Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Int J Mol Sci ; 22(13)2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-34281275

RESUMO

Human estrogens prescribed for hormone replacement therapy (HRT) are known to be potent carcinogens. To find safer estrogens, several chlorinated estrogens were synthesized and their carcinogenic potential were determined. A pellet containing either 2-chloro-17ß-estradiol (2-ClE2) or 4-chloro-17ß-estradiol (4-ClE2) was implanted subcutaneously for 52 weeks into August Copenhagen Irish (ACI) rats, a preferred animal model for human breast cancer. 17ß-Estradiol (E2) frequently induced mammary tumors while both 2-ClE2 and 4-ClE2 did not. Their 17α-ethinyl forms, thought to be orally active estrogens, were also synthesized. Neither 2-chloro-17α-ethinylestradiol (2-ClEE2) nor 4-chloro-17α-ethinylestradiol (4-ClEE2) induced tumors. The less carcinogenic effects were supported by histological examination of mammary glands of ACI rats treated with the chlorinated estrogens. A chlorine atom positioned at the 2- or 4-position of E2 may prevent the metabolic activation, resulting in reducing the carcinogenicity. 2-ClE2 and 4-ClE2 administered subcutaneously and 2-ClEE2 and 4-ClEE2 given orally to ovariectomized rats all showed uterotrophic potency, albeit slightly weaker than that of E2. Our results indicate that less carcinogenic chlorinated estrogens retaining estrogenic potential could be safer alternatives to the carcinogenic estrogens now in use for HRT.


Assuntos
Carcinógenos/toxicidade , Estradiol/análogos & derivados , Terapia de Reposição de Estrogênios/efeitos adversos , Neoplasias Mamárias Experimentais/prevenção & controle , Animais , Testes de Carcinogenicidade , Carcinógenos/síntese química , Dano ao DNA , Estradiol/síntese química , Estradiol/toxicidade , Etinilestradiol/análogos & derivados , Etinilestradiol/síntese química , Etinilestradiol/toxicidade , Feminino , Humanos , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/patologia , Ratos , Ratos Endogâmicos ACI , Útero/efeitos dos fármacos , Útero/patologia
2.
Nucleic Acids Res ; 38(12): e133, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20406772

RESUMO

Estrogen-DNA adducts are potential biomarkers for assessing the risk and development of estrogen-associated cancers. 4-Hydroxyequilenin (4-OHEN) and 4-hydroxyequilin (4-OHEQ), the metabolites of equine estrogens present in common hormone replacement therapy (HRT) formulations, are capable of producing bulky 4-OHEN-DNA adducts. Although the formation of 4-OHEN-DNA adducts has been reported, their quantitative detection in mammalian cells has not been done. To quantify such DNA adducts, we generated a novel monoclonal antibody (4OHEN-1) specific for 4-OHEN-DNA adducts. The primary epitope recognized is one type of stereoisomers of 4-OHEN-dA adducts and of 4-OHEN-dC adducts in DNA. An immunoassay with 4OHEN-1 revealed a linear dose-response between known amounts of 4-OHEN-DNA adducts and the antibody binding to those adducts, with a detection limit of approximately five adducts/10(8) bases in 1 microg DNA sample. In human breast cancer cells, the quantitative immunoassay revealed that 4-OHEN produces five times more 4-OHEN-DNA adducts than does 4-OHEQ. Moreover, in a mouse model for HRT, oral administration of Premarin increased the levels of 4-OHEN-DNA adducts in various tissues, including the uterus and ovaries, in a time-dependent manner. Thus, we succeeded in establishing a novel immunoassay for quantitative detection of 4-OHEN-DNA adducts in mammalian cells.


Assuntos
Anticorpos Monoclonais/imunologia , Adutos de DNA/imunologia , Ensaio de Imunoadsorção Enzimática , Envelhecimento , Animais , Especificidade de Anticorpos , Linhagem Celular Tumoral , Adutos de DNA/análise , Adutos de DNA/química , Equilenina/análogos & derivados , Equilenina/química , Equilenina/metabolismo , Equilina/análogos & derivados , Equilina/química , Equilina/metabolismo , Estrogênios Conjugados (USP)/administração & dosagem , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C
3.
Int J Cancer ; 128(4): 974-82, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20824696

RESUMO

Treatment with tamoxifen (TAM) increases the risk of developing endometrial cancer in women. The carcinogenic effect is thought to involve initiation and/or promotion resulting from DNA damage induced by TAM as well as its estrogenic action. To minimize this serious side-effect while increasing the anti-breast cancer potential, a new benzopyran antiestrogen, 2E-3-{4-[(7-hydroxy-2-oxo-3-phenyl-2H-chromen-4-yl)-methyl]-phenyl}-acrylic acid (SS5020), was synthesized. Unlike TAM, SS5020 exhibits no genotoxic activity to damage DNA. Furthermore, SS5020 does not present significant uterotrophic potential in rats; in contrast, the structurally related compounds, TAM, toremifene, raloxifene (RAL) and SP500263 all have uterotrophic activity. At the human equivalent molar dose of TAM (0.33 or 1.0 mg/kg), SS5020 had much stronger antitumor potential than those same antiestrogens against 7,12-dimethylbenz(a)anthracene-induced mammary carcinoma in rats. The growth of human MCF-7 breast cancer xenograft implanted into athymic nude mice was also effectively suppressed by SS5020. SS5020, lacking genotoxic and estrogenic actions, could be a safer and stronger antiestrogen alternative to TAM and RAL for breast cancer therapy and prevention.


Assuntos
Cinamatos/uso terapêutico , Moduladores de Receptor Estrogênico/uso terapêutico , Neoplasias Mamárias Experimentais/prevenção & controle , Umbeliferonas/uso terapêutico , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Carcinógenos/toxicidade , Cinamatos/síntese química , Cinamatos/química , Adutos de DNA , Moduladores de Receptor Estrogênico/síntese química , Moduladores de Receptor Estrogênico/química , Feminino , Humanos , Neoplasias Mamárias Experimentais/induzido quimicamente , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células Neoplásicas Circulantes , Ratos , Ratos Sprague-Dawley , Tamoxifeno/uso terapêutico , Umbeliferonas/síntese química , Umbeliferonas/química
4.
Int J Cancer ; 127(5): 1021-7, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20039324

RESUMO

Ingestion of aristolochic acids (AA) contained in herbal remedies results in aristolochic acid nephropathy (AAN), which is characterized by chronic renal failure, tubulointerstitial fibrosis and urothelial cancer. AA I and AA II, primary components in AA, have similar genotoxic potential, whereas only AA I shows severe renal toxicity in rodents. AA I is demethylated to form 8-hydroxy-aristolochic acid I (AA Ia) as a major metabolite. However, the nephrotoxicity and genotoxicity of AA Ia has not yet been determined. AA Ia was isolated from urine collected from rats treated with AA I and characterized by NMR and mass spectrometry. The purified AA Ia was administered intraperitoneally to C3H/He male mice for 9 days and its toxicity was compared with AA I. Using (32)P-postlabeling/polyacrylamide gel electrophoresis, the level of AA Ia-derived DNA adducts in renal cortex was approximately 70-110 times lower than that observed with AA I, indicating that AA Ia has only a limited genotoxicity. Supporting this result, when calf thymus DNA was reacted with AA Ia in a buffer containing zinc dust, the formation of AA Ia-DNA adducts was two-orders of magnitude lower than that of AA I. Histopathologic analysis revealed that unlike AA I, no significant changes were detected in the renal cortex of mice treated with AA Ia. Therefore, the contribution of AA Ia to renal toxicity is minimum. We conclude the metabolic pathway of converting AA I to AA Ia functions as the detoxification of AA I.


Assuntos
Ácidos Aristolóquicos/toxicidade , Ácidos Aristolóquicos/urina , Carcinógenos/toxicidade , Adutos de DNA/genética , Nefropatias/induzido quimicamente , Metilação , Animais , Ácidos Aristolóquicos/isolamento & purificação , Cromatografia Líquida de Alta Pressão , DNA/genética , Inativação Metabólica , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Ratos , Ratos Wistar
5.
Int J Cancer ; 127(7): 1718-26, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20073065

RESUMO

Long-term treatment with tamoxifen (TAM) increases the risk of developing endometrial cancer in women. Several antiestrogens developed in last decades have been discontinued from clinical testing because of their undesirable effects on the uterus. To avoid such serious side-effect while increasing the drug's anti-breast cancer potential, new triphenylethylene antiestrogens, 2E-3-{4-[(E)-4-chloro-1-(4-hydroxyphenyl)-2-phenylbut-1-enyl]-phenyl} acrylic acid (SS1020) and 2E-3-{4-[(Z)-4-chloro-1,2-diphenylbut-1-enyl]phenyl}acrylic acid (SS1010), were designed as safer alternatives. Unlike TAM, SS1020 does not present significant uterotrophic potential in rats; in contrast, SS1010, a compound removing a 4-OH moiety from SS1020, represented weak uterotrophic activity. The structurally related compounds 4-hydroxytamoxifen, toremifene, ospemifene, raloxifene (RAL) and GW5638 all have uterotrophic activity. In addition, SS1020 and SS1010 exhibit no genotoxic activity to damage hepatic DNA in rats. Therefore, SS1020 was selected as a safer antiestrogen candidate and used for evaluating the antitumor potential in animals. At the human equivalent doses of TAM, SS1020 had antitumor potential much higher than that of TAM, RAL and GW5638 against 7,12-dimethylbenz(a)anthracene-induced mammary carcinoma in rats. The growth of human MCF-7 breast cancer xenograft implanted into athymic nude mice was also effectively suppressed by SS1020. SS1020, lacking estrogenic and genotoxic actions and having strong antitumor potency superior to that of TAM and RAL, could be a safer alternative for breast cancer therapy and prevention.


Assuntos
Neoplasias do Endométrio/prevenção & controle , Antagonistas de Estrogênios/uso terapêutico , Moduladores de Receptor Estrogênico/uso terapêutico , 9,10-Dimetil-1,2-benzantraceno , Animais , Adutos de DNA/farmacologia , Antagonistas de Estrogênios/farmacologia , Moduladores de Receptor Estrogênico/síntese química , Feminino , Humanos , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos , Ratos , Tamoxifeno/análogos & derivados , Tamoxifeno/uso terapêutico , Útero/efeitos dos fármacos , Útero/fisiologia
6.
Toxicol Lett ; 318: 99-103, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31669098

RESUMO

Fluorination preventing metabolic hydroxylation of 17ß-estradiol (E2) was applied to investigate the mechanisms underlying estrogen-induced carcinogenesis. Either 2-fluoro-17ß-estradiol (2-FE2) or 4-fluoro-17ß-estradiol (4-FE2) was administered subcutaneously for 52 weeks to August Copenhagen Irish (ACI) rats, the preferred animal model for human breast cancer. 4-FE2 induced frequent mammary tumors whereas 2-FE2 did not. The cumulative incidence of mammary tumors in rats treated with 4-FE2 was comparable to that observed with E2. The carcinogenic results were supported by histological examination of mammary glands of fluorinated estrogen-treated ACI rats. To evaluate the estrogenic potential of the fluorinated estrogens, 2-FE2 or 4-FE2 was administrated subcutaneously to ovariectomized rats. Both 4-FE2 and 2-FE2 showed high uterotrophic potency. Our results indicate that estrogenic potential may not be the sole factor driving mammary tumorigenesis. Since fluorination inhibits metabolic hydroxylation of E2 at the substituted position, the carcinogenic effect may occur through the metabolic activation of 2-hydroxylated E2, in combination with the compound's estrogenic potency.


Assuntos
Neoplasias da Mama/induzido quimicamente , Transformação Celular Neoplásica/induzido quimicamente , Estradiol/análogos & derivados , Animais , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/patologia , Estradiol/toxicidade , Feminino , Tamanho do Órgão/efeitos dos fármacos , Ratos Endogâmicos ACI , Medição de Risco , Útero/efeitos dos fármacos , Útero/patologia
7.
Genes Environ ; 41: 9, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30976361

RESUMO

Tamoxifen (TAM) has been prescribed worldwide to patients with and women at high-risk of breast cancer. However, long-term use of TAM increases the incidence of endometrial cancer. The carcinogenic mechanisms of TAM have been extensively investigated. TAM is hydroxylated and sulfonated at α-carbon to form α-hydroxytamoxifen-O-sulfonate. This metabolite readily reacts with genomic DNA, particularly with 2'-deoxyguanosine, leading to DNA replication error. TAM also exerts estrogenic activity at endometrial tissue to induce endometrial hyperplasia. Therefore, our efforts focused on the development of novel and safer anti-estrogens to diminish carcinogenic potential of TAM based on chemical modifications. In this review, we describe a crucial idea of our drug design and introduce our compounds SS1020 and SS5020, possessing high effectiveness, and no genotoxic and estrogenic activities.

8.
Biochemistry ; 47(25): 6695-701, 2008 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-18512958

RESUMO

Treatment with estrogen increases the risk of breast, ovary, and endometrial cancers in women. DNA damage induced by estrogen is thought to be involved in estrogen carcinogenesis. In fact, Y-family human DNA polymerases (pol) eta and kappa, which are highly expressed in the reproductive organs, miscode model estrogen-derived DNA adducts during DNA synthesis. Since the estrogen-DNA adducts are a mixture of 6alpha- and 6beta-diastereoisomers of dG-N(2)-6-estrogen or dA-N(6)-6-estrogen, the stereochemistry of each isomeric adduct on translesion synthesis catalyzed by DNA pols has not been investigated. We have recently established a phosphoramidite chemical procedure to insert 6alpha- or 6beta-isomeric N(2)-(estradiol-6-yl)-2'-deoxyguanosine (dG-N(2)-6-E(2)) into oligodeoxynucleotides. Using such site-specific modified oligomer as a template, the specificity and frequency of miscoding by dG-N(2)-6alpha-E(2) or dG-N(2)-6beta-E(2) were explored using pol eta and a truncated form of pol kappa (pol kappaDeltaC). Translesion synthesis catalyzed by pol eta bypassed both the 6alpha- and 6beta-isomers of dG-N(2)-6-E(2), with a weak blockage at the adduct site, while translesion synthesis catalyzed by pol kappaDeltaC readily bypassed both isomeric adducts. Quantitative analysis of base substitutions and deletions occurring at the adduct site showed that pol kappaDeltaC was more efficient than pol eta by incorporating dCMP opposite both 6alpha- and 6beta-isomeric dG-N(2)-6-E(2) adducts. The miscoding events occurred more frequently with pol eta, but not with pol kappaDeltaC. Pol eta promoted incorporation of dAMP and dTMP at both the 6alpha- and 6beta-isomeric adducts, generating G --> T transversions and G --> A transitions. One- and two-base deletions were also formed. The 6alpha-isomeric adduct promoted slightly lower frequency of dCMP incorporation and higher frequency of dTMP incorporation and one-base deletions, compared with the 6beta-isomeric adduct. These observations were supported by steady-state kinetic studies. Taken together, the miscoding property of the 6alpha-isomeric dG-N(2)-6-E(2) is likely to be similar to that of the 6beta-isomeric adduct.


Assuntos
Adutos de DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Estradiol/análogos & derivados , Guanosina/análogos & derivados , Monofosfato de Adenosina/química , Monofosfato de Adenosina/metabolismo , Sequência de Bases , Catálise , Monofosfato de Citidina/química , Monofosfato de Citidina/metabolismo , Adutos de DNA/química , Adutos de DNA/genética , DNA Polimerase Dirigida por DNA/genética , Eletroforese em Gel de Poliacrilamida , Estradiol/química , Estradiol/metabolismo , Guanosina/química , Guanosina/metabolismo , Humanos , Cinética , Estrutura Molecular , Mutação , Nucleotídeos/química , Nucleotídeos/metabolismo , Oligodesoxirribonucleotídeos/química , Oligodesoxirribonucleotídeos/genética , Oligodesoxirribonucleotídeos/metabolismo , Deleção de Sequência , Estereoisomerismo , Especificidade por Substrato , Timidina Monofosfato/química , Timidina Monofosfato/metabolismo
9.
Int J Cancer ; 122(9): 2142-7, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18183587

RESUMO

Raloxifene (RAL) significantly reduced the incidence of breast cancer in women at high risk of developing the disease. Unlike tamoxifen (TAM), an increased incidence of endometrial cancer was not observed in women treated with RAL. However, RAL, having two hydroxyl moieties, can be conjugated rapidly through phase II metabolism and excreted, making it difficult to achieve adequate bioavailability by oral administration in humans. As a result, higher doses must be administered to obtain an efficacy equivalent to that achieved with TAM. To improve oral bioavailability and antitumor potential, RAL diphosphate was prepared as a prodrug. RAL diphosphate showed several orders of magnitude lower binding potential to both ER alpha and ER beta and weak antiproliferative potency on cultured human MCF-7 and ZR-75-1 breast cancer cells, as compared to RAL. However, RAL diphosphate has a much higher bioavailability than RAL, endowing it with higher antitumor potential than RAL against both 7,12-dimethylbenz(a)anthracene-induced mammary carcinoma in rats and human MCF-7 breast cancer implanted in athymic nude mice. The RAL prodrug may provide greater clinical benefit for breast cancer therapy and prevention.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Moduladores de Receptor Estrogênico/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Organofosfatos/farmacologia , Pró-Fármacos , Cloridrato de Raloxifeno/análogos & derivados , Cloridrato de Raloxifeno/farmacologia , 9,10-Dimetil-1,2-benzantraceno , Administração Oral , Animais , Antineoplásicos Hormonais/sangue , Disponibilidade Biológica , Carcinógenos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Moduladores de Receptor Estrogênico/sangue , Feminino , Humanos , Neoplasias Mamárias Experimentais/induzido quimicamente , Camundongos , Camundongos Nus , Fosforilação , Cloridrato de Raloxifeno/sangue , Ratos , Ratos Sprague-Dawley , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Transplante Heterólogo
10.
J Mol Biol ; 371(5): 1151-62, 2007 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-17603077

RESUMO

4-Hydroxyequilenin (4-OHEN)-dC is a major, potentially mutagenic DNA adduct induced by equine estrogens used for hormone replacement therapy. To study the miscoding property of 4-OHEN-dC and the involvement of Y-family human DNA polymerases (pols) eta, kappa and iota in that process, we incorporated 4-OHEN-dC into oligodeoxynucleotides and used them as templates in primer extension reactions catalyzed by pol eta, kappa and iota. Pol eta inserted dAMP opposite 4-OHEN-dC, accompanied by lesser amounts of dCMP and dTMP incorporation and base deletion. Pol kappa promoted base deletions as well as direct incorporation of dAMP and dCMP. Pol iota worked in conjunction with pol kappa, but not with pol eta, at a replication fork stalled by the adduct, resulting in increased dTMP incorporation. Our results provide a direct evidence that Y-family DNA pols can switch with one another during synthesis past the lesion. No direct incorporation of dGMP, the correct base, was observed with Y-family enzymes. The miscoding potency of 4-OHEN-dC may be associated with the development of reproductive cancers observed in women receiving hormone replacement therapy.


Assuntos
DNA Polimerase Dirigida por DNA/química , Equilenina/química , Equilina/química , Estrogênios/química , Animais , Sequência de Bases , Catálise , Adutos de DNA , Feminino , Terapia de Reposição Hormonal/métodos , Cavalos , Humanos , Cinética , Modelos Químicos , Dados de Sequência Molecular
11.
Chem Res Toxicol ; 21(5): 1120-4, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18447394

RESUMO

Long-term hormone replacement therapy with equine estrogens is associated with a higher risk of breast, ovarian, and endometrial cancers. Reactive oxygen species generated through redox cycling of equine estrogen metabolites may damage cellular DNA. Such oxidative stress may be linked to the development of cancers in reproductive organs. Xeroderma pigmentosa complementation group C-knockout ( Xpc-KO) and wild-type mice were treated with equilenin (EN), and the formation of 7,8-dihydro-8-oxodeoxyguanosine (8-oxodG) was determined as a marker of typical oxidative DNA damage, using liquid chromatography electrospray tandem mass spectrometry. The level of hepatic 8-oxodG in wild-type mice treated with EN (5 or 50 mg/kg/day) was significantly increased by approximately 220% after 1 week, as compared with mice treated with vehicle. In the uterus also, the level of 8-oxodG was significantly increased by more than 150% after 2 weeks. Similar results were observed with Xpc-KO mice, indicating that Xpc does not significantly contribute to the repair of oxidative damage. Oxidative DNA damage generated by equine estrogens may be involved in equine estrogen carcinogenesis.


Assuntos
Dano ao DNA/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Estrogênios/farmacologia , Cavalos , Animais , Proteínas de Ligação a DNA/genética , Equilenina/análogos & derivados , Equilenina/química , Equilenina/farmacologia , Feminino , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Knockout , Estrutura Molecular , Oxirredução
12.
Carcinogenesis ; 28(11): 2363-6, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17361010

RESUMO

N2-ethylidene-2'-deoxyguanosine (N2-ethylidene-dG) is a major DNA adduct induced by acetaldehyde. Although it is unstable in the nucleoside form, it is relatively stable when present in DNA. In this study, we analyzed three acetaldehyde-derived DNA adducts, N2-ethylidene-dG, N2-ethyl-2'-deoxyguanosine (N2-Et-dG) and alpha-methyl-gamma-hydroxy-1,N2-propano-2'-deoxyguanosine (alpha-Me-gamma-OH-PdG) in the liver DNA of aldehyde dehydrogenase (Aldh)-2-knockout mice to determine the influence of alcohol consumption and the Aldh2 genotype on the levels of DNA damage. In control Aldh2+/+ mice, the level of N2-ethylidene-dG adduct in liver DNA was 1.9 +/- 0.7 adducts per 10(7) bases and was not significantly different than that of Aldh2+/- and -/- mice. In alcohol-fed mice (20% ethanol for 5 weeks), the adduct levels of Aldh2+/+, +/- and -/- mice were 7.9 +/- 1.8, 23.3 +/- 4.0 and 79.9 +/- 14.2 adducts per 10(7) bases, respectively, and indicated that adduct level was alcohol and Aldh2 genotype dependent. In contrast, an alcohol- or Aldh2 genotype-dependent increase was not observed for alpha-Me-gamma-OH-PdG, and N2-Et-dG was not detected in any of the analyzed samples. In conclusion, the risk of formation of N2-ethylidene-dG in model animal liver in vivo is significantly higher in the Aldh2-deficient population and these results may contribute to our understanding of in vivo adduct formation in humans.


Assuntos
Aldeído Desidrogenase/metabolismo , Adutos de DNA/biossíntese , Desoxiguanosina/análogos & derivados , Etanol/toxicidade , Fígado/efeitos dos fármacos , Aldeído Desidrogenase/genética , Aldeído-Desidrogenase Mitocondrial , Animais , Cromatografia Líquida , Adutos de DNA/química , Desoxiguanosina/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espectrometria de Massas em Tandem
13.
Methods Mol Biol ; 314: 307-21, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16673890

RESUMO

32P-Postlabeling analysis is a powerful technique for detecting, identifying, and quantifying DNA adducts induced by mutagens or carcinogens. The method involves enzymatic digestion of the DNA sample to nucleoside 3'-monophosphates, and partial purification of the adducted nucleotides followed by their 5'-labeling with 32P. For analysis of DNA adducts, polyethyleneimine-cellulose thin-layer chromatography (TLC) plates have traditionally been used to resolve 32P-labeled DNA adducts (32P-postlabeling/ TLC analysis). However, the TLC procedure is time consuming and labor intensive. To expedite analyses, we recently devised a 32P-postlabeling protocol that utilizes nondenaturing polyacrylamide gel electrophoresis (PAGE) and permits multiple DNA samples to be run on a single gel (32P-postlabeling/PAGE analysis). Using this method, the detection limit for 5 microg of DNA is approx 7 adducts/10(9) nucleotides, similar to that for 32P-postlabeling/TLC. For still higher sensitivity and resolution, high-performance liquid chromatography (HPLC) combined with a radioisotope detector system (32P-postlabeling/HPLC analysis) can be used to increase the detection limit to approx 3 adducts/10(10) nucleotides. Here we describe all three 32P-postlabeling techniques.


Assuntos
Cromatografia Líquida de Alta Pressão , Cromatografia em Camada Fina , Adutos de DNA/análise , Dano ao DNA , Eletroforese em Gel de Poliacrilamida , Marcação por Isótopo , Animais , Células Cultivadas , DNA/química , Desoxirribonucleases/química , Humanos , Nucleosídeos/química , Radioisótopos de Fósforo/análise
14.
Cancer Res ; 64(9): 3144-7, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15126352

RESUMO

Tamoxifen (TAM) possesses antiestrogen activity and is widely used for the treatment or prevention of breast cancer. However, it is also carcinogenic in human uterus and rat liver, highlighting the profound complexity of its actions. To explore the molecular mechanisms of TAM-induced mutagenesis, we analyzed the effects of this drug on gene-disrupted chicken B lymphocyte (DT40) clones deficient in various DNA repair pathways. Rad18, Rev3, and Polkappa are involved in translesion DNA synthesis (TLS), which facilitates recovery from replication blocks on damaged template strands. DT40 cells deficient in TLS were found to be hypersensitive to TAM, exhibiting an increase in chromosomal breaks. Furthermore, these mutants were also hypersensitive to 4-hydroxyestradiol, a physiological metabolite of estrogen. These data suggest a contribution of TLS to the prevention of chromosomal breaks by TAM and estrogen, and they therefore indicate that such error-prone DNA synthesis underlies mutagenesis induced by these agents.


Assuntos
Linfócitos B/efeitos dos fármacos , Quebra Cromossômica , Cromossomos/efeitos dos fármacos , Reparo do DNA/genética , Estrogênios/farmacologia , Tamoxifeno/farmacologia , Animais , Linfócitos B/ultraestrutura , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Linhagem Celular , Galinhas , Cromossomos/genética , DNA/efeitos dos fármacos , DNA/genética , Dano ao DNA
15.
Cancer Res ; 63(15): 4402-6, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12907611

RESUMO

The risk of developing endometrial cancer is increased in breast cancer patients treated with tamoxifen (TAM) and in healthy women undergoing TAM chemoprevention. We have detected previously TAM-DNA adducts in the endometrium of women receiving TAM (Shibutani et al., Carcinogenesis, 21: 1461-1467, 2000). To investigate the genotoxic damage induced by TAM in the uterus and other tissues of primates, we gave adult female cynomolgus monkeys six times the human-equivalent dose of TAM (2 mg/kg body weight/day) for 30 days. DNA samples were prepared from the uterus, ovary, liver, kidney, and brain cortex of three TAM-exposed monkeys and one control monkey and were analyzed as coded specimens. To identify the TAM-DNA adducts, we established a new high-performance liquid chromatography gradient system for (32)P-postlabeling/high-performance liquid chromatography analysis, which can resolve the trans- and cis-diastereoisomers of alpha-(N(2)-deoxyguanosinyl)TAM (dG-N(2)-TAM), alpha-(N(2)-deoxyguanosinyl)-N-desmethylTAM, and alpha-(N(2)-deoxyguanosinyl)tamoxifen N-oxide. Trans-forms of dG-N(2)-TAM and dG-N(2)-N-desTAM adducts were detected in the livers of all three TAM-fed monkeys at levels of 2.7 adducts/10(8) nucleotides and 1.7 adducts/10(8) nucleotides, respectively. The levels of dG-N(2)-TAM adducts observed in the uterus of one monkey and in the ovaries of two monkeys were approximately 10-fold lower than those observed in the livers. TAM exposure also induced dG-N(2)-TAM adduct in the brain cortex of all three monkeys with a value of 1.5 adducts/10(8) nucleotides. No TAM-DNA adducts were detected in the kidneys or in any tissues obtained from the unexposed monkey. Our results suggest that women receiving TAM may form genotoxic damage in many organs, including the reproductive organs.


Assuntos
Adutos de DNA/análise , DNA/metabolismo , Moduladores de Receptor Estrogênico/metabolismo , Moduladores de Receptor Estrogênico/farmacologia , Tamoxifeno/metabolismo , Tamoxifeno/farmacologia , Animais , Córtex Cerebral/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , DNA/efeitos dos fármacos , Adutos de DNA/metabolismo , Feminino , Fígado/metabolismo , Macaca fascicularis , Ovário/metabolismo , Radioisótopos de Fósforo , Útero/metabolismo
16.
DNA Repair (Amst) ; 3(11): 1475-82, 2004 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-15380103

RESUMO

The present study was performed to generate monoclonal antibodies capable of detecting N-acetoxy-2-acetylaminofluorene (NA-AAF)-derived DNA adducts in human cells in situ. As an immunogen, we employed NA-AAF-modified single-stranded DNA coupled electrostatically to methylated protein and we produced five different monoclonal antibodies. All of them showed strong binding to NA-AAF-modified DNA, but had undetectable or minimal binding to undamaged DNA. Competitive inhibition experiments revealed that the epitope recognized by these antibodies is N-(deoxyguanosin-8-yl)-2-acetylaminofluorene (dG-C8-AAF) in DNA, although deacetylated N-(deoxyguanosin-8-yl)-2-aminofluorene in DNA is also recognized with slightly less efficiency. In contrast, these antibodies did not bind to 3-(deoxyguanosin-N(2)-yl)-2-acetylaminofluorene in DNA or to UV-induced lesions in DNA. Interestingly, they showed only minimal binding to small AAF-nucleoside adducts (dG-C8-AAF), indicating that DNA regions flanking a DNA-bound adduct, in addition to the adduct itself, are essential for the stable binding of the antibodies. Using an enzyme-linked immunosorbent assay with the most promising antibody (AAF-1), we detected the concentration-dependent induction of NA-AAF-modified adducts in DNA from repair deficient xeroderma pigmentosum (XP) cells treated with physiological concentrations of NA-AAF. Moreover, the assay enabled to confirm that normal human cells efficiently repaired NA-AAF-induced DNA adducts but not XP-A cells. Most importantly, the formation of NA-AAF-induced DNA adducts in individual nuclei of XP cells could be clearly visualized using indirect immunofluorescence. Thus, we succeeded in establishing novel monoclonal antibodies capable of the in situ detection of NA-AAF-induced DNA adducts in human cells.


Assuntos
Acetoxiacetilaminofluoreno/análise , Acetoxiacetilaminofluoreno/imunologia , Anticorpos Monoclonais , Adutos de DNA/análise , Adutos de DNA/imunologia , Animais , Bovinos , Linhagem Celular , Dano ao DNA , Reparo do DNA , Ensaio de Imunoadsorção Enzimática , Humanos , Hibridomas/imunologia , Camundongos , Microscopia de Fluorescência , Xeroderma Pigmentoso/metabolismo
17.
J Mol Biol ; 344(3): 665-74, 2004 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-15533436

RESUMO

Cellular DNA is damaged by nitric oxide (NO), a multifunctional bioregulator and an environmental pollutant that has been implicated in diseases associated with cancer and chronic inflammation. 2'-Deoxyxanthosine (dX) is a major NO-derived DNA lesion. To explore the mutagenic potential of dX, a 38-mer oligodeoxynucleotide ((5')CATGCTGATGAATTCCTTCXCTTCTTTCCTCTCCCTTT) modified site-specifically with dX at the X position was prepared post-synthetically and used as a DNA template in primer extension reactions catalyzed by calf thymus DNA polymerase (pol) alpha and human DNA pol beta, eta, and kappa. Primer extension reactions catalyzed by pol alpha or beta in the presence of four dNTPs were retarded at the dX lesion while pol eta and kappa readily bypassed the lesion. The fully extended products were analyzed to quantify the miscoding specificity and frequency of dX using two-phase polyacrylamide gel electrophoresis (PAGE). With pol alpha, eta and kappa, incorrect dTMP was preferentially incorporated opposite the lesion, along with lesser amounts of dCMP, the correct base. When pol beta was used, direct incorporation of correct dCMP was primarily observed, accompanied by small amounts of misincorporation of dTMP, dAMP and dGMP. Steady-state kinetic analyses supported the results obtained from the two-phase PAGE assay. dX is a miscoding lesion capable of preferentially generating G-->A mutations. The miscoding frequency varied depending on DNA polymerase used.


Assuntos
Adutos de DNA , Reparo do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Desoxirribonucleosídeos/química , Óxido Nítrico/química , Animais , Sequência de Bases , Cromatografia Líquida de Alta Pressão , Cinética , Oligodesoxirribonucleotídeos , Moldes Genéticos
18.
Chem Biol Interact ; 152(2-3): 131-8, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15840386

RESUMO

2-Acetylaminonaphthalene (2-AAN) has been recognized as a urinary bladder carcinogen in humans. The deacetylated form, 2-aminonaphthalene (2-AN), is metabolized in vivo and reacts primarily with guanine residues in DNA, resulting in the formation of dG-N(2)-aminonaphthalene (dG-N(2)-AN) adduct. Phosphoramidite chemical procedure has recently been established in our laboratory to prepare oligodeoxynucleotides containing a single dG-N(2)-acetylaminonaphthalene (dG-N(2)-AAN) adduct. Oligodeoxynucleotides ((5')TCCTCCTNXCCTCTC, where X is dG or dG-N(2)-AAN and N is C, A, T or G) with different bases 5' flanking to the lesion were prepared and were inserted into a single-strand shuttle vectors and used to establish the mutational frequency and specificity of dG-N(2)-AAN adduct in simian kidney cells. dG-N(2)-AAN adduct promoted preferential incorporation of dCMP, the correct base, opposite the lesion. When the 5' flanking base to the lesion was C, A or T, the mutational frequency was under 2.1%. When G flanked to the lesion, the mutational frequency was slightly increased to 4.2%. Misincorporation of dAMP, dTMP, and/or dGMP varied depending on the 5' flanking base. When dG-N(2)-AAN was positioned at codon 61 of noncoding strand of human c-Ha-ras1 gene ((5')TCCTCCTXGCCTCTC, where X is dG-N(2)-AAN), the mutational frequency was 6.7%; G-->T transversions (4.7%), followed by G-->A transition (2.0%), were observed. These results demonstrated that dG-N(2)-AAN is a weak mutagenic lesion in mammalian cells. The influence of 5' flanking sequence context was observed on the mutational frequency and specificity of this adduct.


Assuntos
2-Naftilamina/análogos & derivados , 2-Naftilamina/toxicidade , Carcinógenos/toxicidade , Adutos de DNA/metabolismo , 2-Naftilamina/metabolismo , Animais , Sequência de Bases , Células COS , Carcinógenos/metabolismo , Chlorocebus aethiops , Adutos de DNA/genética , Desoxiguanosina/genética , Desoxiguanosina/metabolismo , Vetores Genéticos , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos/síntese química , Oligodesoxirribonucleotídeos/metabolismo
19.
Life Sci ; 91(11-12): 415-9, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-23227466

RESUMO

AIMS: This study was carried out to explore anti-breast cancer potential of isoflavone daidzein or its related compounds using appropriate animal models and their anti-tumor mechanism. MAIN METHODS: Daidzein or its major metabolite equol at a dose molar equivalent to tamoxifen [1.0 mg(2.7 µmol)/kg or 10 mg (27 µmol)/kg/day] was treated orally to rats bearing 7,12-dimethylbenz(a)anthracene(DMBA)-induced mammary tumors or ovariectomized athymic nude mice implanted with human MCF-7 breast cancer xenograft and an estrogen pellet. The growth of tumors was monitored for several weeks after the treatment. The cell-cycle and apoptotic stages in mammary tumors collected from rats were analyzed by flow cytometry. Immunohistochemistry analysis was also used to determine the expression of caspase-3. KEY FINDINGS: Oral treatment with daidzein or equol at a human equivalent dose suppressed the growth of both DMBA-induced mammary tumors and human MCF-7 breast cancer xenografts in rodents, the inhibitory activity being superior to that of genistein or tamoxifen. Strong apoptosis induced by daidzein or equol contributes to the anti-tumor potential. SIGNIFICANCE: Daidzein and its metabolite equol showed the potential of inhibiting the growth of mammary tumors in rodents. Daidzein or equol could be used as a core structure to design new drugs for breast cancer therapy. Our results indicate that consumption of daidzein may protect against breast cancer.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores do Crescimento/uso terapêutico , Isoflavonas/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Animais , Apoptose , Feminino , Neoplasias Mamárias Experimentais/patologia , Camundongos , Ratos
20.
Toxicol Lett ; 193(3): 224-8, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20096754

RESUMO

Long-term hormone replacement therapy is associated with an increased risk of breast, ovarian and endometrial cancers in women. Equine estrogens are a principal component of hormone replacement therapy; however, their tumorigenic potential toward mammary tissue and reproductive organs has not been extensively explored. A pellet containing equilin was inserted under the skin of female ACI rats and the development of mammary tumors was monitored. Histological examination revealed premalignant lesions such as apocrine metaplasia in whole-mount preparations of mammary gland from the equilin-treated rats. ACI rats given 10mg equilin developed palpable mammary tumors at 13 weeks of treatment, and 37.5% of the rats developed mammary tumors within 15 weeks. For 2.5mg equilin, palpable tumors were observed in 8.3% of the rats after 8 weeks' treatment; the frequency was lower than that (42.9%) observed with 2.5mg E(2). No tumors were observed in the untreated rats. Evidently, equilin is a mammary carcinogen, and this potential may be associated with development of breast and reproductive cancers in women receiving hormone replacement therapy.


Assuntos
Estrogênios Conjugados (USP)/toxicidade , Neoplasias Mamárias Experimentais/induzido quimicamente , Animais , Terapia de Reposição de Estrogênios/efeitos adversos , Feminino , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/patologia , Ratos , Ratos Endogâmicos ACI
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa