine/analogs & derivatives , 2-Acetylaminofluorene/chemistry , Catalytic Domain , Deoxyguanosine/chemistry , Humans , Models, Molecular , Molecular Structure , Thermodynamics , DNA Polymerase iota
8.
Histochem Cell Biol ; 130(5): 1005-14, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18663461

ABSTRACT

The 2-acetaminofluorene/partial hepatectomy (AAF/Phx) model is widely used to induce oval/progenitor cell proliferation in the rat liver. We have used this model to study the impact of a primary hepatocyte mitogen, triiodothyronine (T3) on the liver regenerating by the recruitment of oval/progenitor cells. Administration of T3 transiently accelerates the proliferation of the oval cells, which is followed by rapid differentiation into small hepatocytes. The oval cell origin of the small hepatocytes has been proven by tracing retrovirally transduced and BrdU marked oval cells. The differentiating oval cells become positive for hepatocyte nuclear factor-4 and start to express hepatocyte specific connexin 32, alpha1 integrin, Prox1, cytochrom P450s, and form CD 26 positive bile canaliculi. At the same time oval cell specific OV-6 and alpha-fetoprotein expression is lost. The upregulation of hepatocyte specific mRNAs: albumin, tyrosine aminotransferase and tryptophan 2,3-dioxygenase detected by real-time PCR also proves hepatocytic maturation. The hepatocytic conversion of oval cells occurs on the seventh day after the Phx in this model while the first small hepatocytes appear 5 days later without T3 treatment. The administration of the primary hepatocyte mitogen T3 accelerates the differentiation of hepatic progenitor cells into hepatocytes in vivo, and that may have therapeutic potential.


Subject(s)
Cell Differentiation/drug effects , Hepatocytes/drug effects , Liver Regeneration/drug effects , Liver/drug effects , Stem Cells/drug effects , Triiodothyronine/pharmacology , 2-Acetylaminofluorene/analogs & derivatives , Animals , Cell Lineage , Cell Proliferation/drug effects , Cell Shape/drug effects , Gene Expression Regulation/drug effects , Hepatectomy , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/metabolism , Liver/pathology , Liver/surgery , Male , Models, Animal , Rats , Rats, Inbred F344 , Stem Cells/metabolism , Stem Cells/pathology , Time Factors
9.
Eur J Cancer Prev ; 16(6): 528-34, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18090125

ABSTRACT

Acetaminophen, a monocyclic phenolic compound and analgesic, when fed at 8900 p.p.m. in the diet, was reported to inhibit the hepatocarcinogenicity in rats of the aromatic amine proximate carcinogen N-hydroxy-N-acetyl-2-aminofluorene. To elucidate the mechanism(s) of this anticarcinogenicity, the present study examined whether acetaminophen at lower doses has the ability to inhibit the initiating effects in the rat liver of the precursor hepatocarcinogen N-acetyl-2-aminofluorene. Male F344 rats were allocated to six groups, which were maintained under reverse light cycle conditions to assure acetaminophen ingestion at the time of N-acetyl-2-aminofluorene administration during the dark phase, which was imposed from 07.00 to 19.00 h. Group 1 served as vehicle control (0.5% carboxymethylcellulose) for N-acetyl-2-aminofluorene, which was administered intragastrically 3 days per week at 2.6 mg/kg for 8 weeks (group 4) to achieve initiation. Acetaminophen was given in the diet either alone at 2400 or 4800 p.p.m. for 9 weeks (groups 2 and 3), or with N-acetyl-2-aminofluorene (groups 5 and 6), starting 1 week before N-acetyl-2-aminofluorene administration. Acetaminophen blood levels were about 1 and 4 microg/ml at the two dietary concentrations. N-acetyl-2-aminofluorene induced hepatocellular preneoplastic lesions measured as hepatocellular altered foci expressing glutathione S-transferase-P, reflecting initiation. Induced foci were reduced with administration of both concentrations of acetaminophen. Acetaminophen by itself produced no DNA adducts nor did it alter the high formation of N-acetyl-2-aminofluorene-DNA adducts, about 200 in 10 nucleotides, measured by nucleotide postlabeling. Acetaminophen did not affect background liver cell proliferation, but significantly reduced N-acetyl-2-aminofluorene-induced increased proliferation measured by proliferating cell nuclear antigen immunostaining. Thus, acetaminophen effectively protected hepatocytes from the initiating effects of N-acetyl-2-aminofluorene, possibly through a cytoprotective effect resulting from slowing the rate of induced cell turnover.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , Acetaminophen/pharmacology , Acetaminophen/therapeutic use , Carcinoma, Hepatocellular/prevention & control , DNA Adducts/drug effects , Liver Neoplasms/prevention & control , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Body Weight/drug effects , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/drug effects , DNA Adducts/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Liver/drug effects , Liver/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Male , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , Rats , Rats, Inbred F344
10.
Biochemistry ; 46(46): 13310-21, 2007 Nov 20.
Article in English | MEDLINE | ID: mdl-17960913

ABSTRACT

The NarI recognition sequence (5'-G1G2CG3CN-3') is the most vulnerable hot spot for frameshift mutagenesis induced by the carcinogen 2-aminofluorene and its analogues in Escherichia coli. Lesioning of the guanine in the G3 position induces an especially high frequency of -2 deletion mutations; vulnerability to these mutations is modulated by the nature of the nucleotide in the N position (C approximately A > G > T). The objective of the present study was to probe the structural basis of this N-mediated influence on the propensity of the G3 lesion to form a slipped mutagenic intermediate (SMI) during translesion synthesis. We studied NarI-based fully paired [(5'-CTCG1G2CG3*CNATC-3')(5'-GATNCGGCCGAG-3'), N = dC or dT] and -2 deletion [(5'-CTCG1G2CG3*CNATC-3')(5'-GATNGCCGAG-3'), N = dC or dT] duplexes, in which G* was either AF [N-(2'-deoxyguanosin-8-yl)-2-aminofluorene] or the 19F probe FAF [N-(2'-deoxyguanosin-8-yl)-7-fluoro-2-aminofluorene]. The latter sequences mimic the bulged SMI for -2 deletion mutations. Dynamic 19F NMR, circular dichroism, and UV melting results indicated that the NarI-dC/-2 deletion duplex adopts exclusively an intercalated conformer, whereas the NarI-dT/-2 deletion duplex exists as multiple conformers. The data support the presence of a putative equilibrium between a carcinogen-intercalated and a carcinogen-exposed SMI for the dT/-2 duplex. A similar dT-induced conformational heterogeneity was observed for the fully paired duplexes in which all three guanines were individually modified by AF or FAF. The frequency of the carcinogen stacked S-conformation was found to be highest (69-75%) at the G3 hot spot in NarI-dC duplexes. Taken together, our results support the hypothesis that the conformational stability of the SMI is a critical determinant for the efficacy of -2 frameshift mutagenesis in the NarI sequence. We also provide evidence for AF/FAF conformational compatibility in the NarI sequences.


Subject(s)
DNA Adducts/chemistry , Deoxyribonucleases, Type II Site-Specific/metabolism , Fluorenes/chemistry , Frameshift Mutation , 2-Acetylaminofluorene/analogs & derivatives , 2-Acetylaminofluorene/chemistry , Base Sequence , Circular Dichroism , Fluorine/chemistry , Models, Molecular , Molecular Sequence Data , Mutagenesis , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , Spectrophotometry, Ultraviolet
11.
Biochemistry ; 46(40): 11263-78, 2007 Oct 09.
Article in English | MEDLINE | ID: mdl-17877372

ABSTRACT

A systematic spectroscopic and computational study was conducted in order to probe the influence of base sequences on stacked (S) versus B-type (B) conformational heterogeneity induced by the major dG adduct derived from the model carcinogen 7-fluoro-2-aminofluorene (FAF). We prepared and characterized eight 12-mer DNA duplexes (-AG*N- series, d[CTTCTAG*NCCTC]; -CG*N- series, d[CTTCTCG*NCCTC]), in which the central guanines (G*) were site-specifically modified with FAF with varying flanking bases (N = G, A, C, T). S/B heterogeneity was examined by CD, UV, and dynamic 19F NMR spectroscopy. All the modified duplexes studied followed a typical dynamic exchange between the S and B conformers in a sequence dependent manner. Specifically, purine bases at the 3'-flanking site promoted the S conformation (G > A > C > T). Simulation analysis showed that the S/B energy barriers were in the 14-16 kcal/mol range. The correlation times (tau = 1/kappa) were found to be in the millisecond range at 20 degrees C. The van der Waals energy force field calculations indicated the importance of the stacking interaction between the carcinogen and neighboring base pairs. Quantum mechanics calculations showed the existence of correlations between the total interaction energies (including electrostatic and solvation effects) and the S/B population ratios. The S/B equilibrium seems to modulate the efficiency of Escherichia coli UvrABC-based nucleotide excision repair in a conformation-specific manner: i.e., greater repair susceptibility for the S over B conformation and for the -AG*N- over the -CG*N- series. The results indicate a novel structure-function relationship, which provides insights into how bulky DNA adducts are accommodated by UvrABC proteins.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , DNA Adducts/chemistry , DNA Repair , 2-Acetylaminofluorene/chemistry , Base Sequence , Circular Dichroism , DNA Adducts/genetics , Magnetic Resonance Spectroscopy , Models, Biological , Nucleic Acid Conformation , Spectrophotometry, Ultraviolet , Temperature
12.
Anticancer Res ; 26(2A): 1105-11, 2006.
Article in English | MEDLINE | ID: mdl-16619513

ABSTRACT

The activation of ketoprofen, which inhibits the outgrowth of azoxymethane-induced aberrant crypt foci in the rat colon, on the inhibition of arylamine N-acetyltransferase (NAT) activity (N-acetylation of substrates), gene expression (mRNA NAT) and 2-aminofluorene (AF)-DNA adduct formation was studied in a human colon tumor (adenocarcinoma) cell line (colo 205). Cellular cytosols (9000 xg supernatant) and intact colon tumor cells were used. The NAT activity in colo 205 cells was inhibited by ketoprofen in a dose- and time -dependent manner in both examined systems. The data also indicated that ketoprofen decreased the apparent value of V(max) of NAT enzymes, being a competitive inhibitor of NAT enzymes. The AF-DNA adduct formation in colo 205 cells was also decreased by ketoprofen. Based on the results from PCR, it was shown that ketoprofen affected mRNA NAT expression in human colon colo 205 cells. The cells were stained with anti-NAT antibody, then analyzed by flow cytometry. The results showed that ketoprofen decreased the percentage of cells stained by anti-NAT. This report is the first to demonstrate that ketoprofen inhibits human colon tumor cell NAT activity, gene expression and DNA adduct formation.


Subject(s)
Arylamine N-Acetyltransferase/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Ketoprofen/pharmacology , 2-Acetylaminofluorene/analogs & derivatives , 2-Acetylaminofluorene/metabolism , 4-Aminobenzoic Acid/metabolism , Acetylation/drug effects , Arylamine N-Acetyltransferase/metabolism , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cytosol/enzymology , DNA Adducts/biosynthesis , Dose-Response Relationship, Drug , Fluorenes , Gene Expression/drug effects , Humans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , para-Aminobenzoates
13.
Nucleic Acids Res ; 34(3): 785-95, 2006.
Article in English | MEDLINE | ID: mdl-16452300

ABSTRACT

Primer extension studies have shown that the Y-family DNA polymerase IV (Dpo4) from Sulfolobus solfataricus P2 can preferentially insert C opposite N-(deoxyguanosin-8-yl)-2-acetylaminofluorene (AAF-dG) [F. Boudsocq, S. Iwai, F. Hanaoka and R. Woodgate (2001) Nucleic Acids Res., 29, 4607-4616]. Our goal is to elucidate on a structural level how AAF-dG can be harbored in the Dpo4 active site opposite an incoming dCTP, using molecular modeling and molecular dynamics simulations, since AAF-dG prefers the syn glycosidic torsion. Both anti and syn conformations of the templating AAF-dG in a Dpo4 ternary complex were investigated. All four dNTPs were studied. We found that an anti glycosidic torsion with C1'-exo deoxyribose conformation allows AAF-dG to be Watson-Crick hydrogen-bonded with dCTP with modest polymerase perturbation, but other nucleotides are more distorting. The AAF is situated in the Dpo4 major groove open pocket with fluorenyl rings 3'- and acetyl 5'-directed along the modified strand, irrespective of dNTP. With AAF-dG syn, the fluorenyl rings are in the small minor groove pocket and the active site region is highly distorted. The anti-AAF-dG conformation with C1'-exo sugar pucker can explain the preferential incorporation of dC by Dpo4. Possible relevance of our new major groove structure for AAF-dG to other polymerases, lesion repair and solution conformations are discussed.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , DNA Adducts/chemistry , DNA Polymerase beta/chemistry , Deoxyguanosine/analogs & derivatives , Models, Molecular , Sulfolobus solfataricus/enzymology , 2-Acetylaminofluorene/chemistry , Base Pairing , Binding Sites , DNA Replication , Deoxyadenine Nucleotides/chemistry , Deoxycytosine Nucleotides/chemistry , Deoxyguanosine/chemistry , Hydrogen Bonding , Nucleic Acid Conformation
14.
Biochemistry ; 44(46): 15387-95, 2005 Nov 22.
Article in English | MEDLINE | ID: mdl-16285743

ABSTRACT

N-Acetyl-2-aminofluorene (AAF) is a chemical carcinogen that reacts with guanines at the C8 position in DNA to form a structure that interferes with DNA replication. In bacteria, the NarI restriction enzyme recognition sequence (G1G2CG3CC) is a very strong mutational hot spot when an AAF adduct is positioned at G3 of this sequence, causing predominantly a -2 frameshift GC dinucleotide deletion mutation. In this study, templates were constructed that contained an AAF adduct at this position, and primers of different lengths were prepared such that the primer ended one nucleotide before or opposite or one nucleotide after the adduct site. Primer extension and gel shift binding assays were used to study the mechanism of bypass by the Escherichia coli DNA polymerase I (Klenow fragment) in the presence of these templates. Primer extension in the presence of all four dNTPs produced a fully extended product using the unmodified template, while with the AAF-modified template synthesis initially stalled at the adduct site and subsequent synthesis resulted in a product that contained the GC dinucleotide deletion. Extension product and gel shift binding analyses were consistent with the formation of a two-nucleotide bulge structure upstream of the active site of the polymerase after a nucleotide is incorporated across from the adduct. These data support a model in which the AAF adduct in the NarI sequence specifically induces a structure upstream of the polymerase active site that leads to the GC frameshift mutation and that it is this structure that allows synthesis past the adduct to occur.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , DNA Adducts/metabolism , DNA Polymerase I/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Frameshift Mutation/genetics , 2-Acetylaminofluorene/pharmacology , Amino Acid Sequence , DNA Primers/chemistry , Deoxyribonucleases, Type II Site-Specific/metabolism , Electrophoretic Mobility Shift Assay , Frameshift Mutation/drug effects , Molecular Sequence Data , Templates, Genetic
15.
Hepatology ; 42(4): 863-70, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16175606

ABSTRACT

Cytokeratins are the largest subfamily of intermediate filament proteins and include more than 20 different gene products, which are expressed in an epithelial tissue-specific manner. We studied by immunohistochemistry and confocal microscopy the distribution of cytokeratin subtypes in the biliary system of adult rat liver. A cytokeratin (CK)19+/7- cholangiocyte population was observed in the smaller branches of the biliary tree including the canals of Hering. They proliferated after 2-acetaminofluorene (AAF) administration, although later the typical oval cells expressed CK7. This observation suggests that cholangiocytes with this cytokeratin phenotype may harbor adult hepatic stem cells. The CK19+/7- cholangiocytes were not present in the rat liver at birth, but developed postnatally. Similar cell populations were not observed in human livers. In conclusion, we propose that the CK19+/7- phenotype may be characteristic for adult hepatic stem cells in rat liver and that these cells are generated de novo after birth.


Subject(s)
Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/metabolism , Keratins/metabolism , Liver/cytology , Liver/metabolism , 2-Acetylaminofluorene/analogs & derivatives , 2-Acetylaminofluorene/pharmacology , Age Factors , Animals , Bile Ducts, Intrahepatic/growth & development , Biomarkers/metabolism , Cell Division/drug effects , Humans , Immunohistochemistry , Keratin-7 , Liver/growth & development , Male , Rats , Rats, Inbred F344 , Stem Cells/cytology , Stem Cells/metabolism
16.
Biochemistry ; 44(19): 7361-8, 2005 May 17.
Article in English | MEDLINE | ID: mdl-15882075

ABSTRACT

Human xeroderma pigmentosum group A (XPA) is an essential protein for nucleotide excision repair (NER). We have previously reported that XPA forms a homodimer in the absence of DNA. However, what oligomeric forms of XPA are involved in DNA damage recognition and how the interaction occurs in terms of biochemical understanding remain unclear. Using the homogeneous XPA protein purified from baculovirus-infected Sf21 insect cells and the methods of gel mobility shift assays, gel filtration chromatography, and UV-cross-linking, we demonstrated that both monomeric and dimeric XPA bound to the DNA adduct of N-acetyl-2-aminofluorene (AAF), while showing little affinity for nondamaged DNA. The binding occurred in a sequential and protein concentration-dependent manner. At relatively low-protein concentrations, XPA formed a complex with DNA adduct as a monomer, while at the higher concentrations, an XPA dimer was involved in the specific binding. Results from fluorescence spectroscopic and competitive binding analyses indicated that the specific binding of XPA to the adduct was significantly facilitated and stabilized by the presence of the second XPA in a positive cooperative manner. This cooperative binding exhibited a Hill coefficient of 1.9 and the step binding constants of K(1) = 1.4 x 10(6) M(-)(1) and K(2) = 1.8 x 10(7) M(-)(1). When interaction of XPA and RPA with DNA was studied, even though binding of RPA-XPA complex to adducted DNA was observed, the presence of RPA had little effect on the overall binding efficiency. Our results suggest that the dominant form for XPA to efficiently bind to DNA damage is the XPA dimer. We hypothesized that the concentration-dependent formation of different types of XPA-damaged DNA complex may play a role in cellular regulation of XPA activity.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , DNA Damage , DNA-Binding Proteins/metabolism , 2-Acetylaminofluorene/metabolism , 2-Acetylaminofluorene/radiation effects , Binding, Competitive/genetics , Chromatography, Gel , Cross-Linking Reagents , DNA Adducts/genetics , DNA Adducts/metabolism , DNA Adducts/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA Replication , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/radiation effects , Dimerization , Fluorescence Polarization , Humans , Protein Binding/genetics , Protein Binding/radiation effects , Replication Protein A , Scintillation Counting , Ultraviolet Rays , Xeroderma Pigmentosum Group A Protein
17.
Nucleic Acids Res ; 33(6): 1961-9, 2005.
Article in English | MEDLINE | ID: mdl-15814813

ABSTRACT

Aromatic amino and nitro compounds are potent carcinogens found in the environment that exert their toxic effects by reacting with DNA following metabolic activation. One important adduct is N-(deoxyguanosin-8-yl)-2-acetylaminofluorene (dG-AAF), which has been extensively used in studies of the mechanisms of DNA repair and mutagenesis. Despite the importance of dG-AAF adducts in DNA, an efficient method for its incorporation into DNA using solid-phase synthesis is still missing. We report the development of a modified 'ultra-mild' DNA synthesis protocol that allows the incorporation of dG-AAF into oligonucleotides of any length accessible by solid-phase DNA synthesis with high efficiency and independent of sequence context. Key to this endeavor was the development of improved deprotection conditions (10% diisopropylamine in methanol supplemented with 0.25 M of beta-mercaptoethanol) designed to remove protecting groups of commercially available 'ultra-mild' phosphoramidite building blocks without compromising the integrity of the exquisitely base-labile acetyl group at N8 of dG-AAF. We demonstrate the suitability of these oligonucleotides in the nucleotide excision repair reaction. Our synthetic approach should facilitate comprehensive studies of the mechanisms of repair and mutagenesis induced by dG-AAF adducts in DNA and should be of general use for the incorporation of base-labile functionalities into DNA.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , 2-Acetylaminofluorene/chemistry , DNA Adducts/chemical synthesis , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/chemistry , Oligodeoxyribonucleotides/chemical synthesis , Biochemistry/methods , DNA Adducts/chemistry , DNA Repair , Fluorenes/chemistry , HeLa Cells , Humans , Oligodeoxyribonucleotides/chemistry
18.
Anticancer Res ; 25(1A): 127-32, 2005.
Article in English | MEDLINE | ID: mdl-15816529

ABSTRACT

It is well documented that arylamine carcinogens are N-acetylated by cytosolic N-acetyltransferase (NAT) enzyme. NAT plays an important role in the metabolizing of those arylamine compounds. 2-Aminofluorene (AF) is an arylamine carcinogen which has been demonstrated to induce carcinogenesis in laboratory animals. Our previous study has shown that a human promyelocytic leukemia cell line, HL-60, displays NAT activity. The purpose of the present study was to determine whether or not wogonin could affect the N-acetylation of AF in HL-60. N-acetylated and non-N-acetylated AF were determined by using high performance liquid chromatography. Wogonin displayed a dose-dependent inhibition of NAT activity in cytosols and intact cells. Wogonin also decreased AF-DNA adduct formation in these cells. The effects of wogonin on the NAT enzymes levels were also examined by Western blotting and flow cytometry and the changes of NAT gene expression were examined by polymerase chain reaction (PCR) and cDNA microarray. The results demonstrated that wogonin inhibited NAT1 mRNA gene expression and the level of NAT enzyme in HL-60 cells. This is the first demonstration that wogonin affects human leukemia cells' NAT activity in vitro.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , Acetyltransferases/antagonists & inhibitors , Flavanones/pharmacology , 2-Acetylaminofluorene/metabolism , Acetylation/drug effects , Acetyltransferases/genetics , Acetyltransferases/metabolism , Cytosol/drug effects , Cytosol/metabolism , DNA Adducts/biosynthesis , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Fluorenes/metabolism , Gene Expression/drug effects , HL-60 Cells , Humans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
19.
Biochemistry ; 43(47): 15005-13, 2004 Nov 30.
Article in English | MEDLINE | ID: mdl-15554708

ABSTRACT

The carcinogen 2-acetylaminofluorene is metabolically activated in cells and reacts with DNA to form N-(deoxyguanosin-8-yl)-2-acetylaminofluorene (dG-C8-AAF), N-(deoxyguanosin-8-yl)-2-aminofluorene (dG-C8-AF), and 3-(deoxyguanosin-N(2)()-yl)-2-acetylaminofluorene (dG-N(2)-AAF) DNA adducts. The dG-N(2)-AAF adduct is the least abundant of the three isomers, but it persists in the tissues of animals treated with this carcinogen. The miscoding and mutagenic properties of dG-C8-AAF and dG-C8-AF have been established; these adducts are readily excised by DNA repair enzymes engaged in nucleotide excision repair. In the present study, oligodeoxynucleotides modified site-specifically with dG-N(2)-AAF were used as DNA templates in primer extension reactions catalyzed by mammalian DNA polymerases. Reactions catalyzed by pol alpha were strongly blocked at a position one base before dG-N(2)-AAF and also opposite this lesion. In contrast, during translesion synthesis catalyzed by pol eta or pol kappa nucleotides were incorporated opposite the lesion. Both pol eta and pol kappa incorporated dCMP, the correct base, opposite dG-N(2)-AAF. In reactions catalyzed by pol eta, small amounts of dAMP misincorporation and one-base deletions were detected at the lesion site. With pol kappa, significant dTMP misincorporation was observed opposite the lesion. Steady-state kinetic analysis confirmed the results obtained from primer extension studies. Single-stranded shuttle vectors containing (5)(')TCCTCCTCXCCTCTC (X = dG-N(2)-AAF, dG-C8-AAF, or dG) were used to establish the frequency and specificity of dG-N(2)-AAF-induced mutations in simian kidney (COS-7) cells. Both lesions promote G --> T transversions overall, with dG-N(2)-AAF being less mutagenic than dG-C8-AAF (3.4% vs 12.5%). We conclude from this study that dG-N(2)-AAF, by virtue of its persistence in tissues, contributes significantly to the mutational spectra observed in AAF-induced mutagenesis and that pol eta, but not pol kappa, may play a role in this process.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , 2-Acetylaminofluorene/toxicity , DNA Adducts/toxicity , Deoxyguanosine/analogs & derivatives , Mutagens/toxicity , 2-Acetylaminofluorene/chemistry , Animals , Base Pair Mismatch , Base Sequence , COS Cells , Chlorocebus aethiops , DNA Adducts/chemistry , DNA Damage/genetics , DNA Mutational Analysis , DNA Repair , DNA-Directed DNA Polymerase/metabolism , Deoxyguanosine/toxicity , Deoxyribonucleotides/chemical synthesis , Deoxyribonucleotides/chemistry , Deoxyribonucleotides/metabolism , Genetic Vectors , Kinetics , Molecular Conformation , Molecular Structure , Mutagenesis, Site-Directed , Mutagens/chemistry , Templates, Genetic
20.
Protein J ; 23(2): 153-66, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15106881

ABSTRACT

Arylamine N-acetyltransferases (NATs) catalyze a variety of biotransformation reactions, including N-acetylation of arylamines and O-acetylation of arylhydroxylamines. Chemical modification of hamster recombinant NAT2 with 2-(bromoacetylamino)fluorene (Br-AAF) and bromoacetanilide revealed that Br-AAF is an affinity label for the enzyme whereas bromoacetanilide inactivates NAT2 through a bimolecular alkylation process. Electrospray ionization quadrupole time-of-flight mass spectrometry analysis of Br-AAF-treated NAT2 showed that a single molecule of 2-acetylaminofluorene had been adducted. Peptide sequencing with tandem mass spectrometry identified the catalytically essential Cys68 as the alkylated amino acid. Br-AAF exhibits similar affinity for hamster NAT1 and NAT2, but is a more effective inactivator of NAT1 because, subsequent to the formation of a reversible enzyme-Br-AAF complex, the rate of alkylation of NAT1 is greater than the rate of alkylation of NAT2. Bromoacetanilide alkylates Cys68 and, to a lesser extent, Cys237 of NAT2; it does not exhibit significant selectivity for either NAT1 or NAT2.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , Arylamine N-Acetyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , 2-Acetylaminofluorene/chemistry , 2-Acetylaminofluorene/metabolism , 2-Acetylaminofluorene/pharmacology , Acetanilides/pharmacology , Acetylation , Animals , Arylamine N-Acetyltransferase/chemistry , Arylamine N-Acetyltransferase/metabolism , Cricetinae , Cystine/chemistry , Isoenzymes/chemistry , Isoenzymes/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...