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










Publication year range
1.
Mol Biol Cell ; 33(14): ar135, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36222847

ABSTRACT

The coatomer protein complex 1 (COPI) is a multisubunit complex that coats intracellular vesicles and is involved in intracellular protein trafficking. Recently we and others found that depletion of COPI complex subunits zeta (COPZ1) and delta (ARCN1) preferentially kills tumor cells relative to normal cells. Here we delineate the specific cellular effects and sequence of events of COPI complex depletion in tumor cells. We find that this depletion leads to the inhibition of mitochondrial oxidative phosphorylation and the elevation of reactive oxygen species (ROS) production, followed by accumulation of lipid droplets (LDs) and autophagy-associated proteins LC3-II and SQSTM1/p62 and, finally, apoptosis of the tumor cells. Inactivation of ROS in COPI-depleted cells with the mitochondrial-specific quencher, mitoquinone mesylate, attenuated apoptosis and markedly decreased both the size and the number of LDs. COPI depletion caused ROS-dependent accumulation of LC3-II and SQSTM1 which colocalizes with LDs. Lack of double-membrane autophagosomes and insensitivity to Atg5 deletion suggested an accumulation of a microlipophagy complex on the surface of LDs induced by depletion of the COPI complex. Our findings suggest a sequence of cellular events triggered by COPI depletion, starting with inhibition of oxidative phosphorylation, followed by ROS activation and accumulation of LDs and apoptosis.


Subject(s)
Autophagy , Neoplasms , Reactive Oxygen Species , Apoptosis , Coat Protein Complex I/metabolism , Lipids , Neoplasms/metabolism
2.
Cell Mol Life Sci ; 66(5): 788-99, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18979208

ABSTRACT

The anti-metabolite 5-fluorouracil (5-FU) is employed clinically to manage solid tumors including colorectal and breast cancer. Intracellular metabolites of 5-FU can exert cytotoxic effects via inhibition of thymidylate synthetase, or through incorporation into RNA and DNA, events that ultimately activate apoptosis. In this review, we cover the current data implicating DNA repair processes in cellular responsiveness to 5-FU treatment. Evidence points to roles for base excision repair (BER) and mismatch repair (MMR). However, mechanistic details remain unexplained, and other pathways have not been exhaustively interrogated. Homologous recombination is of particular interest, because it resolves unrepaired DNA intermediates not properly dealt with by BER or MMR. Furthermore, crosstalk among DNA repair pathways and S-phase checkpoint signaling has not been examined. Ongoing efforts aim to design approaches and reagents that (i) approximate repair capacity and (ii) mediate strategic regulation of DNA repair in order to improve the efficacy of current anticancer treatments.


Subject(s)
Antimetabolites, Antineoplastic/metabolism , DNA Repair , DNA/metabolism , Fluorouracil/metabolism , Animals , Antimetabolites, Antineoplastic/chemistry , Cell Cycle/physiology , DNA/chemistry , DNA Damage , DNA Glycosylases/metabolism , DNA Repair Enzymes/metabolism , Fluorouracil/chemistry , Humans , Molecular Structure , Signal Transduction/physiology
3.
Proc Natl Acad Sci U S A ; 97(25): 13573-8, 2000 Dec 05.
Article in English | MEDLINE | ID: mdl-11106395

ABSTRACT

The human 3-methyladenine DNA glycosylase [alkyladenine DNA glycosylase (AAG)] catalyzes the first step of base excision repair by cleaving damaged bases from DNA. Unlike other DNA glycosylases that are specific for a particular type of damaged base, AAG excises a chemically diverse selection of substrate bases damaged by alkylation or deamination. The 2.1-A crystal structure of AAG complexed to DNA containing 1,N(6)-ethenoadenine suggests how modified bases can be distinguished from normal DNA bases in the enzyme active site. Mutational analyses of residues contacting the alkylated base in the crystal structures suggest that the shape of the damaged base, its hydrogen-bonding characteristics, and its aromaticity all contribute to the selective recognition of damage by AAG.


Subject(s)
Base Pairing , DNA Damage , DNA Glycosylases , N-Glycosyl Hydrolases/metabolism , Binding Sites , Catalysis , Humans , Models, Molecular , Mutagenesis, Site-Directed , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/genetics , Protein Conformation
4.
Carcinogenesis ; 21(5): 901-8, 2000 May.
Article in English | MEDLINE | ID: mdl-10783310

ABSTRACT

3-Methyladenine DNA glycosylases initiate base excision repair by flipping the nucleotide bearing the target base out of double-stranded DNA into an active site pocket for glycosylic bond cleavage and base release. Substrate bases for the murine 3-methyladenine DNA glycosylase (other than 3-methyladenine) include hypoxanthine and 1,N(6)-ethenoadenine, two mutagenic adducts formed by both endogenous and exogenous agents. Using double-stranded DNA oligonucleotides containing damaged bases at specific sites, we studied the relative removal rates for these two adducts when located in different sequence contexts. One of the sequence contexts was an A:T tract, chosen because DNA secondary structure is known to change along the length of this tract, due to a progressive narrowing of the minor groove. Here we report that removal rates for hypoxanthine, but not for 1,N(6)-ethenoadenine, are dramatically affected by its location within the A:T tract. In addition, the removal rates of hypoxanthine and 1,N(6)-ethenoadenine when paired opposite thymine or cytosine were examined, and in each sequence context hypoxanthine removal decreased by at least 20-fold when paired opposite cytosine versus thymine. In contrast, 1, N(6)-ethenoadenine removal was unaffected by the identity of the opposing pyrimidine. We conclude that the removal of certain bases by the mouse 3-methyladenine DNA glycosylase can be modulated by both adjacent and opposing sequence contexts. The influence of DNA sequence context upon DNA repair rates, such as those described here, may contribute to the creation of mutational hot spots in mammalian cells.


Subject(s)
Adenine/analogs & derivatives , DNA Glycosylases , DNA/chemistry , Hypoxanthine/metabolism , N-Glycosyl Hydrolases/metabolism , Nucleic Acid Conformation , Adenine/metabolism , Animals , Base Pairing , Base Sequence , Mice
5.
Bioessays ; 21(8): 668-76, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10440863

ABSTRACT

The genome continuously suffers damage due to its reactivity with chemical and physical agents. Finding such damage in genomes (that can be several million to several billion nucleotide base pairs in size) is a seemingly daunting task. 3-Methyladenine DNA glycosylases can initiate the base excision repair (BER) of an extraordinarily wide range of substrate bases. The advantage of such broad substrate recognition is that these enzymes provide resistance to a wide variety of DNA damaging agents; however, under certain circumstances, the eclectic nature of these enzymes can confer some biological disadvantages. Solving the X-ray crystal structures of two 3-methyladenine DNA glycosylases, and creating cells and animals altered for this activity, contributes to our understanding of their enzyme mechanism and how such enzymes influence the biological response of organisms to several different types of DNA damage.


Subject(s)
DNA Glycosylases , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/metabolism , Animals , DNA Methylation , DNA Repair , Helix-Loop-Helix Motifs , Humans , Mice , Mice, Knockout , Models, Molecular , N-Glycosyl Hydrolases/genetics , Protein Conformation , Substrate Specificity
6.
Cancer Res ; 58(17): 3965-73, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9731510

ABSTRACT

DNA repair status is recognized as an important determinant of the clinical efficacy of cancer chemotherapy. To assess the role that a mammalian DNA glycosylase plays in modulating the toxicity and clastogenicity of the chemotherapeutic DNA cross-linking alkylating agents, we compared the sensitivity of wild-type murine cells to that of isogenic cells bearing homozygous null mutations in the 3-methyladenine DNA glycosylase gene (Aag). We show that Aag protects against the toxic and clastogenic effects of 1,3-bis(2-chloroethyl)-1-nitrosourea and mitomycin C (MMC), as measured by cell killing, sister chromatid exchange, and chromosome aberrations. This protection is accompanied by suppression of apoptosis and a slightly reduced p53 response. Our results identify 3-methyladenine DNA glycosylase-initiated base excision repair as a potentially important determinant of the clinical efficacy and, possibly, the carcinogenicity of these widely used chemotherapeutic agents. However, Aag does not contribute significantly to protection against the toxic and clastogenic effects of several chemotherapeutic nitrogen mustards (namely, mechlorethamine, melphalan, and chlorambucil), at least in the mouse embryonic stem cells used here. We also compare the Aag null phenotype with the Fanconi anemia phenotype, a human disorder characterized by cellular hypersensitivity to DNA cross-linking agents, including MMC. Although Aag null cells are sensitive to MMC-induced growth delay and cell cycle arrest, their sensitivity is modest compared to that of Fanconi anemia cells.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Carmustine/toxicity , DNA Repair , Guanine/analogs & derivatives , Mitomycin/toxicity , Mutagens/toxicity , N-Glycosyl Hydrolases/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , DNA Glycosylases , G2 Phase/drug effects , Guanine/metabolism , Humans , Mice , Mitosis/drug effects
7.
Proc Natl Acad Sci U S A ; 94(24): 13087-92, 1997 Nov 25.
Article in English | MEDLINE | ID: mdl-9371804

ABSTRACT

3-methyladenine (3MeA) DNA glycosylases remove 3MeAs from alkylated DNA to initiate the base excision repair pathway. Here we report the generation of mice deficient in the 3MeA DNA glycosylase encoded by the Aag (Mpg) gene. Alkyladenine DNA glycosylase turns out to be the major DNA glycosylase not only for the cytotoxic 3MeA DNA lesion, but also for the mutagenic 1,N6-ethenoadenine (epsilonA) and hypoxanthine lesions. Aag appears to be the only 3MeA and hypoxanthine DNA glycosylase in liver, testes, kidney, and lung, and the only epsilonA DNA glycosylase in liver, testes, and kidney; another epsilonA DNA glycosylase may be expressed in lung. Although alkyladenine DNA glycosylase has the capacity to remove 8-oxoguanine DNA lesions, it does not appear to be the major glycosylase for 8-oxoguanine repair. Fibroblasts derived from Aag -/- mice are alkylation sensitive, indicating that Aag -/- mice may be similarly sensitive.


Subject(s)
DNA Glycosylases , DNA Repair/genetics , N-Glycosyl Hydrolases/metabolism , Alkylating Agents/pharmacology , Animals , Crosses, Genetic , DNA, Complementary , Female , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Glycosyl Hydrolases/genetics
8.
Nucleic Acids Res ; 25(12): 2359-64, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9171086

ABSTRACT

The covalent sequence specificity of a series of nitrogen mustard and imidazole-containing analogues of distamycin was determined using modified sequencing techniques. The analogues tether benzoic acid mustard (BAM) and possess either one, two or three imidazole units. Examination of the alkylation specificity revealed that BAM produced guanine-N7 lesions in a pattern similar to conventional nitrogen mustards. The monoimidazole-BAM conjugate also produced guanine-N7 alkylation in a similar pattern to BAM, but at a 100-fold lower dose. The diimidazole and triimidazole conjugates did not produce detectable guanine-N7 alkylation but only alkylated at selected sites in the minor groove. Unexpectedly, the alkylation specificity at equivalent doses was nearly identical to that found for the previously reported pyrrole-BAM conjugates. The consensus sequence, 5'-TTTTGPuwas strongly alkylated by the triimidazole conjugate in preference to other similar sites including three occurrences of 5'-TTTTAA. Footprinting studies were carried out to examine the non-covalent DNA binding interactions. These studies revealed that the tripyrrole- BAM conjugate bound non-covalently to the same AT-rich sites as distamycin. In contrast, whereas the Im3lexitropsin bound non-covalently to GC-rich sequences, the triimidazole-BAM conjugate did not detectably footprint to either GC- or AT-rich regions at equivalent doses. The results indicate that the alkylation event is not solely dictated by the non-covalent binding and might be influenced by a unique sequence dependent conformational feature of the consensus sequence 5'-TTTTGPu.


Subject(s)
DNA Damage , DNA/chemistry , Distamycins , Imidazoles , Nitrogen Mustard Compounds , Alkylation , Base Sequence , Binding Sites , Consensus Sequence , DNA Footprinting , DNA-Directed DNA Polymerase , Oligodeoxyribonucleotides , Taq Polymerase
9.
Anticancer Drug Des ; 12(1): 49-60, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9051113

ABSTRACT

The sequence specificity of alkylation for a series of pyrrole- and imidazole-containing analogues of distamycin that tether the nitrogen mustard chlorambucil (CHL) was determined using modified sequencing techniques. Examination of the sequence specificity of alkylation for the imidazole-CHL conjugates using a Taq polymerase stop assay revealed that although the doses required to produce similar amounts of damage were at least 10-fold lower, the sequence specificity of alkylation was essentially identical to that seen for CHL. The guanine-N7 alkylation pattern, which consisted of guanines within runs of guanines, was confirmed using a piperidine cleavage assay. The pyrrole-CHL conjugates also produced a similar pattern of alkylation to that seen for CHL, but one exception was a unique site strongly alkylated only by the di- and tripyrrole-CHL conjugates. The unique lesions, at AG for the dipyrrole-CHL conjugate and G for the tripyrrole-CHL conjugate in the sequence 5'-GAAGAT, were confirmed as minor groove adenine- and guanine-N3 lesions using a thermal cleavage assay.


Subject(s)
Antineoplastic Agents, Alkylating/metabolism , Chlorambucil/metabolism , Distamycins/metabolism , Alkylation
11.
Br J Cancer ; 76(12): 1596-603, 1997.
Article in English | MEDLINE | ID: mdl-9413948

ABSTRACT

The chemistry of the mitomycin C-related drug indoloquinone EO9 would suggest that its mechanism of action is likely to involve DNA damage after reductive activation. The ability of this agent to induce DNA damage in intact cells has been examined using alkaline filter elution. After treatment with pharmacologically relevant concentrations of EO9, both DNA strand breaks and interstrand cross-links were detected in rat Walker tumour cells and human HT29 colon carcinoma cells. These cell lines express relatively high levels of DT-diaphorase (NAD(P)H: quinone acceptor oxidoreductase), which is believed to be involved in EO9 activation. The extent of DNA damage was increased by approximately 30-fold under hypoxia in BE colon carcinoma cells that express non-functional DT-diaphorase, but this dramatic hypoxia enhancement was not seen in HT-29 cells. These data are consistent with cytotoxicity studies that indicate that DT-diaphorase appears to be important in EO9 activation under aerobic conditions, but other enzymes may be more relevant under hypoxia. The involvement of DT-diaphorase in DNA damage induction was further investigated using cell-free assays. DNA cross-links were detectable in plasmid DNA co-incubated with EO9, cofactor and DT-diaphorase but not in the absence of this enzyme. In contrast, using a Taq polymerase stop assay, monofunctional alkylation was detected in plasmid DNA without metabolic activation, although the sequence selectivity was altered after reduction catalysed by DT-diaphorase.


Subject(s)
Antineoplastic Agents/pharmacology , Aziridines/pharmacology , DNA Damage , DNA/drug effects , Indolequinones , Indoles/pharmacology , NAD(P)H Dehydrogenase (Quinone)/physiology , Animals , Aziridines/metabolism , Humans , Indoles/metabolism , Rats , Tumor Cells, Cultured
12.
Biochemistry ; 34(40): 13034-41, 1995 Oct 10.
Article in English | MEDLINE | ID: mdl-7548062

ABSTRACT

The covalent sequence specificity of a series of nitrogen mustard-containing analogues of distamycin was determined using modified sequencing techniques. The analogues tether benzoic acid mustard (BAM) and possess either one, two, or three pyrrole-amide units. Previous characterization of the biological profile of the series revealed an increase in cytotoxicity for each corresponding increase in the number of pyrrole units, while showing poor cross-link formation in isolated and cellular DNA. Examination of the sequence specificity revealed that BAM produced guanine-N7 lesions in similar manner to other conventional nitrogen mustards. The monopyrrole BAM conjugate also produced guanine-N7 alkylation in a similar pattern to BAM. However, alkylation of adenines was also seen that was found to be minor groove adenine-N3 lesions. The dipyrrole and tripyrrole conjugates did not produce detectable guanine-N7 alkylation but only alkylated in AT tracts. In addition, the tripyrrole conjugate preferentially alkylated only a subset of those sites alkylated by the monopyrrole and dipyrrole conjugates. Two sites, 5'-TTTTGG and 5'-TTTTGA, confirmed as guanine-N3 and adenine-N3 lesions, respectively, were strongly alkylated by the tripyrrole conjugate in preference to other similar sites including three occurrences of 5'-TTTTAA. Footprinting studies comparing distamycin and the tripyrrole conjugate showed identical non-covalent recognition of AT-rich sites. Hence, the drug that possessed the most enhanced sequence specificity for alkylation was also the most cytotoxic of this series.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , DNA/chemistry , Distamycins/chemistry , Mechlorethamine/chemistry , Nitrogen Mustard Compounds/chemistry , Base Sequence , Benzoates/chemistry , Benzoic Acid , DNA/biosynthesis , DNA Adducts/chemistry , DNA Damage , DNA Primers/chemistry , Molecular Sequence Data , Structure-Activity Relationship
13.
Bioorg Med Chem ; 3(6): 623-9, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7582941

ABSTRACT

The design, syntheses, photochemical and biological properties of conjugates of pyrene with pyrrole- (1) and imidazole-containing (2) analogues of netropsin are reported. The results of an ethidium displacement assay and circular dichroism (CD) titration studies show both compounds bind with a higher affinity to poly(dA-dT) than to poly(dG-dC). In addition they bind as strongly to T4 coliphage DNA as to calf thymus DNA suggesting the binding occurs in the minor groove. The quenching rate constants of the singlet excited states of agents 1 and 2 by molecular oxygen were found to be 8.5 x 10(9) M-1S-1 and 7.7 x 10(9) M-1S-1, suggesting the involvement of singlet oxygen. Both compounds showed some cytotoxicity to human chronic myeloid leukemia K562 cells in the dark. Upon irradiation the activities were significantly enhanced resulting in photoinduced dose modifications of 8 and 14 for 1 and 2, respectively under the conditions employed. Both agents were markedly more phototoxic than 1-pyrenebutyric acid 8. To address the mechanism of action of compounds 1 and 2 their photoactivated abilities to produce DNA strand breaks were measured. Both agents caused increased single strand breakage with increasing UV exposure. The concentrations (EC50) of 1 and 2 needed to cause 50% single-strand cleavage of pBR322 DNA upon UV-A activation were found to be 40 microM and 45 microM, respectively. In contrast, no DNA strand breaks were observed in the dark with either conjugate or with 8 following irradiation. DNA strand breaks were measured in drug treated K562 cells using alkaline elution.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antineoplastic Agents/pharmacology , DNA/drug effects , Netropsin/analogs & derivatives , Photosensitizing Agents/pharmacology , Pyrenes/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/radiation effects , Bacteriophage T4/genetics , Circular Dichroism , DNA/chemistry , DNA Damage , DNA, Viral/drug effects , Humans , Netropsin/chemistry , Netropsin/radiation effects , Photosensitizing Agents/chemistry , Photosensitizing Agents/radiation effects , Pyrenes/pharmacology , Structure-Activity Relationship , Thymus Gland , Tumor Cells, Cultured , Ultraviolet Rays
14.
Drug Des Discov ; 12(4): 323-35, 1995 Apr.
Article in English | MEDLINE | ID: mdl-9040992

ABSTRACT

The synthesis, DNA binding and biological evaluation of two benzoic acid mustard derivatives of imidazole-containing analogues of distamycin in which the C-terminus is modified to contain a terminal carboxamide are described. The apparent DNA binding constants of compounds 5 and 6 were determined using an ethidium displacement assay, and the results showed that they do not have the AT sequence selectivity of distamycin and they show an acceptance for GC base pairs. Based on their pronounced binding to T4 DNA the data suggest that they bind to the minor groove of DNA. The cytotoxicities of compounds 5 and 6 in human chronic myeloid leukemia cells were determined using a MTT assay, and their IC50 values were 27 and 16 microM, respectively, and higher than the corresponding non-terminal carboxamide-containing analogues 3 and 4. Both compounds were however markedly more active than the non-targeted mustard BAM [N,N-bis (-2-chloroethyl)-4-aminobenzoic acid]. In the NCI panel of cell lines 5 gave a distinctly different pattern of tumor selectivity from 6. While these compounds were shown to alkylate DNA using a CD alkylation assay (35 +/- 10% for 5 and 85 +/- 10% for 6), they produced interstrand crosslinks poorly, even at 100 microM drug concentrations. Based on preliminary data from a polymerase stop assay compounds 3-6 gave different patterns of sequence selection monoalkylation which may contribute to their differing biological activities.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Distamycins/chemical synthesis , Distamycins/pharmacology , Imidazoles/chemical synthesis , Mustard Compounds/chemical synthesis , Mustard Compounds/pharmacology , Antineoplastic Agents/metabolism , DNA, Viral/metabolism , Distamycins/metabolism , Drug Design , Humans , Mustard Compounds/metabolism , Tumor Cells, Cultured/drug effects
15.
Nucleic Acids Res ; 23(1): 81-7, 1995 Jan 11.
Article in English | MEDLINE | ID: mdl-7870593

ABSTRACT

FCE 24517, a novel distamycin derivative possessing potent antitumor activity, is under initial clinical investigation in Europe. In spite of the presence of a benzoyl nitrogen mustard group this compound fails to alkylate the N7 position of guanine, the major site of alkylation by conventional nitrogen mustards. Characterisation of DNA-drug adducts revealed only a very low level of adenine adduct formation. Using a modified Maxam-Gilbert sequencing method the consensus sequence for FCE 24517-adenine adduct formation was found to be 5'-TTTTGA-3'. A single base modification in the hexamer completely abolishes the alkylation of adenine. Using a Taq polymerase stop assay alkylations were confirmed at the A present in the hexamer TTTTGA and, in addition, in one out of three TTTTAA sequences present in the plasmid utilized. The sequence specificity of alkylation by FCE 24517 is therefore the most striking yet observed for an alkylating agent of small molecular weight.


Subject(s)
Antineoplastic Agents/pharmacology , DNA/chemistry , DNA/drug effects , Distamycins/pharmacology , Nitrogen Mustard Compounds/pharmacology , Adenine/chemistry , Alkylation , Antineoplastic Agents/chemistry , Base Sequence , Binding Sites , DNA/genetics , DNA Adducts/chemistry , DNA Adducts/drug effects , DNA Primers/genetics , Distamycins/chemistry , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Nitrogen Mustard Compounds/chemistry , Nucleic Acid Conformation , Plasmids/chemistry , Plasmids/drug effects , Plasmids/genetics
16.
Anticancer Drug Des ; 9(6): 511-25, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7880376

ABSTRACT

Two series of tethered nitrogen mustards based on the minor groove-binding and A/T sequence-specific natural product distamycin have been synthesized and evaluated. The conjugates, which have a modified dimethylamino C-terminus, are comprised of one, two or three pyrrole carboxamide units linked to either benzoic acid mustard (BAM) or chlorambucil (CHL). The DNA binding properties, in vitro cytotoxicities and DNA cross-linking abilities were determined for each of the conjugates. The conjugates were found to bind preferentially to poly(dA.dT) compared to poly(dG.dC) DNA by ethidium displacement and circular dichroism. The di- and tripyrrole conjugates had higher binding affinities than the monopyrrole conjugates. All the conjugates were more cytotoxic than the nitrogen mustards themselves. Cytotoxicity increased with the increase from one to three pyrrole units and the CHL conjugates were more cytotoxic than the corresponding BAM analogues. The CHL conjugates were able to cross-link plasmid DNA at a 10-fold lower dose than CHL itself. The BAM conjugates showed < 10% cross-linking at doses which gave 100% cross-linking with the CHL conjugates. In cells, the CHL conjugates showed significant cross-linking at the IC50 values, while the BAM conjugates showed no evidence of cross-link formation even at 10 times the IC50 value. These results are discussed in reference to a series of previously reported GC-recognizing imidazole analogues possessing the same nitrogen mustard groups.


Subject(s)
Alkylating Agents , DNA/metabolism , Distamycins/chemistry , Nitrogen Mustard Compounds/chemistry , Pyrroles/chemistry , Alkylating Agents/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzoates/chemistry , Benzoic Acid , Binding Sites , Cattle , Cell Death/drug effects , Chlorambucil/chemistry , Circular Dichroism , Cross-Linking Reagents , Ethidium , Humans , Nitrogen Mustard Compounds/metabolism , Nitrogen Mustard Compounds/pharmacology , Plasmids , Poly dA-dT/metabolism , Pyrroles/metabolism , Pyrroles/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured
18.
Anticancer Drug Des ; 8(3): 173-92, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8517912

ABSTRACT

The syntheses of oligoimidazolecarboxamido analogues of distamycin wherein the N-terminus contains either a benzyl-mustard 8 or chlorambucil moiety 9-11 are reported. Based on data from an ethidium displacement assay and CD studies, these compounds, along with the N-benzoyl mustards 6 and 7, were shown to have increased acceptance of GC-rich sequences over distamycin. Compounds 8-11 which contain an electron-donating group (sigma < 0) para to the bischloroethylamino moiety, were found to have significantly enhanced reactivity with DNA compared to the benzoyl mustards 6 and 7. Through dialysis experiments, the benzyl and chlorambucil mustards were shown to alkylate calf thymus DNA more readily than the benzoyl mustards, presumably due to destabilization of the aziridinium intermediate by the electron-withdrawing (sigma > 0) carboxamido group of the benzoyl compounds. Compounds 8-11 were found to alkylate guanine-N7 in the major groove, while compounds 6 and 7 did not, suggesting that they may have different modes of DNA interaction. Mustards 8-11 were also more efficient than 6 and 7 at producing DNA interstrand cross-links in isolated DNA. In general, for these compounds, the cytotoxicity against human chronic myeloid leukemia K562 cells and the panel of human tumor cell lines of the National Cancer Institute increased with the number of imidazole moieties. The IC50 values of compounds 7 and 8 were similar, even though the latter compound was at least 100-fold more efficient at forming DNA cross-links in isolated DNA. Similarly, compounds 9-11 were less cytotoxic than 6 and 7, although they were more efficient cross-linkers in isolated DNA. A direct comparison of the three imidazole-containing benzoyl mustard 7 with the corresponding chlorambucil-containing 11 for their ability to form interstrand cross-links in cells revealed that the former compound showed no cross-linking even at doses in excess of the IC50, whereas the latter produced extensive cross-linking. This further suggests that these compounds exert their biological activity through different mechanisms. It is proposed that the aromatic moiety of compounds 8-11, which bind to the minor groove, may be able to intercalate between GC base pairs and the protruding bischloroethylamino group would be positioned to alkylate and cross-link at guanine-N7 sites in the major groove. However, the benzoyl mustards, which have a rigid amino linkage between the imidazole and aromatic-mustard moieties, do not have the flexibility to intercalate into the DNA.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Alkylating Agents , Antineoplastic Agents/chemical synthesis , DNA Damage , Drug Design , Antineoplastic Agents/chemistry , Base Sequence , Circular Dichroism , DNA/chemistry , Humans , In Vitro Techniques , Mechlorethamine , Models, Molecular , Nucleic Acid Conformation , Spectrophotometry, Ultraviolet , Structure-Activity Relationship , Tumor Cells, Cultured/drug effects
19.
J Med Chem ; 36(7): 863-70, 1993 Apr 02.
Article in English | MEDLINE | ID: mdl-8464041

ABSTRACT

Imidazole containing analogues 7, 10, and 17 of distamycin wherein the C-terminus contain a dimethylamino moiety have been shown to selectively bind to the minor groove of GC-rich sequences. Accordingly, these agents were employed as vectors for the delivery of a variety of alkylating agents to GC-rich sequences. The alkylating agents are attached to the N-terminus of these vectors thus providing the benzoyl N-mustards (8, 15, and 18 that contain one, two, and three imidazole units, respectively) and substituted acetamides 11-14. Results from the ethidium displacement assay for the formamides 7, 10, and 17 and mustards 15 and 18 showed that these agents bind to calf thymus DNA, poly(dA.dT), poly(dG.dC), and also to coliphage T4 DNA, thus confirming their binding in the minor groove. The reduced binding constants of these compounds for poly(dA.dT) while still binding as strongly, or more strongly, to poly(dG.dC) than distamycin provided evidence for their acceptance of GC sequences. Selectivity for GC-rich sequences was also indicated by CD titration studies. Titration of 10, 15, 17, and 18 to poly(dA.dT) produced weak drug-induced CD bands at approximately 330 nm; however, interaction of these agents to poly(dG.dC) in equimolar drug concentrations gave strong bands in this region. Results from dialysis and cross-link gel experiments provided evidence of alkylation and cross-linking of DNA by the mustards which could explain their enhanced cytotoxicity over the formamido analogues. The bifunctional N-mustard-containing analogues 15 and 18 are significantly more cytotoxic than the monoalkylating acetamides 11-14. The mustards also exhibited significant activity against cell lines derived from solid tumors such as melanomas, ovarian cancers, CNS cancers, and small cell lung cancer.


Subject(s)
Alkylating Agents/chemical synthesis , Distamycins/chemical synthesis , Imidazoles/chemical synthesis , Alkylating Agents/toxicity , Animals , Cattle , Circular Dichroism , DNA/metabolism , Distamycins/toxicity , Humans , Imidazoles/toxicity , Structure-Activity Relationship , Tumor Cells, Cultured/drug effects
20.
Biochemistry ; 32(16): 4237-45, 1993 Apr 27.
Article in English | MEDLINE | ID: mdl-8476851

ABSTRACT

The DNA binding properties of a series of imidazole-containing and C-terminus-modified analogues 4-7 of distamycin are described. These analogues contain one to four imidazole units, respectively. Data from the ethidium displacement assay showed that these compounds bind in the minor groove of DNA, with the relative order of binding constants of 6 (Im3) > 7 (Im4) > 5 (Im2) > 4 (Im1). The reduced binding constants of these compounds for poly(dA-dT) relative to distamycin, while they still interact strongly with poly(dG-dC), provided evidence of GC sequence acceptance. The preferences for GC-rich sequences by these compounds were established from a combination of circular dichroism (CD) titration, proton nuclear magnetic resonance (1H-NMR), and methidiumpropylethylenediaminetetraacetate-iron(II) [MPE.Fe-(II)] footprinting studies. In the CD studies, these compounds produced significantly larger DNA-induced ligand bands with poly(dG-dC) than poly(dA-dT) at comparable ligand concentrations. 1H-NMR studies of the binding of 5 to d-[CATGGCCATG]2 provided further evidence of the recognition of GC sequences by these compounds, and suggested that the ligand was located on the underlined sequence in the minor groove with the C-terminus oriented over the T residue. MPE footprinting studies on a GC-rich BamHI/SalI fragment of pBR322 provided unambiguous evidence for the GC sequence selectivity for some of these compounds. Compounds 4 and 7 produced poor footprints on the gels; however, analogues 5 and 6 gave strong footprints.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Base Sequence , Cytosine , DNA/chemistry , Guanine , Imidazoles , Nucleic Acid Conformation , Oligodeoxyribonucleotides/chemistry , Circular Dichroism , DNA, Viral/chemistry , Distamycins , Edetic Acid/analogs & derivatives , Ethidium , Hydrogen Bonding , Imidazoles/chemical synthesis , Indicators and Reagents , Iron Chelating Agents , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Sequence Data , Poly dA-dT/chemistry , Polydeoxyribonucleotides/chemistry , Spectrophotometry, Ultraviolet/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...