Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Endocrinol ; 35(3): 421-32, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16326830

RESUMO

Tamoxifen acts as an oestrogen antagonist in the breast reducing cell proliferation, but in the uterus as an oestrogen agonist resulting in increased cell proliferation. Tamoxifen exerts its tissue-specific effects through the oestrogen receptors (ERalpha or ERbeta). The levels and functions of the two ERs affect the response of the target tissue to oestrogen and tamoxifen. We examined the control of ER stability in breast and uterine cell lines using western blotting and RT-PCR. In MCF-7 breast-derived cells, ERalpha and ERbeta proteins were rapidly degraded via the proteasome pathway in response to oestradiol; conversely tamoxifen stabilised both receptors. In Ishikawa uterine-derived cells, oestradiol and tamoxifen stabilised ERalpha but led to degradation of ERbeta by the proteasome pathway. Further investigations showed that oestradiol induced activation of the non-genomic ERalpha/Akt signalling pathway in MCF-7 cells. We have demonstrated that the alternative Erk signalling pathway is activated in Ishikawa cells following oestradiol treatment in the absence of an active proteasome pathway and therefore increased levels of ERbeta. In conclusion, our data have demonstrated tamoxifen or oestradiol control of ER subtype stability and that non-genomic activation of transcription pathways is cell specific.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Tamoxifeno/farmacologia , Neoplasias Uterinas/tratamento farmacológico , Neoplasias Uterinas/metabolismo , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Especificidade de Órgãos , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Uterinas/genética
2.
Curr Drug Metab ; 4(3): 223-39, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12769667

RESUMO

Tamoxifen, a non-steroidal antiestrogen, is the class representative of a group of drugs that include toremifene, droloxifene and idoxifene. Tamoxifen has been successfully used worldwide as adjuvant therapy in the treatment of women with breast cancer. However, such therapy results in a slightly increased risk of endometrial cancers. Lifetime exposure of rats to high doses of tamoxifen results in a high incidence of liver tumors. Tamoxifen itself is not genotoxic but is activated in the liver to alpha-hydroxytamoxifen. This is further conjugated to form the sulfate ester as the putative reactive intermediate. Studies with recombinant human CYPs show only CYP3A4 is able to catalyze the formation of alpha-hydroxytamoxifen and the irreversible binding of [(14)C]tamoxifen to DNA. CYP3A4 and CYP2D6 convert tamoxifen to N-desmethyltamoxifen. The formation 4-hydroxytamoxifen is catalyzed by CYP2D6 and at a much lower level by CYP2C19. In women, detoxication of alpha-hydroxytamoxifen via a stable glucuronide occurs at a rate in the order of 100 fold higher than in rats whereas rates of sulfation are 3 fold lower than in rats. These factors, together with the low dose of tamoxifen used therapeutically in women, indicates a minimum risk of liver cancers. Results from (32)P-postlabeling and accelerator mass spectrometry suggest that low levels of uterine DNA binding does occur but this is probably too low to play a role in uterine tumor development and it is more likely to be the estrogen agonist action of this class of drug that is the most important factor in tumor development in humans.


Assuntos
Antagonistas de Estrogênios/efeitos adversos , Tamoxifeno/efeitos adversos , Animais , Biotransformação , Antagonistas de Estrogênios/farmacocinética , Antagonistas de Estrogênios/toxicidade , Feminino , Humanos , Inativação Metabólica , Mutagênicos/toxicidade , Tamoxifeno/farmacocinética , Tamoxifeno/toxicidade
3.
J Mol Endocrinol ; 30(1): 1-11, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12580757

RESUMO

Oral dosing of CD-1 mice on days 2-5 after birth with tamoxifen but not raloxifene disrupts the development of the myometrium, resulting in adult uterine adenomyosis. Using laser capture microdissection and RT-PCR we have investigated nerve growth factor (NGF) and cognate receptor expression in uterine cells of 6-day-old pups that may be important in early developmental changes that give rise to adenomyosis. NGF down-regulation is known to occur during terminal myogenic differentiation. NGF was found exclusively in endometrial luminal epithelium of controls. It was up-regulated 18-fold in the luminal epithelium following dosing with tamoxifen but not raloxifene. Western blotting for NGF protein in the whole uterus showed a 25-fold increase after tamoxifen treatment. Expression of the low affinity p75 neutrophin receptor (p75(NTR)) was twofold higher in the myometrium compared with luminal epithelium or stroma. This was not altered following tamoxifen treatment. There was no detectable expression of high affinity tyrosine kinase receptor (trkA(NGFR)). This study shows luminal epithelial cells of the endometrium primarily form NGF. This suggests that NGF normally regulates the differentiation of the mesenchyme into uterine myocytes through paracrine mechanisms and that an early disturbance of this process plays a key role in the subsequent development of adenomyosis.


Assuntos
Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fatores de Crescimento Neural/genética , Cloridrato de Raloxifeno/farmacologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Útero/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Sequência de Bases , Western Blotting , Primers do DNA , Feminino , Imuno-Histoquímica , Lasers , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Útero/citologia , Útero/metabolismo
4.
Toxicol Lett ; 124(1-3): 121-8, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11684364

RESUMO

The use of hydrochlorofluorocarbons (HCFCs) such as HCFC-123 (2,2-dichloro-1,1,1-trifluoroethane) and HCFC-141b (1,1-dichloro-1-fluoroethane) is becoming widespread as replacements for the ozone depleting chlorofluorocarbons. Hepatic activation of HCFC-123 or the unsaturated perchloroethylene through oxidative pathways leads to the formation of the electrophiles trifluoroacetyl chloride or trichloroacetyl chloride, respectively. These can react with epsilon-NH(2) functions of lysine in proteins and give rise to neoantigens. In the case of HCFC-123, this reaction is catalysed primarily by CYP2E1 and to a much lesser extent by the constitutive CYP2C19, CYP2B6 and CYP2C8. For perchloroethylene, the extent of activation is less and the reaction is catalysed primarily by the CYP2B family. While acute hepatotoxicity has been seen in humans exposed to HCFC-123 or halothane, little short- or long-term toxicity in rodents is observed. No immunological related toxicity of perchloroethylene has been reported in exposed humans. Long-term exposure of rats can lead to renal tubule carcinomas and in mice, hepatocellular carcinomas. These toxic reactions do not appear to be directly related to the formation of the putative trichloroacetyl chloride intermediate.


Assuntos
Clorofluorcarbonetos/efeitos adversos , Sistema Enzimático do Citocromo P-450/metabolismo , Neoplasias Renais/induzido quimicamente , Neoplasias Hepáticas/induzido quimicamente , Animais , Carcinoma Hepatocelular , Catálise , Clorofluorcarbonetos/metabolismo , Etano Clorofluorcarbonos , Humanos , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Camundongos , Oxirredução , Ratos
5.
Toxicol Lett ; 124(1-3): 129-38, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11684365

RESUMO

In this study, the metabolic activation of 2,2-dichloro-1,1,1-trifluoroethane (hydrochlorofluorocarbons-123, HCFC-123), halothane or 1,1-dichloro-1-fluoroethane (HCFC-141b) was compared to that of perchloroethylene, using lymphoblastoma derived cell lines expressing human CYP1A1, CYP1A2, CYP2E1, CYP2A6 and CYP3A4 (MCL-5 cells). A dose dependent increase in micronucleus formation was detected over a nominal concentration range of 0.05-2 mM for HCFC-123 and halothane, but this was not seen with HCFC-141b. No dose response for HCFC-123 was seen in a control cHo1 cell line not expressing this cytochrome P450's. Cell lines expressing individual human cytochrome P-450 (CYP) forms were also used to define the enzymes responsible for the clastogenic events and to investigate the formation of immunoreactive protein by microsomal fractions. It was shown that CYP2E1 or CYP2B6 catalysed the clastogenic response, but CYP2D6, CYP3A4, CYP1A2 or CYP1A1 all appeared to be inactive. The formation of neoantigenic trifluoroacetylated protein adducts by microsomal mixtures incubated with HCFC-123 and NADPH was catalysed primarily by CYP2E1 and to a lesser extent by CYP2C19, whereas, only trace levels of immunoreactive protein were seen with microsomes expressing CYP2B6 or CYP2C8. With perchloroethylene as a substrate, the extent of activation was low in comparison with HCFC-123, as judged by the absence of micronuclei formation in the MCL-5 cell line and the weak immunoreactivity of proteins following Western blotting. CYP1A2, CYP2B6 and CYP2C8 appeared to be responsible for perchloroethylene immunoreactivity and in contrast to the findings with the HCFC's, no activation of perchloroethylene by CYP2E1 could be detected. These results show that even though both saturated and unsaturated halocarbons can result in neoantigen formation, there is a marked difference in the specificity of the CYP enzymes involved in their metabolic activation.


Assuntos
Antígenos/análise , Carcinógenos/efeitos adversos , Clorofluorcarbonetos/efeitos adversos , Clorofluorcarbonetos/imunologia , Sistema Enzimático do Citocromo P-450/metabolismo , Tetracloroetileno/efeitos adversos , Western Blotting , Etano Clorofluorcarbonos , Relação Dose-Resposta a Droga , Humanos , Leucemia Linfoide/patologia , Testes para Micronúcleos , Tetracloroetileno/imunologia , Células Tumorais Cultivadas
6.
Toxicol Lett ; 124(1-3): 139-52, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11684366

RESUMO

The bioactivation and cytotoxicity in vitro of 1,1-dichloro-2,2,2-trifluoroethane (HCFC-123) and 1,1-dichloro-1-fluoroethane (HCFC-141b), two replacements for some ozone-depleting chlorofluorocarbons (CFC), were investigated in rat liver microsomes and isolated rat hepatocytes. Both compounds were activated by cytochrome P450 to reactive metabolites, as indicated by: (i) the depletion of exogenous and cellular glutathione, (ii) the increased LDH release from hepatocytes, (iii) the loss of microsomal P450 content and activities, and (iv) the formation of free radical species observed in the presence of the two compounds. Moreover, the formation of two stable metabolites and an increased production of conjugated dienes, a marker of lipid peroxidation, were observed for both HCFC-123 and HCFC-141b. The biotransformation of both compounds by pyridine- and phenobarbital-induced rat liver microsomes and the inhibition of LDH release by 4-methylpyrazole and troleandomycin indicate that P450 2E1, 2B and, possibly, also 3A are the isoforms involved in the bioactivation and toxicity of HCFC-123 and HCFC-141b in the rat.


Assuntos
Clorofluorcarbonetos/metabolismo , Clorofluorcarbonetos/toxicidade , Sistema Enzimático do Citocromo P-450/metabolismo , Animais , Biotransformação , Etano Clorofluorcarbonos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Radicais Livres , Glutationa/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , L-Lactato Desidrogenase/metabolismo , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Fenobarbital/farmacologia , Piridinas/farmacologia , Ratos , Ratos Wistar
7.
Am J Pathol ; 159(2): 623-30, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11485920

RESUMO

Adenomyosis is a fairly frequent disorder in adult women characterized by the haphazard location of endometrial glands and stroma deep within the myometrium of the uterus. This study compared the effects on uterine development of the selective estrogen receptor modulators, tamoxifen, toremifene, and raloxifene with estradiol when given orally to female mice on days 2 to 5 after birth. Uterine adenomyosis was found in all (14 of 14) mice dosed with tamoxifen and most mice (12 of 14) treated with toremifene, but in none of the vehicle-dosed controls, in only one animal treated with raloxifene at 42 and 90 days after dosing and in none of the mice treated with estradiol at 42 days. At 6 days, the uterus in the groups that developed a high incidence of adenomyosis showed histological evidence of disturbed differentiation of the myometrium. Gene-expression XY-scatterplots using Clontech mouse 1.2 Atlas mouse cDNA expression arrays analyzing total uterine RNA showed nerve growth factor-alpha, preadipocyte factor-1, and insulin-like growth factor-2 were key genes differentially modified by tamoxifen or toremifene treatment, relative to the controls. As these genes may play an important role in regulating differentiation and development of the myometrium, these data suggest that adenomyosis may be caused primarily by defects in the formation of the myometrium.


Assuntos
Endometriose/patologia , Estradiol/farmacologia , Miométrio/citologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Células Estromais/citologia , Útero/citologia , Administração Oral , Envelhecimento , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Proteínas de Ligação ao Cálcio , Diferenciação Celular/efeitos dos fármacos , Estradiol/administração & dosagem , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores do Crescimento/genética , Fator de Crescimento Insulin-Like II/genética , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana/genética , Camundongos , Miométrio/efeitos dos fármacos , Miométrio/patologia , Fator de Crescimento Neural/genética , Análise de Sequência com Séries de Oligonucleotídeos , Tamanho do Órgão/efeitos dos fármacos , Cloridrato de Raloxifeno/farmacologia , Proteínas Repressoras/genética , Células Estromais/efeitos dos fármacos , Células Estromais/patologia , Tamoxifeno/farmacologia , Toremifeno/farmacologia , Útero/efeitos dos fármacos , Útero/crescimento & desenvolvimento
8.
J Endocrinol ; 170(3): 555-64, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11524235

RESUMO

This study compares the actions of oestradiol, tamoxifen, toremifene and raloxifene on enzyme and gene expression in uterine tissues of ovariectomised rats over 72 h. The time-course for the induction of ornithine decarboxylase by the compounds showed a rapid biphasic response, while for creatine kinase brain type (BB) there was a continued increase over 72 h. The efficacy of induction showed that, with both markers, oestradiol gave the highest induction level, followed by tamoxifen or toremifene and then raloxifene. RT-PCR demonstrated that all compounds decreased oestrogen receptor (ER) alpha, ERbeta and ERbeta2 gene expression, 8-24 h after the first dose, suggesting that down-regulation of ER is not the primary cause of the difference in efficacy between these compounds. Using cDNA arrays, expression of 512 genes was examined in the uteri of oestradiol- or tamoxifen-treated rats. Both compounds resulted in the up-regulation of heat-shock protein 27, telomerase-associated protein 1 and secretin. However, most surprising was the marked down-regulation of Wilms' tumour and retinoblastoma genes. We speculate that this may result in a loss of regulation of the transition from the G1 to the S phase in the cell cycle and may make cells more vulnerable to the carcinogenic effects of tamoxifen in this tissue.


Assuntos
Antineoplásicos Hormonais/farmacologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Útero/efeitos dos fármacos , Animais , Técnicas de Cultura de Células , Creatina Quinase/metabolismo , Creatina Quinase Forma BB , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Isoenzimas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Ornitina Descarboxilase/metabolismo , Ovariectomia , Cloridrato de Raloxifeno/farmacologia , Ratos , Ratos Wistar , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tamoxifeno/farmacologia , Toremifeno/farmacologia , Útero/enzimologia
9.
Pharmacol Toxicol ; 88(4): 192-7, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11322177

RESUMO

The bioactivation and cytotoxicity of 1,1-dichloro-2,2,2-trifluoroethane (HCFC-123), a replacement for some ozone-depleting chlorofluorocarbons, were investigated using freshly isolated hepatocytes from non-induced male rats. A time- and concentration-dependent increase in the leakage of lactate dehydrogenase and a concentration-dependent loss of total cellular glutathione were observed in cells incubated with 1, 5 and 10 mM HCFC-123 under normoxic or hypoxic (about 4% O2) conditions. Lactate dehydrogenase leakage was completely prevented by pretreating the cell suspension with the free radical trapper N-t-butyl-alpha-phenylnitrone. The aspecific cytochrome P450 (P450) inhibitor, metyrapone, totally prevented the lactate dehydrogenase leakage from hepatocytes, while two isoform-specific P450 inhibitors, 4-methylpyrazole and troleandomycin (a P450 2E1 and a P450 3A inhibitor, respectively), provided a partial protection against HCFC-123 cytotoxicity. Interestingly, pretreatment of cells with glutathione depletors, such as phorone and diethylmaleate, did not enhance the HCFC-123-dependent lactate dehydrogenase leakage. Two stable metabolites of HCFC-123, 1-chloro-2,2,2-trifluoroethane and 1-chloro-2,2-difluoroethene, were detected by gas chromatography/mass spectrometry analysis of the head space of the hepatocyte incubations carried out under hypoxic and, although at a lower level, also normoxic conditions, indicating that reductive metabolism of HCFC-123 by hepatocytes had occurred. The results overall indicate that HCFC-123 is cytotoxic to rat hepatocytes under both normoxic and hypoxic conditions, due to its bioactivation to reactive metabolites, probably free radicals, and that P450 2E1 and, to a lower extent, P450 3A, are involved in the process.


Assuntos
Clorofluorcarbonetos/metabolismo , Halotano/análogos & derivados , Hepatócitos/metabolismo , Animais , Etano Clorofluorcarbonos , Óxidos N-Cíclicos , Relação Dose-Resposta a Droga , Fomepizol , Cromatografia Gasosa-Espectrometria de Massas , Glutationa/metabolismo , Halotano/análise , Halotano/metabolismo , Hepatócitos/citologia , Técnicas In Vitro , Cetonas/farmacologia , L-Lactato Desidrogenase/metabolismo , Masculino , Maleatos/farmacologia , Óxidos de Nitrogênio/farmacologia , Pirazóis/farmacologia , Piridinas/farmacologia , Ratos , Ratos Wistar , Troleandomicina/farmacologia
10.
Carcinogenesis ; 22(4): 553-7, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11285188

RESUMO

It is now generally accepted that activation of tamoxifen occurs as a result of metabolism to alpha-hydroxytamoxifen. In this study, alpha-hydroxytamoxifen was given to female Wistar/Han rats (0.103 or 0.0103 mmol/kg, intraperitoneally, daily for 5 days). This resulted in liver DNA damage, determined by (32)P-post-labelling, of 3333 +/- 795 or 343 +/- 68 adducts/10(8) nucleotides, respectively (mean +/- SD, n = 4). Following HPLC separation, the retention times of the major alpha-hydroxytamoxifen DNA adducts were similar to those seen following the administration of tamoxifen. However, after rats were treated with alpha-hydroxytamoxifen (0.103 mmol/kg) for 5 days and the animals kept for up to 13 months, no liver tumours developed (0/7 rats), even with phenobarbital promotion (0/5 rats). GST-P foci were detected in the liver, but only after 13 months was their number or area significantly increased over the corresponding controls. When alpha-hydroxytamoxifen was given to female lambda/lacI transgenic rats (0.103 mmol/kg orally for 10 days) and the animals killed 46 days later, there was an approximate 1.8-fold increase in mutation frequency but no significant increase in G:C to T:A transversions as described after tamoxifen treatment. It is concluded that DNA damage alone, resulting from the short-term administration of alpha-hydroxytamoxifen, is not sufficient to initiate liver tumours even with phenobarbital promotion. As with tamoxifen, long-term exposure may be required to allow promotion and progression of transformed cells.


Assuntos
Dano ao DNA , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/genética , Tamoxifeno/análogos & derivados , Tamoxifeno/efeitos adversos , Animais , Animais Geneticamente Modificados , Carcinógenos , Cromatografia Líquida de Alta Pressão , Adutos de DNA/metabolismo , Progressão da Doença , Feminino , Glutationa Transferase/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo , Fígado/patologia , Mutação , Fenobarbital , Antígeno Nuclear de Célula em Proliferação/biossíntese , Ratos , Ratos Wistar , Fatores de Tempo
11.
Toxicol Lett ; 120(1-3): 21-9, 2001 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-11323158

RESUMO

Tamoxifen is one of the most effective drugs to be used in the treatment of women with breast cancer and as a chemopreventive agent in women 'at risk' from this disease. Tamoxifen can be regarded as a paradigm for a new range of selective oestrogen receptor modulators that include toremifene, used in the treatment of metastatic breast cancer and raloxifene, presently approved for use in postmenopausal women for the treatment of osteoporosis. Tamoxifen treatment of women leads to a small increase in the incidence of endometrial cancers. It is important to understand the mechanism for this side effect in order to predict the likely human risk for other drugs of this class. Two such mechanisms have been proposed: (1) conversion of the drug to electrophilic metabolites that damage cellular DNA; and (2) an oestrogen agonist action on the uterus, promoting endogenous lesions. In rats, long-term tamoxifen treatment results in liver cancer via a genotoxic mechanism. However, it seems most likely that, in women treated with tamoxifen, endometrial cancer is related to an oestrogen agonist effect of this drug, promoting uterine cell proliferation.


Assuntos
Neoplasias do Endométrio/induzido quimicamente , Antagonistas de Estrogênios/efeitos adversos , Animais , Dano ao DNA , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Humanos , Ratos , Receptores de Estrogênio/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/efeitos adversos , Tamoxifeno/efeitos adversos
12.
Cancer Lett ; 162(1): 117-22, 2001 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-11121869

RESUMO

Administration of tamoxifen (TAM) (20 mg/kg per day p.o.) for 6 weeks to female lambda/lacI transgenic rats caused a 4-fold increase in mutation frequency (MF) at the lacI gene locus in the livers of dosed animals compared with controls. After cessation of dosing, the MF showed a further increase with time at 2, 12 and 24 weeks, respectively. Phenobarbital promotion of similarly treated animals resulted in no increase in mutation frequency compared with TAM alone. Treatment with phenobarbital or TAM+phenobarbital resulted in time-dependent increases in liver weight compared with the corresponding controls. There was an increase in cell proliferation in the phenobarbital and TAM+phenobarbital groups, and at 24 weeks in the TAM dosed animals compared with controls. There was also a progressive increase in the number of GST-P expressing foci in the livers of TAM and TAM + phenobarbital rats compared with controls. The induction of cell proliferation and GSTP foci in the rat liver by phenobarbital is consistent with its ability to promote tamoxifen-initiated liver tumours in the rat. If the lacI gene is regarded as being representative of the rat genome in general (albeit that the gene is bacterial) the above observations suggest that promotion by tamoxifen confers selective advantage on mutated genes at loci that contribute to the tumour phenotype and that promotion of rat liver tumours by tamoxifen is not dependent simply upon the enhancement of cellular proliferation.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Escherichia coli , Antagonistas de Estrogênios/toxicidade , Neoplasias Hepáticas Experimentais/induzido quimicamente , Fígado/efeitos dos fármacos , Mutagênicos/toxicidade , Fenobarbital/toxicidade , Proteínas Repressoras/genética , Tamoxifeno/toxicidade , Animais , Divisão Celular/efeitos dos fármacos , Feminino , Glutationa Transferase/metabolismo , Repressores Lac , Fígado/patologia , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344
13.
Carcinogenesis ; 21(10): 1851-8, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11023543

RESUMO

Tamoxifen is a hepatic genotoxin in rats and mice but a hepatocarcinogen only in rats. It is not associated with DNA adducts and liver tumours in patients. The proposed major pathway for its bioactivation in rats involves alpha-hydroxylation, O-sulphonation and generation of a carbocation that reacts with DNA. Rat liver microsomes catalyse alpha-hydroxylation at approximately 2- and 4-fold the rate achieved by human and murine liver microsomes, respectively. O-glucuronylation will deactivate alpha-hydroxytamoxifen and compete with sulphonation. Rates of O-sulphonation of alpha-hydroxytamoxifen in hepatic cytosol have been determined by a HPLC assay of substrate-dependent 3'-phosphoadenosine 5'-phosphate production. The rank order of O-glucuronylation in hepatic microsomes was estimated by HPLC-mass spectrometry. The rate of sulphonation of trans-alpha-hydroxytamoxifen (25 microM) in cytosol from adult female Sprague-Dawley rats and CD1 mice was 5.3 +/- 0.8 and 3.9 +/- 0.5 pmol/min/mg protein (mean +/- SD, n = 3), respectively. In cytosol fractions from women aged 40-65 years, the rate was 1.1 +/- 0.4 pmol/min/mg protein (mean +/- SD, n = 6). The K(m) for trans-alpha-hydroxytamoxifen in rat, mouse and human cytosol was 84. 6 +/- 3.8, 81.4 +/- 4.6 and 104.3 +/- 5.6 microM (mean +/- SD, n = 3), respectively; the corresponding V:(max) values were 22.4 +/- 3.4, 17.1 +/- 3.1 and 6.3 +/- 1.9 pmol/min/mg protein. These K:(m) were similar to a value obtained by others using purified rat liver hydroxysteroid sulphotransferase a. Turnover of the cis epimer was too slow for accurate determination of rates. Sulphonation of trans-alpha-hydroxytamoxifen in human uterine cytosol was undetectable. The rank order of O-glucuronylation of trans-alpha-hydroxy- tamoxifen in liver microsomes was human > > mouse > rat. In combination, lower rates of alpha-hydroxylation and O-sulphonation and a higher rate of O-glucuronylation in human liver would protect patients from the formation of tamoxifen-DNA adducts.


Assuntos
Carcinógenos/metabolismo , Adutos de DNA/biossíntese , Glucuronosiltransferase/metabolismo , Fígado/metabolismo , Sulfotransferases/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/metabolismo , Tamoxifeno/farmacocinética , Adulto , Idoso , Animais , Biotransformação , Carcinógenos/farmacocinética , Carcinógenos/toxicidade , Citosol/enzimologia , Citosol/metabolismo , Adutos de DNA/antagonistas & inibidores , Feminino , Humanos , Inativação Metabólica , Cinética , Fígado/enzimologia , Camundongos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Tamoxifeno/toxicidade
14.
Crit Rev Toxicol ; 30(5): 571-94, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11055836

RESUMO

The antiestrogen tamoxifen is widely used in the adjuvant therapy of breast cancers in women and helps to prevent the occurrence of breast tumors in healthy women. However, epidemiological studies have shown tamoxifen treatment to be associated with a 2- to 5-fold increased risk of endometrial cancer. In rats but not in mice, long-term administration of tamoxifen results in an increase in hepatocellular carcinomas. Mechanistically, this occurs through metabolic activation of the drug, mainly by the CYP3A family, to an electrophilic species, that causes DNA damage in target tissues, and subsequently leads to gene mutations. It is controversial whether low levels of DNA damage occur in human uterine tissues, and there is no evidence that this can be causally related to the mechanisms of carcinogenesis. In healthy women, the risk:benefits for the use of tamoxifen is in part related to the risk of developing breast cancer. The results from the carcinogenicity studies in rats do not predict the likelihood that women will develop liver cancer or indeed cancers in other organs. The mechanism of endometrial cancer in women remains unresolved, but the experience with tamoxifen has highlighted the potential problems that need to be addressed in the assessment of future generations of selective estrogen receptor modulators.


Assuntos
Neoplasias da Mama/prevenção & controle , Antagonistas de Estrogênios , Neoplasias Hepáticas Experimentais/induzido quimicamente , Tamoxifeno , Animais , Quimioterapia Adjuvante , Neoplasias do Endométrio/induzido quimicamente , Antagonistas de Estrogênios/efeitos adversos , Antagonistas de Estrogênios/metabolismo , Antagonistas de Estrogênios/uso terapêutico , Feminino , Humanos , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , Testes de Mutagenicidade , Ratos , Medição de Risco , Tamoxifeno/efeitos adversos , Tamoxifeno/metabolismo , Tamoxifeno/uso terapêutico
15.
Biochem Pharmacol ; 60(2): 233-9, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10825468

RESUMO

ATPase transporter proteins are commonly found in the hepatocyte canalicular membrane. Some of these, in particular the multidrug resistance (mdr1b) gene, have been previously demonstrated to be inducible genes. In this study, we found that tamoxifen induced expression of the mdr1b gene in the liver up to 40-fold after 14 days' exposure to tamoxifen in the diet at a concentration of 420 ppm. As tamoxifen and its metabolites are primarily excreted into the bile, we investigated if the increased expression of mdr1b in the liver following tamoxifen exposure had any effect on its excretion in rats. We found that the excretion of tamoxifen and its metabolites into bile was increased from 8 +/- 1% to 51 +/- 18% (mean +/- SD) of an administered dose of 180 nmol/kg over a collection period of 3 hr in rats that had received tamoxifen (35 mg/kg) orally for 12 days (plus a 3-day rest) prior to the experiment. These data suggest that prolonged treatment with tamoxifen may result in lower serum and tumour concentrations, due to a self-mediated enhancement of excretion via mdr1b gene-encoded P-glycoprotein. This may have implications for other drugs sharing the same route of excretion and co-administered with tamoxifen.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Canalículos Biliares/metabolismo , Tamoxifeno/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Antineoplásicos Hormonais/farmacocinética , Antineoplásicos Hormonais/farmacologia , Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Taxa de Depuração Metabólica , Ratos , Ratos Endogâmicos Lew , Tamoxifeno/farmacologia
16.
Carcinogenesis ; 21(4): 793-7, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10753217

RESUMO

Tamoxifen was administered orally to neonatal rats on days 2-5 after birth and the subsequent effects on the uterus were characterized, morphometrically, over the following 12 months. Tamoxifen inhibited development of the uterus and glands in the endometrium, indicating a classical oestrogen antagonist action. Between 24 and 35 months after tamoxifen treatment there was a significant increase in the incidence (26%) of uterine adenocarcinomas and a 9% incidence of squamous cell carcinomas of the vagina/cervix in the absence of any oestrogen agonist effect in the uterus. This demonstrates that an oestrogen agonist effect is not an absolute requirement for the carcinogenic effect of tamoxifen in the reproductive tract of the rat. The unopposed oestrogen agonist effect of tamoxifen on the endometrium may not be the only factor involved in the development of endometrial cancers. It is possible that tamoxifen causes these tumours via a genotoxic mechanism similar to that seen in rat liver. However, using (32)P-post-labelling we failed to find evidence of tamoxifen-induced DNA adducts in the uterus. Tamoxifen may affect hormonal imprinting of oestrogen receptor responses in stem cells of the uterus, causing reproductive tract cancers to arise at a later time, in the same way as has been proposed for diethylstilbestrol. If these rodent data extrapolate to humans, then women who are taking tamoxifen as a chemopreventative may have an increased risk of vaginal/cervical cancer, as well as endometrial cancer.


Assuntos
Antineoplásicos Hormonais/toxicidade , Hiperplasia Endometrial/induzido quimicamente , Neoplasias do Endométrio/induzido quimicamente , Tamoxifeno/toxicidade , Neoplasias Vaginais/induzido quimicamente , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Adutos de DNA/análise , Feminino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar
17.
J Biol Chem ; 274(45): 32008-14, 1999 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-10542232

RESUMO

Tamoxifen acts as a strong estrogen antagonist in human breast but as an estrogen agonist in the uterus. The action of tamoxifen is mediated through estrogen receptors (ERalpha and ERbeta), which bind to a variety of responsive elements, to activate transcription. To examine the role of these varied elements in the response to antiestrogens, we studied the activation of a panel of differing promoters, by these compounds, in human breast, bone, and endometrial derived cell lines. No agonistic activity was observed in breast cells, whereas all antiestrogens, particularly tamoxifen, exhibited agonistic effects in uterine cell lines. All antiestrogens studied were agonistic in co-transfections of a collagenase reporter gene and ERbeta, but tamoxifen alone was agonistic with ERalpha in (uterine) HEC-1-A cells. The ERalpha mediated, agonism of tamoxifen was not observed in primary cultures of human uterine stromal cells, whereas the ERbeta-mediated agonism of all selective estrogen receptor modulators was present. This suggests that the two receptors operate by distinct pathways and that the response of cells to antiestrogens is dependent on the ER subtypes expressed.


Assuntos
Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Transcrição Gênica , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Humanos , Osteoblastos/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Transfecção , Células Tumorais Cultivadas
18.
Carcinogenesis ; 20(10): 2011-6, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10506118

RESUMO

The present study compares the formation of DNA adducts, determined by (32)P-postlabelling, in the livers of rats given tamoxifen and the N-demethylated metabolites N-desmethyltamoxifen and N, N-didesmethyltamoxifen. Results show that after 4 days treatment (0.11 mmol/kg i.p.), similar levels of DNA damage were seen after treatment with either tamoxifen or N-desmethyltamoxifen [109 +/- 40 (n = 3) and 100 +/- 33 (n = 4) adducts/10(8) nucleotides, respectively], even though the concentration of tamoxifen in the livers of tamoxifen-treated rats was about half that of N-desmethyltamoxifen in the N-desmethyltamoxifen-treated animals (51 +/- 16 and 100 +/- 8 nmol/g, respectively). Administration of N, N-didesmethyltamoxifen to rats resulted in a 5-fold lower level of damage (19 adducts/10(8) nucleotides, n = 2). Following (32)P-postlabelling and HPLC, hepatic DNA from rats treated with tamoxifen and its metabolites showed distinctive patterns of adducts. Treatment of rats with N,N-didesmethyltamoxifen gave a major product that co-eluted with one of the minor adduct peaks seen in the livers of rats given tamoxifen. Following dosing with N-desmethyltamoxifen, the major product co-eluted with one of the main peaks seen following treatment of rats with tamoxifen. This suggests that tamoxifen can be metabolically converted to N-desmethyltamoxifen prior to activation. However, analysis of the (32)P-postlabelled products from the reaction between alpha-acetoxytamoxifen and calf thymus DNA showed two main peaks, the smaller one of which ( approximately 15% of the total) also co-eluted with that attributed to N-desmethyltamoxifen. This indicates that N-desmethyltamoxifen and N,N-didesmethyltamoxifen are activated in a similar manner to tamoxifen leading to a complex mixture of adducts. Since an HPLC system does not exist that can fully separate all these (32)P-postlabelled adducts, care has to be taken when interpreting results and determining the relative importance of individual adducts and the metabolites they are derived from in the carcinogenic process.


Assuntos
Adutos de DNA/metabolismo , Fígado/efeitos dos fármacos , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Animais , Bovinos , Cromatografia Líquida de Alta Pressão , Nucleotídeos de Desoxiguanina/metabolismo , Feminino , Fígado/metabolismo , Espectrometria de Massas , Radioisótopos de Fósforo , Ratos , Ratos Endogâmicos F344 , Tamoxifeno/administração & dosagem
19.
Carcinogenesis ; 20(7): 1153-60, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10383884

RESUMO

The anti-oestrogen tamoxifen is widely used for adjuvant therapy in the treatment of women with breast cancer and has a low incidence of serious side-effects. It could also play a role as a breast cancer chemopreventive agent. However, epidemiological studies in both tamoxifen-treated breast cancer patients and in healthy women have shown that treatment results in a small increase in the incidence of endometrial cancers. While the use of tamoxifen in breast cancer patients is clearly justified, the situation for its use as a chemopreventive agent in healthy women is not so clear cut. Reasons for caution come from studies in rats that show that tamoxifen is a genotoxic mutagenic liver carcinogen. Initiation of tumours in the rat is the result of metabolic activation of tamoxifen by CYP enzymes to an electrophile(s) that binds irreversibly to DNA. This is not related to the oestrogen receptor status of the tissue. The extent of DNA damage, detected by 32P-post-labelling or accelerator mass spectrometry, is dependent both on the dose and the length of exposure. Studies have been carried out to see if such binding occurs in the uterine endometrium from tamoxifen-treated women. Results are presently inconclusive, but if such irreversible DNA binding occurs, it is at very low levels. Based on a mechanistic understanding of tamoxifen-induced liver carcinogenesis in the rat, it seems that in humans hepatic DNA damage will be close to the limit of detection by 32P-post-labelling and liver cancer will not be a significant carcinogenic risk. We cannot be certain of the mode of action of tamoxifen that results in the increase in endometrial cancers in treated women but it seems unlikely that this will be associated with a classical genotoxic mechanism.


Assuntos
Anticarcinógenos/efeitos adversos , Antineoplásicos Hormonais/efeitos adversos , Carcinógenos/efeitos adversos , Antagonistas de Estrogênios/efeitos adversos , Tamoxifeno/efeitos adversos , Animais , Anticarcinógenos/metabolismo , Anticarcinógenos/uso terapêutico , Antineoplásicos Hormonais/metabolismo , Antineoplásicos Hormonais/uso terapêutico , Biotransformação , Neoplasias da Mama/prevenção & controle , Dano ao DNA , Desenho de Fármacos , Neoplasias do Endométrio/induzido quimicamente , Neoplasias do Endométrio/epidemiologia , Antagonistas de Estrogênios/metabolismo , Antagonistas de Estrogênios/uso terapêutico , Feminino , Humanos , Incidência , Neoplasias Hepáticas Experimentais/epidemiologia , Masculino , Camundongos , Ratos , Receptores de Estrogênio/efeitos dos fármacos , Medição de Risco , Tamoxifeno/metabolismo , Tamoxifeno/uso terapêutico , Útero/efeitos dos fármacos
20.
Carcinogenesis ; 20(1): 153-60, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9934863

RESUMO

The metabolic formation of a-hydroxytamoxifen, a reactive metabolite of tamoxifen in rat liver, was characterized and quantified in vitro (hepatic microsomal incubations) and in vivo (bile-duct cannulated animals). This minor metabolite was identified by chromatographic and mass spectral comparisons with the authentic compound. The rates of formation of alpha-hydroxytamoxifen in incubations (30 min) of tamoxifen (25 microM) with liver microsomal preparations from women (pool of six), female CD1 mice or female Sprague-Dawley rats, as quantified by liquid chromatography-mass spectrometry (LC-MS), were 1.15+/-0.03, 0.30+/-0.05 and 2.70+/-0.35 pmol/min/mg protein, respectively. Selective inhibition of microsomal P450 indicated that alpha-hydroxylation was catalysed predominantly by CYP3A in humans. Bile-duct cannulated and anaesthetized female rats and mice given [14C]tamoxifen (43 micromol/kg, i.v.) excreted, respectively, 24 and 21% of the administered radioactivity in bile over 5 and 3.5 h. The major radiolabelled biliary metabolite in rats, characterized by LC-MS after enzymic hydrolysis of conjugates, was the glucuronide of 4-hydroxytamoxifen (10% of dose) and only 0.1% of the dose was recovered as alpha-hydroxytamoxifen. After administration of alpha-hydroxytamoxifen (43 micromol/kg, i.v.) to rats, only 1.19% of the administered compound was recovered from a glucuronide metabolite in bile, indicating a possible 0.84% alpha-hydroxylation of tamoxifen in vivo. There was, however, no indication of the presence in bile of either O-sulphonate or glutathione conjugates derived from alpha-hydroxytamoxifen. This study shows for the first time that alpha-hydroxytamoxifen can be glucuronylated in rat liver. Whereas sulphonation results in electrophilic genotoxic intermediates, glucuronidation may represent a means of detoxifying alpha-hydroxytamoxifen.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Microssomos Hepáticos/enzimologia , Mutagênicos/farmacocinética , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacocinética , Adulto , Animais , Bile/metabolismo , Biotransformação , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/metabolismo , Feminino , Glucuronatos/metabolismo , Humanos , Hidroxilação , Inativação Metabólica , Camundongos , Pessoa de Meia-Idade , Estrutura Molecular , Mutagênicos/análise , Mutagênicos/toxicidade , Oxirredutases N-Desmetilantes/metabolismo , Ratos , Ratos Sprague-Dawley , Sulfonas/metabolismo , Tamoxifeno/análise , Tamoxifeno/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...