Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Reprod Toxicol ; 108: 1-9, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34974146

RESUMO

Tamoxifen, a selective non-steroidal estrogen receptor modulator, is the standard adjuvant endocrine treatment for breast cancer. Since information on the risk of using tamoxifen during pregnancy is still scarce, this study evaluated whether the in utero and lactational treatment with this drug could compromise reproductive and behavioural parameters in male offspring. Pregnant Wistar rats were exposed to three doses of tamoxifen (0.12; 0.6; 3 µg/kg), by gavage, from gestational day 15 to lactational day 20. Tamoxifen exposure did not alter the anogenital distance in the male offspring; however, there was a significant increase in the body weight in the 0.12 µg/kg dose and a decrease in the 0.6 µg/kg dose. The male offspring treated with the highest dose exhibited a delay in the onset of puberty, evidenced by an increase in the age of preputial separation. Regarding sperm parameters, there was an increase in the sperm count in the cauda epididymis in the intermediate and highest dose groups, in addition to an increase in the number of static sperm and a decrease in the progressive sperm in the same groups. Moreover, an increase in the number of hyperplasia of the epithelial clear cells was observed in the epididymis. In conclusion, the present study demonstrated that maternal exposure to tamoxifen compromised the installation of puberty of the male offspring and the maturation of the epididymis, affecting sperm storage and motility in the adult life.


Assuntos
Comportamento Animal/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Espermatozoides/efeitos dos fármacos , Tamoxifeno/toxicidade , Animais , Epididimo/efeitos dos fármacos , Epididimo/crescimento & desenvolvimento , Feminino , Hipotálamo/citologia , Lactação , Masculino , Troca Materno-Fetal , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Gravidez , Ratos Wistar , Receptores Androgênicos/metabolismo , Maturidade Sexual/efeitos dos fármacos , Contagem de Espermatozoides , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatozoides/fisiologia
2.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34911761

RESUMO

Arterial remodeling is an important adaptive mechanism that maintains normal fluid shear stress in a variety of physiologic and pathologic conditions. Inward remodeling, a process that leads to reduction in arterial diameter, plays a critical role in progression of such common diseases as hypertension and atherosclerosis. Yet, despite its pathogenic importance, molecular mechanisms controlling inward remodeling remain undefined. Mitogen-activated protein kinases (MAPKs) perform a number of functions ranging from control of proliferation to migration and cell-fate transitions. While the MAPK ERK1/2 signaling pathway has been extensively examined in the endothelium, less is known about the role of the MEKK3/ERK5 pathway in vascular remodeling. To better define the role played by this signaling cascade, we studied the effect of endothelial-specific deletion of its key upstream MAP3K, MEKK3, in adult mice. The gene's deletion resulted in a gradual inward remodeling of both pulmonary and systematic arteries, leading to spontaneous hypertension in both vascular circuits and accelerated progression of atherosclerosis in hyperlipidemic mice. Molecular analysis revealed activation of TGFß-signaling both in vitro and in vivo. Endothelial-specific TGFßR1 knockout prevented inward arterial remodeling in MEKK3 endothelial knockout mice. These data point to the unexpected participation of endothelial MEKK3 in regulation of TGFßR1-Smad2/3 signaling and inward arterial remodeling in artery diseases.


Assuntos
Hipertensão Pulmonar/patologia , MAP Quinase Quinase Quinase 1/metabolismo , MAP Quinase Quinase Quinase 3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Remodelação Vascular/fisiologia , Animais , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Genótipo , Membro Posterior/irrigação sanguínea , Células Endoteliais da Veia Umbilical Humana , Humanos , Hipertensão Pulmonar/metabolismo , Isquemia , MAP Quinase Quinase Quinase 1/genética , MAP Quinase Quinase Quinase 3/genética , Camundongos , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Transdução de Sinais , Tamoxifeno/toxicidade , Fator de Crescimento Transformador beta/genética
3.
Recent Pat Anticancer Drug Discov ; 14(3): 280-291, 2019 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-31538904

RESUMO

BACKGROUND: Due to the low water solubility of Docetaxel (DTX), it is formulated with ethanol and Tween 80 with lots of side effects. For this reason, special attention has been paid to formulate it in new drug nano-carriers. OBJECTIVE: The goal of this study was to evaluate the safety, antitumor activity and tissue distribution of the novel synthesized Raloxifene (RA) targeted polymeric micelles. METHODS: DTX-loaded RA-targeted polymeric micelles composed of poly(styrene-maleic acid)- poly(amide-ether-ester-imide)-poly(ethylene glycol) (SMA-PAEE-PEG) were prepared and their antitumor activity was studied in MC4-L2 tumor-bearing mice compared with non-targeted micelles and free DTX. Safety of the micelles was studied by Hematoxylin and Eosin (H&E) staining of tumors and major organs of the mice. The drug accumulation in the tumor and major organs was measured by HPLC method. RESULTS: The results showed better tumor growth inhibition and increased survival of mice treated with DTX-loaded in targeted micelles compared to the non-targeted micelles and free DTX. Histopathological studies, H&E staining of tumors and immunohistochemical examination showed the potential of DTX-loaded RA-targeted micelles to inhibit tumor cells proliferation. The higher accumulation of the DTX in the tumor tissue after injection of the micelles compared to the free DTX may indicate the higher uptake of the targeted micelles by the G-Protein-Coupled Estrogen Receptors (GPER). CONCLUSION: The results indicate that RA-conjugated polymeric micelles may be a strong and effective drug delivery system for DTX therapy and uptake of the drug into tumor cells, and overcome the disadvantages and side effects of conventional DTX.


Assuntos
Docetaxel/farmacocinética , Docetaxel/toxicidade , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Cloridrato de Raloxifeno/farmacocinética , Cloridrato de Raloxifeno/toxicidade , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Docetaxel/administração & dosagem , Portadores de Fármacos/administração & dosagem , Feminino , Humanos , Maleatos/administração & dosagem , Maleatos/química , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Micelas , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Poliestirenos/administração & dosagem , Poliestirenos/química , Distribuição Aleatória , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem , Moduladores Seletivos de Receptor Estrogênico/farmacocinética , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Distribuição Tecidual
4.
Arterioscler Thromb Vasc Biol ; 38(6): 1346-1357, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29650694

RESUMO

OBJECTIVE: Estrogens exert beneficial effect on the blood vascular system. However, their role on the lymphatic system has been poorly investigated. We studied the protective effect of the 17ß estradiol-the most potent endogenous estrogen-in lymphedema-a lymphatic dysfunction, which results in a massive fluid and fat accumulation in the limb. APPROACH AND RESULTS: Screening of DNA motifs able to mobilize ERs (estrogen receptors) and quantitative real-time polymerase chain reaction analysis revealed that estradiol promotes transcriptional activation of lymphangiogenesis-related gene expression including VEGF (vascular endothelial growth factor)-D, VEGFR (VEGF receptor)-3, lyve-1, and HASs (hyaluronan synthases). Using an original model of secondary lymphedema, we observed a protective effect of estradiol on lymphedema by reducing dermal backflow-a representative feature of the pathology. Blocking ERα by tamoxifen-the selective estrogen modulator-led to a remodeling of the lymphatic network associated with a strong lymphatic leakage. Moreover, the protection of lymphedema by estradiol treatment was abrogated by the endothelial deletion of the receptor ERα in Tie2-Cre; ERαlox/lox mice, which exhibit dilated lymphatic vessels. This remodeling correlated with a decrease in lymphangiogenic gene expression. In vitro, blocking ERα by tamoxifen in lymphatic endothelial cells decreased cell-cell junctions, inhibited migration and sprouting, and resulted in an inhibition of Erk but not of Akt phosphorylation. CONCLUSIONS: Estradiol protection from developing lymphedema is mediated by an activation of its receptor ERα and is antagonized by tamoxifen. These findings reveal a new facet of the estrogen influence in the management of the lymphatic system and provide more evidence that secondary lymphedema is worsened by hormone therapy.


Assuntos
Linfedema Relacionado a Câncer de Mama/prevenção & controle , Estradiol/administração & dosagem , Receptor alfa de Estrogênio/agonistas , Terapia de Reposição Hormonal , Linfangiogênese/efeitos dos fármacos , Vasos Linfáticos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Linfedema Relacionado a Câncer de Mama/metabolismo , Linfedema Relacionado a Câncer de Mama/patologia , Linfedema Relacionado a Câncer de Mama/fisiopatologia , Modelos Animais de Doenças , Implantes de Medicamento , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Vasos Linfáticos/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovariectomia , Fosforilação , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Tamoxifeno/toxicidade
5.
Neurotoxicology ; 56: 64-75, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27395752

RESUMO

Selective estrogen receptor modulators (SERMs) are a class of therapeutic chemicals which present tissue-specific estrogen receptor modulating activity. Neonatal exposure to SERMs has been reported to adversely affect central nervous system development, however, mechanism and involvement of hypothalamic kisspeptin neurone in this impairment remains undetermined. To clarify this uncertainty, neonates from female Donryu rats were subcutaneously injected with raloxifene (RLX) at 0.1, 1, and 10mg/kg or tamoxifen (TMX) at 10mg/kg on postnatal day 0, and then hypothalamic KiSS1 mRNA expression and gonadotropin levels were investigated during young adulthood and estrous cycling was monitored until middle age. Treatment with RLX or TMX at 10mg/kg significantly depressed luteinizing hormone surge levels and KiSS1 mRNA expression in the anteroventral periventricular nucleus (AVPV), the control center of estrous cyclicity. The 10mg/kg TMX group also showed decreased levels of follicle-stimulating hormone and KiSS1 mRNA expression in the arcuate nucleus (ARC). Early cessation of normal estrous cycling was observed in the 10mg/kg RLX group, while the estrous cycle in the 10mg/kg TMX group had ceased by the start of the analysis. The same dose of tamoxifen or raloxifene had either weak-estrogenic or anti-estrogenic activity on the uterus, respectively; however, treatment in adulthood with both SERMs did not affect KiSS1 mRNA expression in either the AVPV or ARC in the present study. These results indicate that neonatal exposure to SERMs could disrupt neuroendocrine development and postnatal reproductive function through the alteration of kisspeptin neurons.


Assuntos
Deficiências do Desenvolvimento/induzido quimicamente , Doenças do Sistema Endócrino/induzido quimicamente , Hipotálamo/patologia , Kisspeptinas/metabolismo , Neurônios/metabolismo , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Fatores Etários , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Deficiências do Desenvolvimento/patologia , Modelos Animais de Doenças , Doenças do Sistema Endócrino/patologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Ciclo Estral/efeitos dos fármacos , Feminino , Hormônios/metabolismo , Hipotálamo/efeitos dos fármacos , Kisspeptinas/genética , Neurônios/efeitos dos fármacos , Ovariectomia , Gravidez , Progesterona/farmacologia , Cloridrato de Raloxifeno/farmacologia , Ratos , Tamoxifeno/farmacologia
6.
Toxicol Lett ; 253: 27-35, 2016 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-27108949

RESUMO

Exposure to environmental endocrine active compounds correlates with altered susceptibility to disease in human populations. Chemical risk assessment is single compound based, although exposure often takes place as heterogeneous mixtures of man-made and natural substances within complex matrices like diet. Here we studied whether the effects of cadmium and enterolactone on endocrine endpoints in dietary exposure can be predicted based on pure compound effects. Ovariectomized estrogen reporter ERE-luciferase (ERE-luc) mice were maintained on diets that intrinsically contain increasing concentrations of cadmium and enterolactone precursors for three and 21 days. The activation of the ERE-luc, epidermal growth factor receptor (EGFR), mitogen activated protein kinase (MAPK)-ERK1/2, and classical estrogen responses were measured. Interactions between the diets and endogenous hormone were evaluated by challenging the animals with 17ß-estradiol. Compared to animals on basal purified diet, mice consuming experimental diets were exposed to significantly higher levels of cadmium and enterolactone, yet the exposure remained comparable to typical human dietary intake. Surprisingly, we could not detect effects on endpoints regulated by pure enterolactone, such as ERE-luc activation. However, cadmium accumulation in the liver was accompanied with activation of EGFR and MAPK-ERK1/2 in line with our earlier CdCl2 studies. Further, attenuation of 17ß-estradiol-induced ERE-luc response in liver by experimental diets was observed. Our findings indicate that the exposure context can have substantial effects on the activity of endocrine active compounds in vivo. Thus, whenever possible, a context that mimics human exposure should be tested along with pure compounds.


Assuntos
4-Butirolactona/análogos & derivados , Cádmio/toxicidade , Dieta/efeitos adversos , Receptores ErbB/efeitos dos fármacos , Estrogênios/metabolismo , Lignanas/toxicidade , Fígado/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Transdução de Sinais/efeitos dos fármacos , Útero/efeitos dos fármacos , 4-Butirolactona/administração & dosagem , 4-Butirolactona/toxicidade , Animais , Pão/efeitos adversos , Cádmio/administração & dosagem , Receptores ErbB/metabolismo , Feminino , Linho/toxicidade , Genes Reporter , Lignanas/administração & dosagem , Fígado/enzimologia , Luciferases/biossíntese , Luciferases/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovariectomia , Elementos de Resposta , Medição de Risco , Sementes/toxicidade , Fatores de Tempo , Triticum/toxicidade , Útero/metabolismo
7.
J Appl Toxicol ; 36(6): 853-62, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26584595

RESUMO

Tamoxifen is a widely used anticancer drug with both an estrogen agonist and antagonist effect. This study focused on its endocrine disrupting effect, and overall environmental significance. Zebrafish embryos were exposed to different concentrations (0.5, 5, 50 and 500 µg l(-1) ) of tamoxifen for 96 h. The results showed a complex effect of tamoxifen on zebrafish embryo development. For the 500 µg l(-1) exposure group, the heart rate was decreased by 20% and mild defects in caudal fin and skin were observed. Expressions of a series of genes related to endocrine and morphological changes were subsequently tested through quantitative real-time polymerase chain reaction. Bisphenol A as a known estrogen was also tested as an endocrine-related comparison. Among the expression of endocrine-related genes, esr1, ar, cyp19a1b, hsd3b1 and ugt1a1 were all increased by tamoxifen exposure, similar to bisphenol A. The cyp19a1b is a key gene that controls estrogen synthesis. Exposure to 0.5, 5, 50 and 500 µg l(-1) of tamoxifen caused upregulation of cyp19a1b expression to 152%, 568%, 953% and 2024% compared to controls, higher than the effects from the same concentrations of bisphenol A treatment, yet vtg1 was suppressed by 24% from exposure to 500 µg l(-1) tamoxifen. The expression of metabolic-related genes such as cyp1a, cyp1c2, cyp3a65, gpx1a, gstp1, gsr and genes related to observed morphological changes such as krt17 were also found to be upregulated by high concentrations of tamoxifen. These findings indicated the potential environmental effect of tamoxifen on teleost early development. Copyright © 2015 John Wiley & Sons, Ltd.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Tamoxifeno/toxicidade , Poluentes Químicos da Água/toxicidade , Peixe-Zebra , Nadadeiras de Animais/anormalidades , Nadadeiras de Animais/efeitos dos fármacos , Nadadeiras de Animais/embriologia , Animais , Antineoplásicos Hormonais/toxicidade , Aromatase/genética , Aromatase/metabolismo , Embrião não Mamífero/anormalidades , Embrião não Mamífero/enzimologia , Embrião não Mamífero/metabolismo , Estrogênios não Esteroides/toxicidade , Frequência Cardíaca/efeitos dos fármacos , Larva/efeitos dos fármacos , Larva/crescimento & desenvolvimento , Larva/metabolismo , Concentração Osmolar , Distribuição Aleatória , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Pele/efeitos dos fármacos , Pele/embriologia , Anormalidades da Pele/induzido quimicamente , Anormalidades da Pele/embriologia , Anormalidades da Pele/veterinária , Teratogênicos/toxicidade , Vitelogeninas/antagonistas & inibidores , Vitelogeninas/genética , Vitelogeninas/metabolismo , Peixe-Zebra/anormalidades , Peixe-Zebra/embriologia , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/agonistas , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Zigoto/efeitos dos fármacos , Zigoto/crescimento & desenvolvimento , Zigoto/metabolismo
8.
Drug Metab Pers Ther ; 31(1): 35-40, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26581074

RESUMO

BACKGROUND: Metabolite profiles of ospemifene, a novel nonsteroidal selective estrogen receptor modulator, were surveyed as part of its development. METHODS: The pharmacokinetics of ospemifene and its two major, pharmacologically active metabolites 4-hydroxyospemifene and 4'-hydroxyospemifene, was elucidated in studies of volunteer humans given various doses of ospemifene and in experiments of several animal species (rat, mouse, dog, and cynomolgus monkey), which had been used either for pharmacological or toxicological studies of ospemifene. Metabolites produced in in vitro human and animal liver preparations were compared between species and with the metabolite profiles in the in vivo investigations. RESULTS: Considerable interspecies differences were observed in the metabolite profiles and quantities. The major human metabolite, 4-hydroxyospemifene, was produced in substantial amounts both in vitro and in vivo in most animal species, except dog, and thus the exposure to this metabolite seems adequate in the most important toxicology species, the rat and the cynomolgus monkey. 4'-Hydroxyospemifene was equally abundant in vitro and in vivo metabolite in mice and dogs, and consequently, its contribution to the total exposure of ospemifene-related activity would be adequately covered in animal experiments. Other ospemifene metabolites were variably detected in different species, but probably they are not of consequence to pharmacology or toxicology of ospemifene. CONCLUSIONS: Overall, there are quantitative and also some qualitative differences in the metabolism of ospemifene in different species. Generally, in vitro metabolite profiles were predictive for in vivo profiles. The contribution of two major hydroxyl metabolites to activity and toxicity of ospemifene is adequately covered by at least some animal species.


Assuntos
Microssomos Hepáticos/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/análogos & derivados , Animais , Cães , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Especificidade da Espécie , Tamoxifeno/metabolismo , Tamoxifeno/toxicidade
9.
Environ Toxicol ; 31(12): 1834-1850, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26303313

RESUMO

Tamoxifen is an endocrine-active pharmaceutical (EAP) that is used world-wide. Because tamoxifen is a ubiquitous pharmaceutical and interacts with estrogen receptors, a case study was conducted with this compound to (1) determine effects on reproductive endpoints in a nontarget species (i.e., a fish), (2) compare biologically-active metabolites across species, (3) assess whether in vitro assays predict in vivo results, and (4) investigate metabolomic profiles in tamoxifen-treated fish to better understand the biological mechanisms of tamoxifen toxicity. In reproductive assays, tamoxifen exposure caused a significant reduction in egg production and significantly increased ovarian aromatase activity in spawning adult cunner fish (Tautogolabrus adspersus). In plasma from tamoxifen-exposed cunner, the predominant metabolite was 4-hydroxytamoxifen, while in rats it was N-desmethyltamoxifen. Because 4-hydroxytamoxifen is a more biologically active metabolite than N-desmethyltamoxifen, this difference could result in a different level of risk for the two species. The results of in vitro assays with fish hepatic microsomes to assess tamoxifen metabolism did not match in vivo results, indicating probable differences in excretion of tamoxifen metabolites in fish compared with rats. For the first time, a complete in vitro characterization of the metabolism of tamoxifen using fish microsomes is presented. Furthermore, a metabolomic investigation of cunner gonad extracts demonstrates that tamoxifen alters the biochemical profile in this nontarget species. Understanding the consequence of tamoxifen exposure in nontarget species, and assessing the discrepancies between sex- and species-mediated endpoints, is a step toward understanding how to accurately assess the risks posed by EAPs, such as tamoxifen, in the aquatic environment. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1834-1850, 2016.


Assuntos
Perciformes/fisiologia , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Tamoxifeno/toxicidade , Poluentes Químicos da Água/toxicidade , Animais , Bioensaio , Ovos , Exposição Ambiental , Feminino , Microssomos Hepáticos/metabolismo , Reprodução/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/metabolismo
10.
ChemMedChem ; 10(8): 1403-12, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26076185

RESUMO

In this study, a series of new 4,4'-diaminotriphenylmethanes was efficiently synthesized from aromatic aldehydes and 2,5-dimethoxybenzenamine under microwave irradiation in the presence of Sc(OTf)3 as a catalyst. Antiproliferative activity was assessed by using the MCF-7 estrogen receptor (ER)-positive breast cancer cell line, and antagonist/agonist transcriptional activities were determined. Docking studies and competition studies of triphenylmethanes and radiolabeled estradiol determined that these compounds do not bind the ER, indicating that triphenylmethane-induced changes in proliferative and transcriptional activities differ from conventional mechanisms of action triggered by other selective ER modulators.


Assuntos
Metano/química , Receptores de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/síntese química , Sítios de Ligação , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Metano/síntese química , Metano/toxicidade , Simulação de Acoplamento Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Estrogênio/química , Receptores de Estrogênio/genética , Moduladores Seletivos de Receptor Estrogênico/química , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Ativação Transcricional/efeitos dos fármacos
11.
Biochim Biophys Acta ; 1852(5): 1038-48, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25703139

RESUMO

Hernia is a disease with defects in collagen synthesis/metabolism. However, the underlying mechanisms for hernia formation have not been fully defined. Tamoxifen is a selective estrogen receptor modulator and used for patients with breast cancer. Tamoxifen also has pleiotropic and side effects. Herein, we report that tamoxifen treatment resulted in an appearance of a large bulge in the low abdomen between the hind legs in male but not in female mice. The autopsy demonstrated that the low abdominal wall was broken and a large amount of intestine herniated out of the abdominal cavity. Histological analysis indicated that tamoxifen caused structural abnormalities in the low abdominal wall which were associated with decreased type II collagen content. Furthermore, we determined increased matrix metalloproteinase-2 (MMP-2) and MMP-13 expression in the tissue. In vitro, tamoxifen induced MMP-2 and MMP-13 expression in fibroblasts. The promoter activity analysis and ChIP assay demonstrate that induction of MMP-13 expression was associated with activation of JNK-AP-1 and ERK1/2 signaling pathways while induction of MMP-2 expression was related to activation of the ERK1/2 signaling pathway. Taken together, our study establishes a novel murine hernia model, defines a severe side effect of tamoxifen, and suggests a caution to male patients receiving tamoxifen treatment.


Assuntos
Hérnia/genética , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 2 da Matriz/genética , Tamoxifeno/farmacologia , Parede Abdominal/patologia , Animais , Western Blotting , Colágeno Tipo II/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hérnia/induzido quimicamente , Hérnia/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Fatores Sexuais , Tamoxifeno/toxicidade
12.
Hum Exp Toxicol ; 34(3): 284-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24925364

RESUMO

OBJECTIVE: Clomiphene citrate (CC) is a selective estrogen receptor modulator and is used for the treatment of in vitro fertilization, intracytoplasmic sperm injection, intrauterine insemination, and so on. In this study, sister chromatid exchanges (SCEs) and cell cycle delays were analyzed to investigate genotoxicity and cytotoxicity of CC in peripheral blood lymphocytes of healthy women. METHODS: Human peripheral blood lymphocytes obtained from two donors were used to detect genotoxicity and cytotoxicity of CC. Lymphocytes were treated with various concentrations (0.40, 0.80, 1.60, and 3.20 µg/ml) of CC. A negative (distilled water) and a positive control (mitomycin-C = 0.20 µg/ml) were also used simultaneously with test substance-treated cultures. SCEs and cell division delays were measured from 25 cells and 100 cells perdonor, respectively. RESULTS: CC significantly increased the mean SCE value at all concentrations compared with the negative control. This increase was found to be dose dependent (r = 0.83) and at the highest concentration, nearly two times higher increase was observed than the negative control. However, replication index was not affected by the CC treatment. CONCLUSION: The present study shows that CC is genotoxic for human lymphocytes in vitro. Further investigations, especially in vivo are now needed in different test organisms to clarify the genotoxic activity of CC, which should also help to better understand genotoxic mechanism of this ovulation-stimulating drug.


Assuntos
Clomifeno/toxicidade , Linfócitos/efeitos dos fármacos , Mutagênicos/toxicidade , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Adulto , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Humanos , Linfócitos/citologia , Troca de Cromátide Irmã , Adulto Jovem
13.
Artigo em Inglês | MEDLINE | ID: mdl-24189048

RESUMO

Clomiphene citrate (CC) is a selective estrogen-receptor modulator that is primarily used to enhance follicular development in women receiving in vitro fertilization (IVF) treatment. Although some studies suggested large increases in ovarian cancer risk related to fertility medications, this association has not been confirmed in other studies. Whether there could be a residual, small risk is still an open question. It is known that genomic instability and multiple genetic changes may be required in carcinogenesis. Genomic instability such as single-base changes, chromosomal rearrangements or aneuploidy may accelerate this process. Genomic instability is not only central to carcinogenesis, but it is also a factor in some neurodegenerative diseases such as amyotrophic lateral sclerosis or the neuromuscular disease myotonic dystrophy. For these reasons, this study was planned to examine genotoxic effects of CC in human lymphocytes by use of the chromosome aberration (CA) assay, the micronucleus (MN) test, the comet assay, and the test for bacterial mutagenicity in Salmonella typhimurium strains TA98 and TA100 (Ames test). Concentrations of 0.40, 0.80, 1.60, and 3.20µg/ml of CC significantly increased the frequency of chromosomal aberrations (p<0.01 and p<0.001) and micronuclei (p<0.05, p<0.01 and p<0.001) in cultured human lymphocytes, and of DNA damage (tail length, p<0.05, except 0.80µg/ml) in isolated lymphocytes compared with their respective controls. The highest CC concentration at 24h and highest two concentrations after the 48-h treatment significantly decreased the mitotic index. The Ames test showed that the concentrations of CC used in this study induced neither base-pair substitutions nor frame-shift mutations in S. typhimurium strains TA98 and TA100.


Assuntos
Aberrações Cromossômicas , Clomifeno/toxicidade , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Adulto , Dano ao DNA , Feminino , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/ultraestrutura , Micronúcleos com Defeito Cromossômico , Testes de Mutagenicidade
14.
Curr Med Chem ; 20(33): 4177-84, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23895678

RESUMO

Ormeloxifene is a non-steroidal Selective Estrogen Receptor Modulator (SERM) that is used as an oral contraceptive. Recent studies have shown its potent anti-cancer activities in breast, head and neck, and chronic myeloid leukemia cells. Several in vivo and clinical studies have reported that ormeloxifene possesses an excellent therapeutic index and has been well-tolerated, without any haematological, biochemical or histopathological toxicity, even with chronic administration. A reasonably long period of time and an enormous financial commitment are required to develop a lead compound into a clinically approved anti-cancer drug. For these reasons and to circumvent these obstacles, ormeloxifene is a promising candidate on a fast track for the development or repurposing established drugs as anti-cancer agents for cancer treatment. The current review summarizes recent findings on ormeloxifene as an anti-cancer agent and future prospects of this clinically safe pharmacophore.


Assuntos
Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Benzopiranos/toxicidade , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Benzopiranos/química , Benzopiranos/uso terapêutico , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Ensaios Clínicos como Assunto , Humanos , Neoplasias/tratamento farmacológico , Moduladores Seletivos de Receptor Estrogênico/química , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico
15.
Toxicol Pathol ; 41(7): 1001-10, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23416963

RESUMO

Bazedoxifene acetate (BZA) is a selective estrogen receptor modulator that is approved in a number of countries for the prevention and/or treatment of osteoporosis in postmenopausal women. To assess carcinogenic potential, BZA was administered ad libitum in the diet to male and female rats for 2 years. The achieved mean dosages of BZA were approximately 1.31 to 56.9 mg/kg/day at dietary concentrations of 0.003% to 0.1%. BZA treatment resulted in a reduction and a delayed onset in total tumor burden in both male and female rats. Survival rates were enhanced due to decreased pituitary and mammary tumors and decreased body weight gain in BZA-treated animals compared with controls. In male rats only, an increase in renal tubular tumors was observed. The greater increase in tumor incidence in male rats given BZA was associated with the increased survival and increased time for development of late onset tumors. These findings are consistent with a non-genotoxic mechanism, unique to male rats, that involves test article-induced corticomedullary mineralization, renal tubular injury, and exacerbation of naturally occurring chronic progressive nephropathy in aged male rats that led to a sequela of proliferative changes and tumor formation.


Assuntos
Indóis/toxicidade , Neoplasias Renais/induzido quimicamente , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Análise de Variância , Animais , Peso Corporal/efeitos dos fármacos , Testes de Carcinogenicidade , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Indóis/administração & dosagem , Masculino , Ratos , Insuficiência Renal Crônica/induzido quimicamente , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem , Análise de Sobrevida
16.
Arthritis Rheum ; 65(3): 780-91, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23280471

RESUMO

OBJECTIVE: To ascertain whether engineered expression of kallikreins within the kidneys, using an inducible Cre/loxP system, can ameliorate murine lupus nephritis. METHODS: In mice with a lupus-prone genetic background, we engineered the expression of tamoxifen-inducible Cre recombinase under the control of a kidney-specific promoter whose activation initiates murine kallikrein-1 expression within the kidneys. These transgenic mice were injected with either tamoxifen or vehicle at age 2 months and then were monitored for 8 months for kallikrein expression and disease. RESULTS: Elevated expression of kallikrein was detected in the kidney and urine of tamoxifen-injected mice but not in controls. At age 10 months, all vehicle-injected mice developed severe lupus nephritis, as evidenced by increased proteinuria (mean ± SD 13.43 ± 5.65 mg/24 hours), increased blood urea nitrogen (BUN) and serum creatinine levels (39.86 ± 13.45 mg/dl and 15.23 ± 6.89 mg/dl, respectively), and severe renal pathology. In contrast, the tamoxifen-injected mice showed significantly reduced proteinuria (6.6 ± 4.12 mg/24 hours), decreased BUN and serum creatinine levels (15.71 ± 8.17 mg/dl and 6.64 ± 3.39 mg/dl, respectively), and milder renal pathology. Tamoxifen-induced up-regulation of renal kallikrein expression increased nitric oxide production and dampened renal superoxide production and inflammatory cell infiltration, alluding to some of the pathways through which kallikreins may be operating within the kidneys. CONCLUSION: Local expression of kallikreins within the kidney has the capacity to dampen lupus nephritis, possibly by modulating inflammation and oxidative stress.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Túbulos Renais/fisiologia , Nefrite Lúpica/genética , Estresse Oxidativo/fisiologia , Calicreínas Teciduais/genética , Animais , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Integrases/genética , Túbulos Renais/citologia , Óperon Lac/genética , Nefrite Lúpica/metabolismo , Nefrite Lúpica/fisiopatologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos , Camundongos Transgênicos , Óxido Nítrico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Superóxidos/metabolismo , Tamoxifeno/toxicidade , Calicreínas Teciduais/metabolismo
17.
J Cell Physiol ; 228(4): 724-33, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22949219

RESUMO

Bazedoxifene Acetate (BZA) is a selective estrogen receptor modulator (SERM) that is approved for the prevention and/or treatment of osteoporosis in postmenopausal women. To assess for carcinogenic potential, BZA was administered ad libitum in the diet to rats for 2 years. BZA caused an increase in benign ovarian tumors in female rats and decreased incidences of mammary tumors (females) and pituitary tumors (males and females). In addition, BZA provided a significant survival benefit at all dosages tested, which correlated with a significant reduction in pituitary and mammary gland tumors and decreased body weight gain (both genders). Additional studies were subsequently conducted in rats and monkeys to further explore the mechanisms likely responsible for the observed effects. Results from studies in hypophysectomized and chemically castrated female rats indicated that BZA did not directly stimulate formation of ovarian cysts, but an intact pituitary was required for cyst formation. Further, BZA increased estradiol concentrations in rats and monkeys. In monkeys, BZA increased concentrations of luteinizing hormone (LH) after onset of treatment and prohibited the preovulatory surge of LH until after cessation of treatment. These hormonal changes suggest that BZA inhibited both the positive and negative feedback effects of estrogen on gonadotropins and the resulting increase in LH caused formation and persistence of ovarian cysts, which eventually transformed into benign ovarian granulosa cell tumors in the rat carcinogenicity study. These results also suggest that the reductions in pituitary and mammary gland tumors were attributed to BZA-related antagonism of endogenous estrogens at the estrogen receptors.


Assuntos
Carcinógenos/farmacologia , Indóis/farmacologia , Receptores de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Testes de Carcinogenicidade/métodos , Carcinógenos/toxicidade , Cistos/induzido quimicamente , Cistos/metabolismo , Cistos/patologia , Estradiol/metabolismo , Estrogênios/metabolismo , Feminino , Contaminação de Alimentos , Haplorrinos , Indóis/toxicidade , Hormônio Luteinizante/metabolismo , Masculino , Neoplasias Ovarianas/induzido quimicamente , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Hipófise/patologia , Neoplasias Hipofisárias/induzido quimicamente , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Ratos , Ratos Sprague-Dawley , Moduladores Seletivos de Receptor Estrogênico/toxicidade
18.
Gastroenterology ; 142(1): 21-24.e7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22001866

RESUMO

Tamoxifen, a selective estrogen receptor modulator, is widely used in research and clinically in patients. We find that treatment of normal mice with a single ≥3 mg/20 g body weight dose of tamoxifen leads to apoptosis of >90% of all gastric parietal cells (PCs) and metaplasia of zymogenic chief cells within 3 days. Remarkably, gastric histology returns to nearly normal by 3 weeks. Tamoxifen toxicity occurs by oral and intraperitoneal administration, in both sexes, in multiple strains, and does not depend on estrogen, though acid secretion inhibition is partially protective. Thus, substantial gastric toxicity is a heretofore unappreciated tamoxifen side effect.


Assuntos
Celulas Principais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Tamoxifeno/toxicidade , Administração Oral , Animais , Atrofia , Celulas Principais Gástricas/patologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Injeções Intraperitoneais , Integrases/genética , Óperon Lac , Masculino , Metaplasia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Parietais Gástricas/patologia , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem , Especificidade da Espécie , Tamoxifeno/administração & dosagem , Fatores de Tempo
19.
Toxicol Pathol ; 40(1): 55-61, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22021168

RESUMO

Here, we report the effects of estrogen and the selective estrogen receptor modulator (SERM) levormeloxifene on adrenocortical measures in ovariectomized female cynomolgus monkeys (Macaca fascicularis). Animals were randomized into one of five treatment groups, each containing 23 to 26 animals: (1) placebo, (2) 0.016 mg/kg 17ß-estradiol (E(2)), (3) 0.5 mg/kg levormeloxifene (L(1)), (4) 1.0 mg/kg levormeloxifene (L(2)), and (5) 5.0 mg/kg levormeloxifene (L(3)). Treatments were administered orally each day for 18 mo. All doses of levormeloxifene resulted in adrenal weights at least 50% greater than placebo (p < .0001 for all). The target dose of levormeloxifene (L(2)) resulted in higher serum concentrations of cortisol (+63%), dehydroepiandrosterone-sulfate (+73%), and androstenedione (+37%) compared with the placebo group (p < .05 for all). In contrast, E(2) resulted in no significant differences in adrenal weight or adrenocortical steroids. Oral E(2) and all SERM doses resulted in similar reductions in serum gonadotropins and at least threefold greater uterine weight versus placebo (p < .0001 for all). Results indicate that the SERM levormeloxifene, in contrast to E(2), may have robust stimulatory effects on adrenocortical hormones in a postmenopausal model. These findings warrant further investigation into long-term SERM effects on adrenocortical function.


Assuntos
Córtex Suprarrenal/efeitos dos fármacos , Pirrolidinas/toxicidade , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Córtex Suprarrenal/metabolismo , Androgênios/sangue , Animais , Peso Corporal , Feminino , Gonadotropinas/sangue , Hidrocortisona/sangue , Macaca fascicularis , Tamanho do Órgão , Ovariectomia , Hipófise/efeitos dos fármacos , Pirrolidinas/administração & dosagem , Análise de Regressão , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem , Estatísticas não Paramétricas , Estimulação Química , Testes de Toxicidade Crônica
20.
Hear Res ; 282(1-2): 161-6, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21907781

RESUMO

Tamoxifen has been used extensively in the treatment of breast cancer and other neoplasms. In addition to its well-known action on estrogen receptors it is also known to acutely block chloride channels that participate in cell volume regulation. Tamoxifen's role in preventing cochlear outer hair cell (OHC) swelling in vitro suggested that OHC swelling noted following noise exposure could potentially be a therapeutic target for tamoxifen in its role as a chloride channel blocker to help prevent noise-induced hearing loss. To investigate this possibility, the effects of exposure to tamoxifen on physiologic measures of cochlear function in the presence and absence of subsequent noise exposure were studied. Male Mongolian gerbils (2-4 months old) were randomly assigned to different groups. Tamoxifen at ∼10 mg/kg was administered to one of the groups. Five hours later they were exposed to a one-third octave band of noise centered at 8 kHz in a sound-isolation chamber for 30 min at 108 dB SPL. Compound action potential (CAP) thresholds and distortion product otoacoustic emission (DPOAE) levels were measured 30-35 days following noise exposure. Tamoxifen administration did not produce any changes in CAP thresholds and DPOAE levels when administered by itself in the absence of noise. Tamoxifen causes a significant increase in CAP thresholds from 8 to 15 kHz following noise exposure compared to CAP thresholds in animals exposed to noise alone. No significant differences were seen in the DPOAE levels in the f(2) = 8-15 kHz frequency range where maximum noise-induced increases in CAP thresholds were seen. Contrary to our original expectation, it is concluded that tamoxifen potentiates the degree of damage to the cochlea resulting from noise exposure.


Assuntos
Cóclea/efeitos dos fármacos , Perda Auditiva Provocada por Ruído/etiologia , Ruído , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Tamoxifeno/toxicidade , Animais , Limiar Auditivo/efeitos dos fármacos , Cóclea/lesões , Cóclea/fisiopatologia , Potenciais Evocados/efeitos dos fármacos , Gerbillinae , Perda Auditiva Provocada por Ruído/induzido quimicamente , Perda Auditiva Provocada por Ruído/fisiopatologia , Masculino , Emissões Otoacústicas Espontâneas/efeitos dos fármacos , Fatores de Risco , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...