Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 427
Filter
Add more filters










Publication year range
1.
Am J Med Genet A ; 149A(3): 328-35, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19206178

ABSTRACT

Snyder-Robinson syndrome (SRS, OMIM 309583) is a rare X-linked syndrome characterized by mental retardation, marfanoid habitus, skeletal defects, osteoporosis, and facial asymmetry. Linkage analysis localized the related gene to Xp21.3-p22.12, and a G-to-A transition at point +5 of intron 4 of the spermine synthase gene, which caused truncation of the SMS protein and loss of enzyme activity, was identified in the original family. Here we describe another family with Snyder-Robinson syndrome in two Mexican brothers and a novel mutation (c.496T>G) in the exon 5 of the SMS gene confirming its involvement in this rare X-linked mental retardation syndrome.


Subject(s)
Chromosomes, Human, X , Genes, X-Linked , Mental Retardation, X-Linked/genetics , Mutation, Missense , Spermine Synthase/genetics , Adult , DNA Mutational Analysis , Exons , Genetic Linkage , Humans , Intellectual Disability/genetics , Male , Marfan Syndrome/genetics , Osteoporosis/genetics , Pedigree , Scoliosis/genetics , Siblings , Young Adult
2.
J Med Genet ; 45(8): 539-43, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18550699

ABSTRACT

We report the identification of a novel mutation at a highly conserved residue within the N-terminal region of spermine synthase (SMS) in a second family with Snyder-Robinson X-linked mental retardation syndrome (OMIM 309583). This missense mutation, p.G56S, greatly reduces SMS activity and leads to severe epilepsy and cognitive impairment. Our findings contribute to a better delineation and expansion of the clinical spectrum of Snyder-Robinson syndrome, support the important role of the N-terminus in the function of the SMS protein, and provide further evidence for the importance of SMS activity in the development of intellectual processing and other aspects of human development.


Subject(s)
Mental Retardation, X-Linked/genetics , Mutation, Missense , Spermine Synthase/genetics , Adult , Child , DNA Mutational Analysis , Female , Genes, Recessive , Humans , Male , Pedigree , Syndrome
3.
Amino Acids ; 33(2): 231-40, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17396215

ABSTRACT

The polyamines spermidine and spermine have been hypothesized to possess different functions in the protection of DNA from reactive oxygen species. The growth and survival of mouse fibroblasts unable to synthesize spermine were compared to their normal counterparts in their native and polyamine-depleted states in response to oxidative stress. The results of these studies suggest that when present at normal or supraphysiological concentrations, either spermidine or spermine can protect cells from reactive oxygen species. However, when polyamine pools are pharmacologically manipulated to produce cells with low levels of predominately spermine or spermidine, spermine appears to be more effective. Importantly, when cells are depleted of both glutathione and endogenous polyamines, they exhibit increased sensitivity to hydrogen peroxide as compared to glutathione depletion alone, suggesting that polyamines not only play a role in protecting cells from oxidative stress but this role is distinct from that played by glutathione.


Subject(s)
Hydrogen Peroxide/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Spermidine/physiology , Spermine/physiology , Animals , Apoptosis , Cells, Cultured , DNA Damage , Eflornithine/pharmacology , Glutathione/pharmacology , Guanidines/pharmacology , In Situ Nick-End Labeling , Mice
4.
Biochem Soc Trans ; 35(Pt 2): 295-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17371264

ABSTRACT

Studies over many years have suggested that increased polyamine synthesis may be necessary for neoplastic growth. This review summarizes recent work on the regulation of putrescine production both de novo and via the degradation of higher polyamines and provides a summary of studies using transgenic mice in which the levels of proteins that regulate these processes (L-ornithine decarboxylase, antizyme and spermidine/spermine-N(1)-acetyltransferase) are altered.


Subject(s)
Neoplasms/pathology , Polyamines/metabolism , Cell Division , Homeostasis , Humans , Ornithine Decarboxylase/metabolism , Putrescine/metabolism
5.
Amino Acids ; 27(2): 207-14, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15378410

ABSTRACT

In a previous research, we have shown that adequate levels of polyamines are required in transformed mouse fibroblasts for the correlated activations of MAPK subtypes (ERK and JNK) and caspases induced by etoposide and leading to apoptosis. We report now that the treatment of fibroblasts with etoposide also elicited a progressive and sustained increase of NF-kappaB activation. The DNA binding activity of p65 NF-kappaB subunit was increased up to approximately 4-fold and was accompanied by enhancement of p65 phosphorylation. A two days pre-treatment of fibroblasts with alpha-difluoromethylornithine (DFMO), which caused polyamine depletion, provoked a slight activating effect when given alone, but markedly inhibited the etoposide-induced increases in p65 DNA binding and phosphorylation. The NF-kappaB inhibiting effect of DFMO was prevented by the addition of exogenous putrescine, which restored the intracellular content of polyamines. Selective inhibitors of the etoposide-stimulated MAPK subtypes also reduced NF-kappaB activation. Moreover, pharmacological NF-kappaB inhibition reduced the increase in caspase activity and cell death elicited by etoposide, suggesting that NF-kappaB is involved in signaling to apoptosis. The results of the present study, together with our previous findings, suggest that polyamines play a permissive role in the pathways triggered by etoposide and leading to cell death of fibroblasts, by supporting the activation of MAPKs, NF-kappaB and caspases.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Etoposide/pharmacology , Fibroblasts/metabolism , NF-kappa B/metabolism , Polyamines/chemistry , Animals , Apoptosis , Blotting, Western , Caspases/metabolism , Cell Line, Transformed , Cell Nucleus/metabolism , Coumarins/chemistry , DNA/metabolism , Eflornithine/chemistry , Enzyme Inhibitors/pharmacology , Etoposide/chemistry , Flavonoids/pharmacology , In Situ Nick-End Labeling , MAP Kinase Signaling System , Mice , Phosphorylation , Protein Binding , Time Factors
6.
Biochem Soc Trans ; 31(2): 356-60, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12653638

ABSTRACT

Transgenic mice expressing proteins altering polyamine levels in a tissue-specific manner have considerable promise for evaluation of the roles of polyamines in normal, hypertrophic and neoplastic growth. This short review summarizes the available transgenic models. Mice with large increases in ornithine decarboxylase (ODC), S-adenosylmethionine decarboxylase or antizyme, a protein regulating polyamine synthesis by reducing polyamine transport and ODC in the heart, have been produced using constructs in which the protein is expressed from the alpha -myosin heavy-chain promoter. These mice are useful in studies of the role of polyamines in hypertrophic growth. Expression from keratin promoters has been used to target increased synthesis of ODC, spermidine/spermine-N(1)-acetyltransferase (SSAT) and antizyme in the skin. Such expression of ODC leads to an increased sensitivity to chemical and UV carcinogenesis. Expression of antizyme inhibits carcinogenesis in skin and forestomach. Expression of SSAT increases the incidence of skin papillomas and their progression to carcinomas in response to a two-stage carcinogenesis protocol. These results establish the importance of polyamines in carcinogenesis and neoplastic growth and these transgenic mice will be valuable experimental tools to evaluate the importance of polyamines in mediating responses to oncogenes and studies of cancer chemoprevention.


Subject(s)
Biogenic Polyamines/physiology , Mice, Transgenic , Neoplasms/etiology , Acetyltransferases/biosynthesis , Acetyltransferases/metabolism , Animals , Biogenic Polyamines/biosynthesis , Biogenic Polyamines/metabolism , Humans , Hypertrophy/enzymology , Hypertrophy/etiology , Mice , Neoplasms/enzymology , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/metabolism
7.
Environ Mol Mutagen ; 38(2-3): 235-43, 2001.
Article in English | MEDLINE | ID: mdl-11746760

ABSTRACT

O6-alkylguanine DNA-alkyltransferase (AGT) is a widely distributed DNA repair protein that protects living organisms from endogenous and exogenous alkylation damage to DNA at the O6-position of guanine. The search of the C. elegans genome database for an AGT protein revealed the presence of a protein (cAGT-2) with some similarity to known AGTs in addition to the easily recognized cAGT-1 protein. The predicted protein sequence of cAGT-2 contains the amino acid sequence -ProCysHisPro- at the presumed active site of the protein, whereas all other known AGTs have -ProCysHisArg-. A truncated version of the cAGT-2 protein was expressed in E. coli. This purified recombinant protein was able to repair O6-methylguanine and O4-methylthymine adducts in DNA in vitro and also reacted with the bulky benzyl adduct in O6-benzylguanine. This fragment of cAGT-2 (104 amino acids) is the smallest protein possessing AGT activity yet described. The full-length cAGT-2 protein (274 amino acids) totally lacks the N-terminal domain present in all other known AGTs but has a long C-terminal extension that has significant homology to histone 1C. Expression of cAGT-2 in an E. coli strain lacking endogenous AGT activity provided modest but statistically significant resistance to the toxicity of N-methyl-N'-nitro-N-nitrosoguanidine, confirming that cAGT-2 is an alkyltransferase.


Subject(s)
Alkyl and Aryl Transferases/analysis , Caenorhabditis elegans/enzymology , DNA Repair , Alkyl and Aryl Transferases/genetics , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Molecular Sequence Data , Sequence Alignment
8.
Biochemistry ; 40(32): 9484-94, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11583147

ABSTRACT

S-Adenosylmethionine decarboxylase belongs to a small class of amino acid decarboxylases that use a covalently bound pyruvate as a prosthetic group. It is an essential enzyme for polyamine biosynthesis and provides an important target for the design of anti-parasitic and cancer chemotherapeutic agents. We have determined the structures of S-adenosylmethionine decarboxylase complexed with the competitive inhibitors methylglyoxal bis(guanylhydrazone) and 4-amidinoindan-1-one-2'-amidinohydrazone as well as the irreversible inhibitors 5'-deoxy-5'-[N-methyl-N-[(2-aminooxy)ethyl]amino]adenosine, 5'-deoxy-5'-[N-methyl-N-(3-hydrazinopropyl)amino]adenosine, and the methyl ester analogue of S-adenosylmethionine. These structures elucidate residues important for substrate binding and show how those residues interact with both covalently and noncovalently bound inhibitors. S-Adenosylmethionine decarboxylase has a four-layer alphabeta betaalpha sandwich fold with residues from both beta-sheets contributing to substrate and inhibitor binding. The side chains of conserved residues Phe7, Phe223, and Glu247 and the backbone carbonyl of Leu65 play important roles in binding and positioning the ligands. The catalytically important residues Cys82, Ser229, and His243 are positioned near the methionyl group of the substrate. One molecule of putrescine per monomer is observed between the two beta-sheets but far away from the active site. The activating effects of putrescine may be due to conformational changes in the enzyme, to electrostatic effects, or both. The adenosyl moiety of the bound ligand is observed in the unusual syn conformation. The five structures reported here provide a framework for interpretation of S-adenosylmethionine decarboxylase inhibition data and suggest strategies for the development of more potent and more specific inhibitors of S-adenosylmethionine decarboxylase.


Subject(s)
Adenosylmethionine Decarboxylase/chemistry , Adenosylmethionine Decarboxylase/metabolism , Protein Structure, Tertiary , Adenosylmethionine Decarboxylase/antagonists & inhibitors , Adenosylmethionine Decarboxylase/genetics , Binding Sites , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Protein Binding , Protein Folding , Putrescine/chemistry , Putrescine/metabolism , Substrate Specificity
9.
Biochemistry ; 40(32): 9495-504, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11583148

ABSTRACT

S-Adenosylmethionine decarboxylase (AdoMetDC) is synthesized as a proenzyme that cleaves itself in a putrescine-stimulated reaction via an N-->O acyl shift and beta-elimination to produce an active enzyme with a catalytically essential pyruvoyl residue at the new N-terminus. N-->O acyl shifts initiate the self-processing of other proteins such as inteins and amidohydrolases, but their mechanisms in such proteins are not well understood. We have solved the crystal structure of the H243A mutant of AdoMetDC to 1.5 A resolution. The mutant protein is trapped in the ester form, providing clear evidence for the structure of the ester intermediate in the processing of pyruvoyl enzymes. In addition, a putrescine molecule is bound in a charged region within the beta-sandwich, and cross-links the two beta-sheets through hydrogen bonds to several acidic residues and ordered water molecules. The high-resolution structure provides insight into the mechanism for the self-processing reaction and provides evidence for the mechanism for simulation of the self-processing reaction by putrescine. Studies of the effects of putrescine or 4-aminobutanol on the processing of mutant AdoMetDC proenzymes are consistent with a model in which a single activator molecule interacts with buried Asp174, Glu178, and Glu256, leading to an alteration in the position of Glu11, resulting in stimulation of self-processing.


Subject(s)
Adenosylmethionine Decarboxylase/chemistry , Protein Precursors/metabolism , Protein Structure, Tertiary , Putrescine/metabolism , Adenosylmethionine Decarboxylase/antagonists & inhibitors , Adenosylmethionine Decarboxylase/genetics , Adenosylmethionine Decarboxylase/metabolism , Binding Sites , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Protein Binding , Protein Processing, Post-Translational , Putrescine/chemistry , Water/chemistry
10.
Cancer Res ; 61(16): 6073-81, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507056

ABSTRACT

To directly evaluate the role of increased ornithine decarboxylase (ODC) and polyamines in mouse skin carcinogenesis, we used bovine keratin 5 (K5) and keratin 6 (K6) promoter elements to direct the expression of antizyme (AZ) to specific skin cell populations. AZ is a multifunctional regulator of polyamine metabolism that inhibits ODC activity, stimulates ODC degradation, and suppresses polyamine uptake. K5-AZ mice treated with 12-O-tetradecanoylphorbol-13-acetate (TPA) at 0 and 24 h exhibit increases in epidermal and dermal ODC activity that are reduced in magnitude. K6-AZ mice treated similarly do not show any increased ODC activity or protein after a second application due to TPA-induced expression of AZ protein. Epidermal and dermal polyamine content, particularly spermidine, is reduced in untreated K5-AZ mice and TPA-treated K5-AZ and K6-AZ mice. Susceptibility to 7,12-dimethylbenz(a)anthracene/TPA carcinogenesis was also investigated for two K6-AZ transgenic lines [K6-AZ(52) and K6-AZ(18)] and a single K5-AZ line. K6-AZ(52) mice had a substantial delay in tumor onset and a >80% reduction in tumor multiplicity compared with normal littermates. K6-AZ(18) and K5-AZ mice also developed fewer papillomas than littermate controls (35% and 50%, respectively), and the combination of these lines to produce double transgenic animals yielded an additive decrease (70%) in tumor multiplicity. These mice demonstrate for the first time that AZ suppresses tumor growth in an animal cancer model and provide a valuable model system to evaluate the role of ODC and polyamines in skin tumorigenesis.


Subject(s)
Ornithine Decarboxylase Inhibitors , Protein Biosynthesis , Skin Neoplasms/enzymology , Skin Neoplasms/prevention & control , Skin/metabolism , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Biogenic Polyamines/metabolism , Carcinogens/toxicity , Cattle , Enzyme Induction/drug effects , Enzyme Inhibitors/metabolism , Keratin-15 , Keratin-5 , Keratins/genetics , Mice , Mice, Transgenic , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/genetics , Promoter Regions, Genetic , Proteins/genetics , Proteins/metabolism , Skin/drug effects , Skin/enzymology , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate/toxicity
11.
Biochem J ; 358(Pt 1): 25-32, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11485548

ABSTRACT

These studies were designed to determine the consequences of constitutive overexpression of ornithine decarboxylase (ODC) in the heart. Induction of ODC is known to occur in response to agents that induce cardiac hypertrophy. However, it is not known whether high ODC levels are sufficient for the development of a hypertrophic phenotype. Transgenic mice were generated with cardiac-specific expression of a stable ODC protein using the alpha-myosin heavy-chain promoter. Founder lines with >1000-fold overexpression of ODC in the heart were established, resulting in a 50-fold overaccumulation of putrescine, 4-fold elevation in spermidine, a slight increase in spermine and accumulation of large amounts of cadaverine compared with littermate controls. Despite these significant alterations in polyamines, myocardial hypertrophy, as measured by ratio of heart to body weight, did not develop, although atrial natriuretic factor RNA was slightly elevated in transgenic ventricles. However, stimulation of beta-adrenergic signalling by isoproterenol resulted in severe hypertrophy and even death in ODC-overexpressing mice without further altering polyamine levels, compared with only a mild hypertrophy in littermates. When beta1-adrenergic stimulation was blocked by simultaneous treatment with isoproterenol and the beta1 antagonist atenolol, a significant, although reduced, hypertrophy was still present in the hearts of transgenic mice, suggesting that both beta1 and beta2 adrenergic receptors contribute to the hypertrophic phenotype. Therefore these mice provide a model to study the in vivo co-operativity between high ODC activity and activation of other pathways leading to hypertrophy in the heart.


Subject(s)
Adrenergic beta-Agonists/metabolism , Cardiomegaly/enzymology , Ornithine Decarboxylase/biosynthesis , Adrenergic beta-Agonists/pharmacology , Animals , Atenolol/pharmacology , Blotting, Southern , Cadaverine/biosynthesis , Hypertrophy/enzymology , Immunohistochemistry , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Myocardium/enzymology , Promoter Regions, Genetic , Putrescine/biosynthesis , RNA, Messenger/metabolism , Spermidine/biosynthesis , Spermine/biosynthesis , Tissue Distribution
12.
Biochem J ; 358(Pt 1): 137-45, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11485561

ABSTRACT

Spermidine/spermine N(1)-acetyltransferase (SSAT), a key enzyme in mammalian polyamine catabolism, undergoes rapid turnover (half-life approx. 30 min) and is highly inducible in response to polyamine analogues such as bis(ethyl)spermine (BE-3-4-3), which greatly stabilize the enzyme. Rapid degradation of SSAT in reticulocyte lysates was preceded by formation of a ladder of ubiquitinated forms, and required the production of high-molecular-mass complexes with ubiquitin (HMM-SSAT-Ubs). Mutation of all 11 lysines in SSAT separately to arginine demonstrated that no single lysine residue is critical for its degradation in vitro, but mutant K87R had a significantly longer half-life, suggesting that lysine-87 may be the preferred site for ubiquitination. Mutations at the C-terminus of SSAT, such as E171Q, resulted in marked stabilization of the protein, due to the lack of formation of the HMM-SSAT-Ubs. Addition of BE-3-4-3 prevented the accumulation of ubiquitin conjugates and the proteasomal degradation of wild-type SSAT. These results indicate that conformational changes brought about by the binding of polyamine analogues prevent the efficient polyubiquitination of SSAT, leading to a major increase in the amount of SSAT protein, and that alteration of the C-terminal end of the protein has a similar effect in preventing the productive interaction with an E2 or E3 component of the ubiquitin pathway.


Subject(s)
Acetyltransferases/chemistry , Cysteine Endopeptidases/chemistry , Multienzyme Complexes/chemistry , Polyamines , Spermidine/chemistry , Spermine/chemistry , Animals , Arginine/chemistry , Binding Sites , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Lysine/chemistry , Mutation , Plasmids/metabolism , Proteasome Endopeptidase Complex , Protein Binding , Protein Conformation , Rabbits , Rats , Reticulocytes/chemistry , Reticulocytes/metabolism , Time Factors , Ubiquitins/chemistry , Ubiquitins/metabolism
13.
J Biol Chem ; 276(37): 34458-64, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11461922

ABSTRACT

Ornithine decarboxylase is the initial and rate-limiting enzyme in the polyamine biosynthetic pathway. Polyamines are found in all mammalian cells and are required for cell growth. We previously demonstrated that N-hydroxyarginine and nitric oxide inhibit tumor cell proliferation by inhibiting arginase and ornithine decarboxylase, respectively, and, therefore, polyamine synthesis. In addition, we showed that nitric oxide inhibits purified ornithine decarboxylase by S-nitrosylation. Herein we provide evidence for the chemical mechanism by which nitric oxide and S-nitrosothiols react with cysteine residues in ornithine decarboxylase to form an S-nitrosothiol(s) on the protein. The diazeniumdiolate nitric oxide donor agent 1-diethyl-2-hydroxy-2-nitroso-hydrazine acts through an oxygen-dependent mechanism leading to formation of the nitrosating agents N(2)O(3) and/or N(2)O(4). S-Nitrosoglutathione inhibits ornithine decarboxylase by an oxygen-independent mechanism likely by S-transnitrosation. In addition, we provide evidence for the S-nitrosylation of 4 cysteine residues per ornithine decarboxylase monomer including cysteine 360, which is critical for enzyme activity. Finally S-nitrosylated ornithine decarboxylase was isolated from intact cells treated with nitric oxide, suggesting that nitric oxide may regulate ornithine decarboxylase activity by S-nitrosylation in vivo.


Subject(s)
Nitric Oxide/physiology , Ornithine Decarboxylase Inhibitors , Binding Sites , Cyclic N-Oxides/pharmacology , Cysteine/metabolism , Glutathione/analogs & derivatives , Glutathione/pharmacology , Hydrazines/pharmacology , Imidazoles/pharmacology , Luminescent Measurements , Nitrogen Oxides , Nitroso Compounds/pharmacology , Ornithine Decarboxylase/chemistry , Ornithine Decarboxylase/metabolism , Photolysis , S-Nitrosoglutathione
14.
Cancer Epidemiol Biomarkers Prev ; 10(3): 191-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11303587

ABSTRACT

Alpha-difluoromethylornithine (DFMO) is an irreversible inhibitor of ornithine decarboxylase, the first enzyme in polyamine synthesis. Previous work showed simultaneous administration of DFMO and a zinc-deficient (ZD) diet to weanling rats from the beginning inhibited the onset of zinc-deficiency-induced esophageal cell proliferation by activating apoptosis and reduced the incidence of N-nitrosomethylbenzylamine (NMBA)-induced esophageal cancer. Because esophageal cancer initiation by NMBA is very rapid in ZD rats, this study determined whether DFMO is effective in preventing esophageal carcinogenesis when administered after the establishment of a carcinogenic environment. Weanling rats were given a ZD diet for 5 weeks to establish sustained increased esophageal cell proliferation and then an intragastric dose of NMBA. Thereafter, 20 rats were switched to DFMO-containing water while nine control ZD animals remained on deionized water; all of the animals continued on the ZD diet. Esophagi were collected 15 weeks later. The upper portion was processed for immunohistochemical analysis of cell proliferation, apoptosis, and expression of related genes, and the lower was processed for polyamine content. DFMO substantially reduces the levels of esophageal putrescine and spermidine and esophageal tumor incidence from 89 to 10% in ZD rats. Importantly, DFMO-treated ZD esophagi display increased rate of apoptosis accompanied by intense bax expression and greatly reduced cell proliferation by proliferating cell nuclear antigen expression. In addition, the p16(ink4a)/retinoblastoma control at G1 to S, deregulated in ZD esophagi, is restored after DFMO treatment. These results demonstrate that DFMO, a highly effective chemopreventive agent in esophageal carcinogenesis, reverses and counteracts esophageal cell proliferation/cancer initiation in ZD animals by way of stimulating apoptosis.


Subject(s)
Apoptosis/drug effects , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Esophageal Neoplasms/pathology , Esophageal Neoplasms/prevention & control , Proto-Oncogene Proteins c-bcl-2 , Analysis of Variance , Animals , Blotting, Western , Cell Division/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/analysis , Diet , Dimethylnitrosamine/analogs & derivatives , Disease Models, Animal , Drug Interactions , Esophageal Neoplasms/chemically induced , Genes, bcl-2 , Immunohistochemistry , Male , Polyamines/analysis , Proliferating Cell Nuclear Antigen , Proto-Oncogene Proteins/analysis , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity , Zinc/deficiency , bcl-2-Associated X Protein
15.
Clin Cancer Res ; 7(2): 421-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11234899

ABSTRACT

Bis-2-chloroethylnitrosourea (BCNU) or temozolomide (TMZ) were tested alone or in combination with the AGT inhibitors O6-benzyl-2'-deoxyguanosine (dBG) or O6-benzylguanine (BG) against human glial tumor xenografts growing s.c. in athymic mice. Four glioblastoma (SWB77, SWB40, SWB39, and D-54) and one anaplastic oligodendroglioma (SWB61) xenografts having O6-alkylguanine-DNA alkyltransferase (AGT) activities of 75, 45, 10, < 10, and 16 fmol/mg protein, respectively, were used. BCNU at 35 mg/m2 was ineffective against these tumors, although 70 mg/m2 (LD10, 75 mg/m2) produced a marked tumor growth delay (T-C) in D54 but had no effect against SWB40 or SWB77. Coadministration of BG or dBG and BCNU necessitated reduction of the BCNU dose to a maximum of 30 and 35 mg/m2, respectively, because of increased toxicity. Optimized treatment with dBG (250 mg/m2) and BCNU (35 mg/m2) resulted in T-Cs of 30, 29, 11, 16, and 14 days for SWB77, SWB40, SWB39, D-54 and SWB61, respectively. These delays were more pronounced than those induced with optimized, isotoxic treatments with BG (180 mg/m2) and BCNU (30 mg/m2). In comparison to BCNU, TMZ was less toxic, with an LD10 of 400 mg/m2. TMZ (300 mg/m2) was more effective than BCNU against SWB77, SWB40, and SWB61, inducing T-Cs of 23, 53, and 56 days, respectively. BG and dBG enhanced the toxicity of TMZ in athymic mice by decreasing the LD10 from 400 to 200 mg/m2. TMZ (180 mg/m2) with either BG (180 mg/m2) or dBG (250 mg/m2) resulted in T-Cs of 31 and 49 days in SWB77, respectively, as compared with 16 days for TMZ (180 mg/m2) alone. In SWB40, the combination of TMZ with dBG, but not with BG, was significantly more effective than the maximum tolerated dose of TMZ (300 mg/m2) alone. The combination of TMZ with AGT inactivators had no benefit, as compared with TMZ alone, against xenografts with marginal AGT activity. In conclusion, at equimolar doses dBG was less toxic than BG in athymic mice when combined with either BCNU or TMZ. In this regard, BCNU or TMZ can be used at higher doses in combination with dBG than with BG. This study further demonstrates that there is a significant benefit of depleting AGT with nonspecific AGT inhibitors prior to treatment with either BCNU or TMZ in tumors having AGT activity >45 fmol/mg protein.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Carmustine/therapeutic use , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Deoxyguanosine/analogs & derivatives , Glioma/drug therapy , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols , Apoptosis , Deoxyguanosine/therapeutic use , Drug Resistance, Neoplasm , Enzyme Inhibitors/therapeutic use , Glioma/enzymology , Glioma/pathology , Guanine/analogs & derivatives , Guanine/therapeutic use , Humans , Mice , Mice, Nude , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Temozolomide , Transplantation, Heterologous , Tumor Cells, Cultured
16.
Biochem J ; 355(Pt 1): 199-206, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11256964

ABSTRACT

Activation of the caspase proteases represents a central point in apoptosis. The requirement for spermine for the processes leading to caspase activation has been studied in transformed embryonic fibroblasts obtained from gyro (Gy) mutant male mice. These cells lack spermine synthase activity and thus provide a valuable model to study the role of spermine in cell processes. Gy fibroblasts do not contain spermine and have a higher spermidine content. However, when compared with fibroblasts obtained from normal male littermates (N cells), Gy fibroblasts were observed to grow normally. The lack of spermine did not affect the expression of Bcl-2, and caspases 3 and 9 were activated by etoposide in both N and Gy cells, indicating that spermine is dispensable for caspase activation. Spermine deficiency did not significantly influence caspase activity in cells treated with etoposide, cycloheximide or staurosporine, but sensitized the cells to UV irradiation, which triggered significantly higher caspase activity in Gy cells compared with N cells. alpha-Difluoromethylornithine (DFMO), an inhibitor of polyamine synthesis that is able to deplete cells of putrescine and spermidine, but usually does not influence spermine content, was able to produce a more complete polyamine depletion in Gy cells. This depletion, which included spermine deficiency, dramatically increased caspase activation and cell death in Gy fibroblasts exposed to UV irradiation. On the other hand, in either N or Gy cells, DFMO treatment did not influence caspase activity triggered by staurosporine, but inhibited it when the inducers were cycloheximide or etoposide. In Gy cells depleted of polyamines by DFMO, polyamine replenishment with either spermidine or spermine was sufficient to restore caspase activity induced by etoposide, indicating that, in this model, polyamines have an interchangeable role in supporting caspase activation. Therefore, spermine is not required for such activation, and the effect and specificity of polyamine depletion on caspase activity may be very different, depending on the role of polyamines in the specific death pathways engaged by different stimuli. Some inducers of apoptosis, for example etoposide, absolutely require polyamines for caspase activation, yet the lack of polyamines, particularly spermine, strongly increases caspase activation when induced by UV irradiation.


Subject(s)
Caspases/metabolism , Polyamines/metabolism , Spermine Synthase/metabolism , Animals , Blotting, Western , Cells, Cultured , Cycloheximide/pharmacology , Eflornithine/pharmacology , Enzyme Activation , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/metabolism , Male , Mice , Mice, Mutant Strains , Protein Synthesis Inhibitors/pharmacology , Spermine Synthase/genetics
17.
J Pharmacol Exp Ther ; 296(3): 958-65, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181929

ABSTRACT

Inactivation of the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT) enhances tumor cell killing by therapeutic alkylating agents. O(6)-Benzylguanine (b(6)G) can inactivate AGT and is currently in clinical trials to enhance therapy. Short oligodeoxyribonucleotides containing b(6)G are much more effective inactivators, but their use for therapeutic purposes is likely to be compromised by metabolic instability. We have therefore examined the ability to inactivate AGT of an 11-mer oligodeoxyribonucleotide containing b(6)G (11-mpBG) when modified with terminal methylphosphonate linkages to protect it from nucleases. This modification did not reduce the ability to serve as a substrate/inactivator for AGT, and 11-mpBG had an ED(50) value of 1.3 nM, more than 300-fold lower than that for b(6)G. A similar oligodeoxyribonucleotide containing O(6)-methylguanine (m(6)G) was also found to be a good substrate (ED(50) value of 10 nM), but the benzylated form was repaired more rapidly and preferentially. When added to HT29 cell cultures, 5 microM 11-mpBG was able to cause a prolonged inactivation of cellular AGT for at least 72 h and to greatly sensitize the cells to killing by 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). The 11-mpMG was ineffective at up to 20 microM, suggesting that the benzyl group allows better uptake into the cell. However, even with 11-mpBG, the 1000-fold decrease in potency toward AGT in HT29 cells compared to that toward the protein in vitro suggests that uptake may be a limiting factor. These results suggest that oligodeoxyribonucleotides such as 11-mpBG may prove to be useful drugs for potentiation of alkylating agent chemotherapy if uptake can be improved.


Subject(s)
Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , Guanine/chemistry , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Oligodeoxyribonucleotides/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Carmustine/pharmacology , Cell Survival/drug effects , Drug Resistance , Enzyme Inhibitors/chemistry , Enzyme Repression , Guanine/pharmacology , HT29 Cells , Humans , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Oligodeoxyribonucleotides/chemistry , Organophosphorus Compounds/chemistry
18.
Cancer Res ; 61(1): 53-8, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11196197

ABSTRACT

Alkylating agents such as N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) are known to covalently link alkyl groups at the position 6 of guanines (O6MG) in DNA. O6-alkylguanine-DNA alkyltransferase (AGT) specifically removes the methyl group of the O6MG. Using purified human topoisomerase I (Top1), we found an 8-10-fold enhancement of Top1 cleavage complexes when O6MG is incorporated in oligonucleotides at the +1 position relative to a unique Top1 cleavage site. Top1 poisoning by O6MG is attributable to a decrease of the Top1-mediated DNA religation as well as an increase in the enzyme cleavage step. Increased cleavage is probably linked to a change in the hydrogen bonding pattern, such as in the case of the 8-oxoguanine, whereas inhibition of religation could be attributed to altered base pairing, such as abasic sites or base mismatches, because incorporation of a 6-thioguanine did not affect Top1 activity. Top1-DNA covalent complexes are also induced in MNNG-treated CHO cells constitutively lacking the AGT enzyme. Conversely, no increase could be detected in CHO cells transfected with the wild-type human AGT. Moreover, we show that yeasts overexpressing the human Top1 are more sensitive to MNNG, whereas knock-out Top1 strain cells display some resistance to the drug. Altogether, these results suggest a role for Top1 poisoning by alkylated bases in the antiproliferative activity of alkylating agents as well as in the DNA lesions resulting from endogenous and carcinogenic DNA modifications.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Guanine/analogs & derivatives , Guanine/metabolism , Methylnitronitrosoguanidine/toxicity , Alkylating Agents/toxicity , Animals , CHO Cells/drug effects , CHO Cells/enzymology , CHO Cells/metabolism , Cricetinae , DNA/metabolism , Humans , O(6)-Methylguanine-DNA Methyltransferase/deficiency , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Transfection
19.
J Med Chem ; 43(22): 4071-83, 2000 Nov 02.
Article in English | MEDLINE | ID: mdl-11063604

ABSTRACT

A series of O(6)-allyl- and O(6)-(2-oxoalkyl)guanines were synthesized and evaluated, in comparison with the corresponding O(6)-alkylguanines, as potential inhibitors of the DNA-repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT). Simple O(6)-alkyl- and O(6)-cycloalkylguanines were weak AGT inactivators compared with O(6)-allylguanine (IC(50) = 8.5 +/- 0.6 microM) with IC(50) values ranging from 100 to 1000 microM. The introduction of substituents at C-2 of the allyl group of O(6)-allylguanine reduced activity compared with the parent compound, while analogous compounds in the O(6)-(2-oxoalkyl)guanine series exhibited very poor activity (150-1000 microM). O(6)-Cycloalkenylguanines proved to be excellent AGT inactivators, with 1-cyclobutenylmethylguanine (IC(50) = 0.55 +/- 0.02 microM) and 1-cyclopentenylmethylguanine (IC(50) = 0.39 +/- 0.04 microM) exhibiting potency approaching that of the benchmark AGT inhibitor O(6)-benzylguanine (IC(50) = 0.18 +/- 0.02 microM). 1-Cyclopentenylmethylguanine also inactivated AGT in intact HT29 human colorectal carcinoma cells (IC(50) = 0.20 +/- 0.07 microM) and potentiated the cytotoxicity of the monomethylating antitumor agent Temozolomide by approximately 3- and 10-fold, respectively, in the HT29 and Colo205 tumor cell lines. The observation that four mutant AGT enzymes resistant to O(6)-benzylguanine also proved strongly cross-resistant to 1-cyclopentenylmethylguanine indicates that the O(6)-substituent of each compound makes similar binding interactions within the active site of AGT.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Enzyme Inhibitors/chemical synthesis , Guanine/chemical synthesis , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Cell Extracts , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/pharmacology , Humans , Mutation , O(6)-Methylguanine-DNA Methyltransferase/genetics , Structure-Activity Relationship , Temozolomide , Tumor Cells, Cultured
20.
Biochem J ; 351 Pt 2: 439-47, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11023830

ABSTRACT

Mutant Gy male mice, which have previously been described as having disruption of the phosphate-regulating Phex gene and a spermine synthase gene [Meyer, Henley, Meyer, Morgan, McDonald, Mills and Price (1998) Genomics, 48, 289-295; Lorenz, Francis, Gempel, Böddrich, Josten, Schmahl and Schmidt (1998) Hum. Mol. Genet. 7, 541-547], as well as mutant Hyp male mice, which have disruption of the Phex gene only, were examined along with their respective normal male littermates. Biochemical analyses of extracts of brains, hearts and livers of 5-week-old mice showed that Gy males lacked any significant spermine synthase activity as well as spermine content. Organs of Gy males had a higher spermidine content. This was caused not only by the lack of conversion of spermidine into spermine, but also because of compensatory increases in the activities of other polyamine biosynthetic enzymes. Gy males were half the body weight of their normal male littermates at weaning age. Hyp males, however, were no different in size when compared with their controls. High mortality of Gy males occurs by weaning age and this mortality was shown to be largely post-natal. Embryonic fibroblasts were isolated from Gy males and their normal male littermates and were similarly shown to lack any significant spermine synthase activity as well as spermine content. The lack of spermine, however, had no significant effect on the growth of immortalized fibroblasts or of primary fibroblast cultures. Similarly, there was no difference in the time of senescence of primary fibroblast cultures from Gy males compared with cultures derived from normal male littermates. However, the lack of spermine did increase the sensitivity of immortalized fibroblasts to killing by the chloroethylating agent 1, 3-bis(2-chloroethyl)-N-nitrosourea. Therefore both the Gy male mice and derived embryonic fibroblasts provide valuable models to study the importance of spermine and spermine synthase, without the use of inhibitors which may have additional side effects.


Subject(s)
Fibroblasts/enzymology , Polyamines/metabolism , Spermine Synthase/deficiency , Animals , Antineoplastic Agents, Alkylating/pharmacology , Body Weight , Brain/metabolism , Carmustine/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myocardium/metabolism , Spermine/biosynthesis , Time Factors , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...