Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Aquat Toxicol ; 263: 106694, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37716317

RESUMEN

The objective of this study was to assess the effects of a model substance with anti-progestogenic activity on development of African clawed frog (Xenopus laevis) from tadpole to juvenile stage. Mifepristone, a synthetic progesterone receptor-blocking steroid hormone used in medicine as an abortifacient, was chosen as a model compound with anti-progestogenic activity. In the experiment, African clawed frog tadpoles were exposed to mifepristone at three concentrations (2, 21, and 215 ng L-1). A control group was exposed to dimethyl sulfoxide (DMSO; 0.001 %). The experiment started when tadpoles reached stages 47-48 according to Nieuwkoop and Faber (NF; 1994) and continued until stage NF 66, when metamorphosis was complete. Exposure to mifepristone had no significant effect on the rate of tadpole development, occurrence of morphological anomalies, weight, body length, or sex ratio. Mortality was within an acceptable range of 0-3.6 % throughout the test and did not differ among the groups. Histopathological examination of the gonads and thyroid gland revealed no significant changes. Therefore, we can conclude that mifepristone had no negative effect on development of the African clawed frog up to juvenile stage. Nevertheless, at the highest tested mifepristone concentration (215 ng L-1), gene expression analysis revealed up-regulation of mRNA expression of nuclear progesterone receptor (npr), membrane progesterone receptor (mpr), estrogen receptor beta (esrß), and luteinizing hormone (lh) in the brain-pituitary complex of exposed frogs at stage NF 66. Higher mRNA expression of npr was also found in frogs exposed to 22 ng L-1 mifepristone compared to the solvent control. These findings confirmed the anti-progestogenic activity of mifepristone in frogs because the up-regulation of progesterone receptors occurs if progesterone availability in the body is reduced. All the observed changes in combination may have negative consequences for reproduction and reproductive behavior later in life.


Asunto(s)
Progestinas , Contaminantes Químicos del Agua , Animales , Progestinas/farmacología , Mifepristona/toxicidad , Xenopus laevis , Receptores de Progesterona/genética , Contaminantes Químicos del Agua/toxicidad , Metamorfosis Biológica , ARN Mensajero , Larva
2.
J Toxicol Sci ; 47(7): 301-308, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35786681

RESUMEN

We examined that an estradiol-dominant state against progesterone could affect hematological parameters through hemodilution because estradiol is known to increase plasma volume via oncotic pressure. We performed a 2- and 3-week repeated oral dose study with mifepristone, a progesterone receptor antagonist, in female rats and examined erythrocyte counts, hemoglobin, hematocrit, plasma volume, levels of estradiol and progesterone, water intake, and water loss. Mifepristone treatment decreased some hematological parameters mildly and increased plasma volume. There were no remarkable changes in the balance of water intake and water loss through urination. Both estradiol and progesterone levels and the ratio of estradiol to progesterone increased. Therefore, our findings indicate that repeated mifepristone treatment increases estradiol levels and plasma volume, resulting in lower erythrocyte counts, hemoglobin, and hematocrit. The present study proved the possible contribution of estradiol to understanding the toxicological significance of mifepristone-induced hemodilution.


Asunto(s)
Estradiol , Mifepristona , Animales , Femenino , Hemodilución/métodos , Hemoglobinas , Mifepristona/toxicidad , Progesterona , Ratas , Agua
3.
Exp Eye Res ; 190: 107854, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31669405

RESUMEN

Death of retinal photoreceptors is the basis of prevalent blinding diseases. Since steroids might have a therapeutic role in retinal degenerations, we compared the protective effects of dexamethasone and progesterone on photoreceptor death induced by mifepristone and light exposure. Therefore, we studied the effective protection doses for each steroid in the two models. In addition, we analyzed changes in the levels of pro- and antiapoptotic molecules, glucocorticoid receptors α and ß (GRα and GRß), and rhodopsin under conditions of successful protection and photoreceptor survival. Mifepristone and light exposure selectively damaged photoreceptors. In light exposed retinas, photoreceptors mainly disappeared in the dorsotemporal region, while mifepristone produced a uniform damage. Dexamethasone and progesterone, at the same dose of 4 mg/kg/day for 2 days, preserved over 88% photoreceptor nuclei in both models. Assessment of cell death regulators showed that, in control retinas, both steroids activated BCL-XL, a prosurvival molecule, and decreased BID, a proapoptotic regulator. After steroid treatment of damaged retinas, BCL-XL, BCL2 and BAX showed characteristic patterns depending on the use of dexamethasone or progesterone on mifepristone or light exposed retinas. By contrast, BID decreased with any injury-steroid combination. Changes in GRα or GRß levels did not correlate with survival but were consistent with a mechanism of ligand induced downregulation of receptor expression. GRß might be upregulated by progesterone. Both dexamethasone and progesterone increased retinal rhodopsin stores, suggesting a link between photoreceptor protection and transduction pathways. Results show that dexamethasone and progesterone induced comparable but not identical protection responses in each model.


Asunto(s)
Dexametasona/farmacología , Glucocorticoides/farmacología , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Progesterona/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Degeneración Retiniana/prevención & control , Animales , Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Western Blotting , Caspasa 3 , Supervivencia Celular/fisiología , Antagonistas de Hormonas/toxicidad , Inmunohistoquímica , Luz/efectos adversos , Masculino , Ratones Endogámicos BALB C , Mifepristona/toxicidad , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/metabolismo , Receptores de Glucocorticoides/metabolismo , Degeneración Retiniana/etiología , Degeneración Retiniana/metabolismo , Rodopsina/metabolismo , Proteína bcl-X/metabolismo
4.
Aquat Toxicol ; 215: 105289, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31491707

RESUMEN

Mifepristone (RU486), a clinical abortion agent and potential endocrine disruptor, binds to progestin and glucocorticoid receptors and has multiple functional importance in reproductive physiology. A long-term exposure of RU486 resulted in masculinization of female fish, however, the epigenetic landscape remains elusive. Recent studies demonstrated that long non-coding RNAs (lncRNAs) might play potential roles in epigenetic modulation of sex differentiation, ovarian cancer and germline stem cell survival. To further understand the influence of RU486 exposure on epigenetic regulation, we performed a comparative investigation on sex-biased gonadal lncRNAs profiles using control XX/XY and RU486-induced sex reversed XX Nile tilapia (Oreochromis niloticus) by RNA-seq. In total, 962 sexually differentially expressed lncRNAs and their target genes were screened from the gonads of control and sex reversed fish. In comparison with the control XX group, sex reversal induced by RU486 treatment led to significant up-regulation of 757 lncRNAs and down-regulation of 221 lncRNAs. Hierarchical clustering analysis revealed that global lncRNA expression profiles in RU486-treated XX group clustered into the same branch with the control XY, whereas XX control group formed a separate branch. The KEGG pathway enrichment analysis showed that the cis-target genes between RU486-XX and control-XX were concentrated in NOD - like receptor signaling pathway, Cell adhesion molecules (CAMs) and Biosynthesis of amino acids. Real-time PCR and in situ hybridization experiments demonstrate that lncRNAs showing intense fluctuation during RU486 treatment are also sexually dimorphic during early sex differentiation, which further proves the intimate relationship between lncRNAs and sex differentiation and sexual transdifferentiation. Taken together, our data strongly indicates that a long-term exposure of RU486 resulted in sex reversal of XX female fish and the altered expression of sexually dimorphic lncRNAs might partially account for the sex reversal via epigenetic modification.


Asunto(s)
Cíclidos/genética , Cíclidos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Gónadas/metabolismo , Mifepristona/toxicidad , Progestinas/antagonistas & inhibidores , ARN Largo no Codificante/genética , Caracteres Sexuales , Animales , Femenino , Genoma , Gónadas/efectos de los fármacos , Masculino , Sistemas de Lectura Abierta/genética , Ovario/efectos de los fármacos , Ovario/metabolismo , ARN Largo no Codificante/metabolismo , Reproducibilidad de los Resultados , Testículo/efectos de los fármacos , Testículo/metabolismo , Factores de Tiempo , Distribución Tisular/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad
5.
Water Res ; 137: 64-71, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29544204

RESUMEN

Vast numbers of xenobiotics are known still to be present in treated municipal wastewater treatment plant (WWTP) effluents. Some of these possess endocrine-disrupting potency and pose risks for exposed aquatic animals. We searched for 17 potential environmental contaminants having affinity to the progesterone receptor. Relative potency values of these progesterone receptor-active chemicals were obtained. On the basis of relative potencies and measured environmental concentrations, the contribution of progestins to measured progestagenic activities was evaluated. Wastewaters (influent and effluent) and surrounding surface waters (upstream and downstream) at six municipal WWTPs were screened using instrumental chemical analysis and in vitro reporter gene bioassay. We showed the presence of target compounds and (anti-)progestagenic activities in municipal wastewater and surface water. Nine and seven progestins were identified in influent and effluent wastewaters, respectively. Only two compounds, progesterone and medroxyprogesterone were found in surface waters. Progestagenic agonistic activities in influents were partially masked by strong anti-progestagenic activities that were detected in all influents and ranged from 2.63 to 83 ng/L of mifepristone equivalents (EQs). Progestagenic activities were detected in all effluents and ranged from 0.06 to 0.47 ng/L of reference compound ORG 2058 EQs (a synthetic progestin equivalents), thus indicating incomplete removal of progestins during wastewater treatment processing. This activity poses a continuing risk for the aquatic environment. By contrast, anti-progestagenic activities showed better removal efficiency in WWTPs compared to progestagenic agonistic activities. Anti-progestagenic activities were found in only three of six effluents and ranged from 0.26 to 2.1 ng/L mifepristone EQs. We explained most of the progestagenic activity in municipal WWTP effluents by the presence of synthetic progestins and progesterone, which contributed 65-96% of such activity in samples where no antagonistic activity was found. The progestins medroxyprogesterone acetate, megestrol acetate and progesterone contributed most to the progestagenic activity detected in municipal effluents. Anti-progestagenic activities were found in some municipal effluents, but no causative agents were revealed because two analysed selective progesterone receptor modulators (SPRMs) with anti-progestagenic activities, mifepristone and ulipristal acetate, were not present in the effluents.


Asunto(s)
Progesterona/toxicidad , Progestinas/toxicidad , Aguas Residuales/toxicidad , Contaminantes Químicos del Agua/análisis , Contaminantes Químicos del Agua/toxicidad , Línea Celular , República Checa , Ecotoxicología/métodos , Monitoreo del Ambiente , Humanos , Medroxiprogesterona/análisis , Medroxiprogesterona/toxicidad , Mifepristona/toxicidad , Progesterona/análisis , Progestinas/análisis , Receptores de Progesterona/metabolismo , Eslovaquia , Eliminación de Residuos Líquidos/métodos , Aguas Residuales/análisis
6.
Cell Death Differ ; 24(2): 288-299, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27911443

RESUMEN

We observed that the transient induction of mtDNA double strand breaks (DSBs) in cultured cells led to activation of cell cycle arrest proteins (p21/p53 pathway) and decreased cell growth, mediated through reactive oxygen species (ROS). To investigate this process in vivo we developed a mouse model where we could transiently induce mtDNA DSBs ubiquitously. This transient mtDNA damage in mice caused an accelerated aging phenotype, preferentially affecting proliferating tissues. One of the earliest phenotypes was accelerated thymus shrinkage by apoptosis and differentiation into adipose tissue, mimicking age-related thymic involution. This phenotype was accompanied by increased ROS and activation of cell cycle arrest proteins. Treatment with antioxidants improved the phenotype but the knocking out of p21 or p53 did not. Our results demonstrate that transient mtDNA DSBs can accelerate aging of certain tissues by increasing ROS. Surprisingly, this mtDNA DSB-associated senescence phenotype does not require p21/p53, even if this pathway is activated in the process.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , ADN Mitocondrial/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilcisteína/farmacología , Envejecimiento , Animales , Apoptosis , Puntos de Control del Ciclo Celular/efectos de los fármacos , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mifepristona/toxicidad , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Timocitos/citología , Timocitos/efectos de los fármacos , Timocitos/metabolismo , Proteína p53 Supresora de Tumor/genética
7.
Toxicol Mech Methods ; 26(1): 36-45, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26907462

RESUMEN

OBJECTIVE: Mifepristone (RU486) is an oral first-line contraceptive used by hundreds of millions of women, and recently it was tested for anticancer activity in both genders worldwide. We are developing metapristone (the N-monodemethyl RU486) as a potential metastasis chemopreventive. The present acute and 30-d subacute toxicity study aimed at examining and compared in parallel the potential toxicity of the two drugs. METHODS: The single-dose acute toxicity and 30-d subacute toxicity studies were conducted in mice and rats, respectively, by gavaging metapristone or mifepristone at various doses. Blood samples and organs were collected for blood chemistry, hematology and histology analyses. RESULTS: Oral mifepristone (3000 mg/kg) caused 30% and 40% death in female and male mice, respectively, within 15 h post-dosing. In comparison, the same dose of metapristone produced 30% acute death in males only. Thirty-day oral administration of the two drugs to rats (12.5, 50 and 200 mg/kg/day) caused reversible hepatotoxicity that only occurred at 200 mg/kg/day group, evidenced by the elevated liver enzyme activity and liver organ weight. CONCLUSION: The present study, for the first time, reveals reversible hepatotoxicity in rats caused by the 30-d consecutive administration at the high dose, and warns the potential hepatotoxicity caused by long-term administrations of high doses of mifepristone or metapristone in clinical trials but not by the acute single abortion doses.


Asunto(s)
Abortivos Esteroideos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Mifepristona/análogos & derivados , Mifepristona/toxicidad , Abortivos Esteroideos/administración & dosificación , Animales , Femenino , Masculino , Mifepristona/administración & dosificación , Ratas
8.
Biomed Pharmacother ; 78: 291-300, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26898454

RESUMEN

Malignant melanoma, the most deadly form of skin cancer, has a high propensity for metastatic spread and is notoriously chemotherapy-resistant. Metapristone is the primary metabolite of mifepristone (RU486) and shows biological activities similar to RU486. In the present study, we comprehensively investigated the efficacy of metapristone as a metastatic chemopreventive against melanoma B16F10 cells in vitro and in vivo, and evaluated the safety profile of both drugs in mice. Metapristone showed less cytostatic effect in vitro and in vivo in comparison with mifepristone. However, metapristone interfered the adhesion of B16F10 cells to fibronectin by down-regulating cellular expression of integrin α4. Chemopreventive pretreatment followed by oral administration of metapristone and mifepristone (2.5, 10, 50 mg/kg/day for 35 days) to melanoma C57BL/6 mouse model showed significant attenuation of pulmonary metastatic development. Oral administration of high doses of metapristone and mifepristone to normal mice for 35 days (25, 100, 250 mg/kg/day) resulted in a dose-dependent increase in mouse liver weight that was more severe with mifepristone than metapristone. The long-term toxicity study revealed more changes by mifepristone in counts of erythrocytes, leukocytes and platelets than by metapristone. In conclusion, metapristone may fit into a new class of cancer metastatic chemopreventive agents. It showed a safety and efficacy profile better than mifepristone.


Asunto(s)
Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Mifepristona/análogos & derivados , Mifepristona/efectos adversos , Mifepristona/uso terapéutico , Animales , Anticarcinógenos/efectos adversos , Anticarcinógenos/uso terapéutico , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimioprevención , Modelos Animales de Enfermedad , Femenino , Integrina alfa4/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Mifepristona/toxicidad , Resultado del Tratamiento
9.
Sci Rep ; 5: 7830, 2015 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-25597938

RESUMEN

Mifepristone (RU486), a synthetic steroid compound used as an abortifacient drug, has received considerable attention to its anticancer activity recently. To explore the possibility of using mifepristone as a cancer metastasis chemopreventive, we performed a systems pharmacology analysis of mifepristone-related molecules in the present study. Data were collected by using Natural Language Processing (NLP) and 513 mifepristone-related genes were dug out and classified functionally using a gene ontology (GO) hierarchy, followed by KEGG pathway enrichment analysis. Potential signal pathways and targets involved in cancer were obtained by integrative network analysis. Total thirty-three proteins were involved in focal adhesion-the key signaling pathway associated with cancer metastasis. Molecular and cellular assays further demonstrated that mifepristone had the ability to prevent breast cancer cells from migration and interfere with their adhesion to endothelial cells. Moreover, mifepristone inhibited the expression of focal adhesion kinase (FAK), paxillin, and the formation of FAK/Src/Paxillin complex, which are correlated with cell adhesion and migration. This study set a good example to identify chemotherapeutic potential seamlessly from systems pharmacology to cellular pharmacology, and the revealed hub genes may be the promising targets for cancer metastasis chemoprevention.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Mifepristona/toxicidad , Paxillin/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Redes y Vías Metabólicas/efectos de los fármacos
10.
Aquat Toxicol ; 144-145: 96-104, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24177212

RESUMEN

Here, we analyzed the transcriptional effects of the antiprogestin mifepristone (MIF, RU486) and progesterone (P4) in zebrafish as well as their in vitro activities in yeast-based reporter gene assays. This study is associated with the reproduction study in adult zebrafish and embryos exposed for 21 days to 5, 39, 77 ng/L MIF, and 25 ng/L P4 (Blüthgen et al., 2013a). The in vitro activities of MIF and P4 were investigated using a series of recombinant yeast-based assays (YES, YAS, YPS) and compared to transcriptional alterations obtained in fish tissues and embryos from the exposure study. MIF elicited antiestrogenic, androgenic and progestogenic activities in recombinant yeast, similar to P4, and no antiprogestogenic activity in vitro. The transcriptional alterations of steroid hormone receptors were similar in adult males and females, and more pronounced in embryos. MIF tended to transcriptionally down-regulate the androgen (ar), progesterone (pgr) and glucocorticoid (gr) receptors in adult fish and embryos. Transcripts of the estrogen receptor (esr1) and vitellogenin (vtg1) were not significantly altered. A trend for down-regulation was observed for transcripts of genes belonging to steroidogenic enzymes including 17ß-hydroxysteroid dehydrogenase type 3 (hsd17b3), 3 ß-hydroxysteroid dehydrogenase (hsd3b), P450 aromatase A (cyp19a) and 11ß-hydroxylase (cyp11b). P4 resulted in similar transcriptional alterations as MIF. The data indicate that gene expression changes (here and later gene expression is taken as synonym to gene transcription) and in vitro activities match only in part including the lack of antiprogestogenic activity of MIF. Additionally, effects on reproduction and gonad histology described in the associated report (Blüthgen et al., 2013a) can only partly be explained by gene expression data presented here.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Mifepristona/toxicidad , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/fisiología , Animales , Femenino , Masculino , Vitelogeninas/genética , Vitelogeninas/metabolismo , Pez Cebra/embriología
11.
Aquat Toxicol ; 144-145: 83-95, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24161497

RESUMEN

Effects of synthetic progestins have recently been reported in fish, but potential effects of the synthetic antiprogestin mifepristone (MIF), also called RU486, have not been studied. The present study provides first insights into reproductive effects of MIF in zebrafish in comparison to the progesterone receptor agonist, progesterone (P4). We carried out a reproductive study using breeding groups of adult zebrafish. After a 14 day pre-exposure, zebrafish were exposed for 21 days to 5, 39, 77 ng/L MIF, 25 ng/L P4 and water and solvent controls. In addition, embryos originating from exposed adult fish were continuously exposed to 3, 15, 26 ng/L MIF, and 254 ng/L P4, respectively, for 96 h post fertilization. We found a significant U-shaped increase in egg production after exposure to 5 and 77 ng/L MIF, but no effects at 25 ng/L P4. Levels of sex steroid hormones in blood plasma of adult males (11-ketotestosterone) and females (17 ß-estradiol) were not altered. In addition to an increase of mature vitellogenic oocytes in ovaries of females exposed to MIF and P4, we observed several histopathological changes in ovaries, including post-ovulatory follicles, atretic follicles and proteinaceous fluid. Male gonads showed no or less alterations and no histopathological effects. Fertility of eggs and hatching success of embryos (F1 generation) was not affected at 3-26 ng/L MIF and 254 ng/L P4, respectively. The data lead to the conclusion that trace quantities of MIF affect reproduction of zebrafish and ovaries of female zebrafish. Effects on transcriptional changes in adult and embryonic zebrafish of this study in comparison to in vitro effects are reported in the associated report (Blüthgen et al., 2013a).


Asunto(s)
Mifepristona/toxicidad , Reproducción/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/fisiología , Animales , Embrión no Mamífero , Femenino , Hormonas Esteroides Gonadales/metabolismo , Gónadas/efectos de los fármacos , Masculino , Ovario/efectos de los fármacos , Pez Cebra/embriología
12.
Psychoneuroendocrinology ; 36(6): 824-33, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21163582

RESUMEN

Progesterone withdrawal has been proposed as an underlying factor in premenstrual syndrome and postpartum depression. Progesterone withdrawal induces forced swim test (FST) immobility in mice, a depression-like behavior, but the contribution of specific receptors to this effect is unclear. The role of progesterone's GABA(A) receptor-modulating metabolite allopregnanolone in depression- and anxiety-related behaviors has been extensively documented, but little attention has been paid to the role of progesterone receptors. We administered the classic progesterone receptor antagonist mifepristone (RU-38486) and the specific progesterone receptor antagonist CDB-4124 to mice that had been primed with progesterone for five days, and found that both compounds induced FST immobility reliably, robustly, and in a dose-dependent fashion. Although CDB-4124 increased FST immobility, it did not suppress initial activity in a locomotor test. These findings suggest that decreased progesterone receptor activity contributes to depression-like behavior in mice, consistent with the hypothesis that progesterone withdrawal may contribute to the symptoms of premenstrual syndrome or postpartum depression.


Asunto(s)
Depresión/inducido químicamente , Norpregnadienos/toxicidad , Progesterona/fisiología , Receptores de Progesterona/antagonistas & inhibidores , Animales , Depresión/fisiopatología , Depresión Posparto , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Finasterida/farmacología , Finasterida/toxicidad , Locomoción/efectos de los fármacos , Ratones , Ratones Endogámicos DBA , Mifepristona/farmacología , Mifepristona/toxicidad , Norpregnadienos/administración & dosificación , Norpregnadienos/farmacología , Síndrome Premenstrual , Progesterona/farmacología , Receptores de Progesterona/fisiología , Método Simple Ciego , Natación
13.
J Toxicol Sci ; 34 Suppl 1: SP31-42, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19265287

RESUMEN

In order to assess ovarian pathological changes and their relationship to changes in female fertility parameters, mifepristone, a progesterone receptor antagonist, was selected as the test article and was administered orally to female rats at dose levels of 0, 0.8, 4, 20 and 100 mg/kg for 2 or 4 weeks in repeated dose-toxicity studies and in a female fertility study at dose levels of 0, 0.8, 4 and 20 mg/kg from > 2 weeks before copulation to postcoital day 7. In the repeated dose toxicity studies, persistent estrus was seen in the vaginal smears, and multiple cysts in the ovaries at necropsy, increases in luteinized cysts and hypertrophy of previously formed corpora lutea were observed in the histopathological examination of ovaries in rats receiving 20 mg/kg or more for 2 or 4 weeks. In female fertility studies, persistent vaginal cornification was also observed at 20 mg/kg and the precoital interval was significantly shortened. All of the animals were completely infertile when dosed with 20 mg/kg during the post-coital period. An increase in pre-implantation losses was observed in the animals treated with 20 mg/kg during the pre-coital phase, while treatment with 4 mg/kg mifepristone during the post-coital phase induced an increase in post-implantation losses. These results suggested that a 2-week administration period would be sufficient to detect the ovarian toxicity of mifepristone in repeated dose toxicity study and the pathological findings in the ovaries would reflect the alterations in female reproductive endpoints in the female fertility study.


Asunto(s)
Fertilidad/efectos de los fármacos , Antagonistas de Hormonas/toxicidad , Mifepristona/toxicidad , Ovario/efectos de los fármacos , Pruebas de Toxicidad/métodos , Administración Oral , Animales , Esquema de Medicación , Desarrollo Embrionario/efectos de los fármacos , Ciclo Estral/efectos de los fármacos , Ciclo Estral/fisiología , Femenino , Fertilidad/fisiología , Antagonistas de Hormonas/administración & dosificación , Infertilidad Femenina/inducido químicamente , Infertilidad Femenina/fisiopatología , Japón , Masculino , Mifepristona/administración & dosificación , Quistes Ováricos/inducido químicamente , Quistes Ováricos/patología , Ovario/metabolismo , Ovario/patología , Embarazo , Asociación entre el Sector Público-Privado , Ratas , Ratas Sprague-Dawley , Sociedades Científicas , Organismos Libres de Patógenos Específicos , Vagina/efectos de los fármacos , Vagina/patología
14.
Toxicol In Vitro ; 22(1): 261-6, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17928190

RESUMEN

Inducible gene expression systems are being used in many in vitro and in vivo applications for target discovery, target validation and as components in exploratory therapeutic agents. Ideally, the ligands, which activate the systems, are benign so that the effects can be strictly attributed to the induced protein. As a first step to defining the potential effects of these inducers, we tested three of them, doxycycline, muristerone A and mifepristone (for tet-, ecdysone- and progesterone antagonist-inducible systems respectively), for toxicity across a panel of normal cells and cancer cell lines. In contrast to both muristerone A and mifepristone that showed no significant toxicity on any of the tested cells, we observed that doxycycline induced cell death in selected cancer and primary cell lines. The different susceptibility of cell lines to the ligands commonly used in these inducible systems suggests that it is important to consider the effects of the inducers prior to their use in experimental in vitro cell culture systems.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Doxiciclina/administración & dosificación , Doxiciclina/toxicidad , Sistemas de Liberación de Medicamentos , Ecdisterona/administración & dosificación , Ecdisterona/análogos & derivados , Ecdisterona/toxicidad , Humanos , Ligandos , Mifepristona/administración & dosificación , Mifepristona/toxicidad
15.
Contraception ; 68(6): 409-20, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14698070

RESUMEN

Roussel Uclaf in partnership with the INSERM unit of Prof. E.E. Baulieu first discovered mifepristone (RU486) as part of a large research program on steroidal compounds with antihormone properties. Exhibiting a strong affinity to the progesterone and the glucocorticoid receptors, mifepristone exerted competitive antagonism to these hormones both in in vitro and in animal experiments. Due to its antiprogesterone activity, it was proposed that mifepristone be used for the termination of early human pregnancy. Mifepristone, at a dose of 600 mg initially used alone, was then used with a subsequent low dose of prostaglandin that led to a success rate of 95% as a medical method for early termination of pregnancy (TOP). Its use was extended to other indications, such as cervical dilatation prior to surgical TOP in the first trimester, therapeutic TOP for medical reasons beyond the first trimester, and for labor induction in case of fetal death in utero. The efficacy and safety of this treatment has been confirmed based on its use for over a decade, with close adherence to the approved recommendations. This paper describes the safety studies conducted in animals as well as the safety follow-up and side effects reported with use of the compound in various indications either approved or unapproved. The rationale for warnings and contraindications for use of the product are also explained. At lower doses, the molecule has proven promising for contraceptive purposes with few reported side effects. However, development of the product for this indication would require long-term studies. Although political and philosophical obstacles have delayed research, the use of mifepristone for other potential indications in gynecology or oncology should be investigated.


Asunto(s)
Anticonceptivos Sintéticos Poscoito/farmacología , Mifepristona/farmacología , Animales , Ensayos Clínicos como Asunto , Anticonceptivos Sintéticos Poscoito/efectos adversos , Anticonceptivos Sintéticos Poscoito/química , Anticonceptivos Sintéticos Poscoito/toxicidad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Mifepristona/efectos adversos , Mifepristona/química , Mifepristona/toxicidad , Modelos Animales , Progesterona/antagonistas & inhibidores , Seguridad
16.
J Neurosci ; 23(13): 5536-44, 2003 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-12843254

RESUMEN

The innate immune system plays a crucial role in protecting the host against infectious microorganisms. An inappropriate control of this system may have profound consequences, because of the maintained production of specific proinflammatory molecules. Glucocorticoids are the most efficient endogenous molecules that provide negative feedback on proinflammatory signaling and gene expression. Here we show that activation of this system is not detrimental for the brain but a profound neurodegeneration takes place in animals treated with the glucocorticoid receptor inhibitor Mifepristone (RU486). This drug increased the inflammatory reaction induced by a single intracerebral bolus of lipopolysaccharide (LPS). Inhibition of tumor necrosis factor alpha (TNF-alpha) totally abolished the neurotoxic effect of the endotoxin, and chronic infusion of the cytokine mimicked the treatment combining RU486 and LPS. The neuronal damage caused by TNF-alpha is dependent on both nitric oxide and caspase pathways. In controlling the cerebral innate immunity and microglial TNF-alpha production, glucocorticoids play a major role in protecting the brain against bacterial cell wall components.


Asunto(s)
Encéfalo/fisiología , Glucocorticoides/fisiología , Inmunidad Innata/fisiología , Mifepristona/toxicidad , Enfermedades Neurodegenerativas/inducido químicamente , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Caspasas/metabolismo , Citocinas/fisiología , Vías de Administración de Medicamentos , Sinergismo Farmacológico , Retroalimentación Fisiológica/inmunología , Glucocorticoides/farmacología , Inmunidad Innata/inmunología , Lipopolisacáridos/toxicidad , Masculino , Microglía/metabolismo , FN-kappa B/metabolismo , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/farmacología , Óxido Nítrico/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología , Factor de Necrosis Tumoral alfa/toxicidad
17.
Mol Cancer ; 2: 3, 2003 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-12605714

RESUMEN

BACKGROUND: Glucocorticoids inhibit hepatocellular proliferation and modulate the expression of oncogenes and tumor suppressor genes via mechanisms involving the glucocorticoid receptor. Glucocorticoids also produce a receptor-mediated inhibitory effect on both basal and hormone-stimulated expression of a newly discovered family of molecules important for shutting off cytokine action. We therefore hypothesized that inhibiting glucocorticoid receptors may disturb hepatocellular growth and apoptosis. Consequently, we investigated the effect of RU486, a potent antagonist of the glucocorticoid receptor, on basal levels of hepatocellular proliferation and apoptosis in male B6C3F1 mice. Furthermore, we evaluated the effect of this compound on cellular genes involved in the regulation of these important processes. RESULTS: Data show that treatment of male B6F3C1 mice with RU486 (2 mg/kg/d, ip) for 7 days dramatically inhibited liver cell proliferation by about 45% and programmed hepatocellular death by approximately 66%. RU 486 also significantly increased hepatic expression of the oncogenes mdm2 and JunB, while reducing that of the tumor suppressor gene p53. CONCLUSION: Exposure to RU486 may ultimately enhance the susceptibility of the liver to cancer risk by diminishing its ability to purge itself of pre-cancerous cells via apoptosis. This effect may be mediated through increases in the hepatic expression of the oncogene mdm2, coupled with decreases in that of the tumor suppressor gene p53. The decrease in hepatocellular proliferation caused by RU 486 may be related to effects other than its anti-glucocorticoid activity.


Asunto(s)
Antagonistas de Hormonas/farmacología , Neoplasias Hepáticas/inducido químicamente , Mifepristona/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Antagonistas de Hormonas/toxicidad , Masculino , Ratones , Ratones Endogámicos , Mifepristona/toxicidad , Proteínas Nucleares/genética , Oncogenes/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-mdm2 , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/genética
18.
Toxicol Sci ; 54(2): 338-54, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10774816

RESUMEN

After previously examining 12 compounds with known endocrine activities, we have now evaluated 4 additional compounds in a Tier I screening battery for detecting endocrine-active compounds (EACs): a weak estrogen receptor (ER) agonist (coumestrol; COUM), an androgen receptor (AR) agonist (testosterone; TEST), a progesterone receptor (PR) agonist (progesterone; PROG), and a PR antagonist (mifepristone; RU486). The Tier I battery incorporates 2 short-term in vivo tests (5-day ovariectomized female battery; 15-day intact male battery) and an in vitro yeast transactivation system (YTS). The Tier I battery is designed to identify compounds that have the potential to act as agonists or antagonists to the estrogen, androgen, progesterone, or dopamine receptors; steroid biosynthesis inhibitors (aromatase, 5alpha-reductase, and testosterone biosynthesis); or compounds that alter thyroid function. In addition to the Tier I battery, a 15-day dietary restriction experiment was performed using male rats to assess confounding due to treatment-related decreases in body weight. In the Tier I female battery, TEST administration increased uterine weight, uterine stromal cell proliferation, and altered hormonal concentrations (increased serum testosterone [T] and prolactin [PRL]; and decreased serum FSH and LH). In the male battery, TEST increased accessory sex gland weights, altered hormonal concentrations (increased serum T, dihydrotestosterone [DHT], estradiol [E2], and PRL; decreased serum FSH and LH), and produced microscopic changes of the testis (Leydig cell atrophy and spermatid retention). In the YTS, TEST activated gene transcription in the yeast containing the AR or PR. In the female battery, COUM administration increased uterine weight, uterine stromal cell proliferation, and uterine epithelial cell height, and increased serum PRL concentrations. In the male battery, COUM altered hormonal concentrations (decreased serum T, DHT, E2; increased serum PRL) and, in the YTS, COUM activated gene transcription in the yeast containing the ER. In the female battery, PROG administration increased uterine weight, uterine stromal cell proliferation, and uterine epithelial cell height and altered hormonal concentrations (increased serum progesterone and decreased serum FSH and LH). In the male battery, PROG decreased epididymis and accessory sex gland weights, altered hormonal concentrations (decreased serum T, PRL, FSH, and LH; increased serum progesterone and E2), and produced microscopic changes of the testis (Leydig cell atrophy). In the YTS, PROG activated gene transcription in the yeast containing the AR or PR. In the female battery, RU486 administration increased uterine weight and decreased uterine stromal cell proliferation. In the male battery, RU486 decreased epididymis and accessory sex gland weights and increased serum FSH and LH concentrations. In the YTS, RU486 activated gene transcription in the yeast containing the ER, AR, or PR. Dietary restriction data demonstrate that confounding due to decrements in body weight are not observed when body weight decrements are 10% or less in the Tier I male battery. In addition, minimal confounding is observed at body decrements of 15% (relative liver weight, T3, and T4). Hence, compounds can be evaluated in this Tier I at levels that produce a 10% decrease in body weight without confounding of the selected endpoints. Using the responses obtained for all the endpoints in the Tier I battery, a distinct "fingerprint" was produced for each type of endocrine activity against which compounds with unknown activity can be compared. These data demonstrate that the described Tier I battery is useful for identifying EACs and they extend the compounds evaluated to 16.


Asunto(s)
Cumestrol/toxicidad , Sistema Endocrino/efectos de los fármacos , Antagonistas de Hormonas/toxicidad , Mifepristona/toxicidad , Progesterona/toxicidad , Testosterona/toxicidad , Animales , Líquidos Corporales/efectos de los fármacos , Líquidos Corporales/fisiología , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Sistema Endocrino/patología , Estro/efectos de los fármacos , Femenino , Gónadas/efectos de los fármacos , Gónadas/patología , Masculino , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Pruebas de Toxicidad/métodos , Útero/efectos de los fármacos , Útero/patología , Útero/fisiología
19.
Liver ; 18(5): 343-51, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9831364

RESUMEN

AIMS/BACKGROUND: Estrogen is known to affect hepatobiliary function; however, it is unusual for high serum levels of estrogen to actually result in clinically detectable hyperbilirubinemia. Women affected by cholestatic jaundice during pregnancy share this genetic susceptibility with two Cricetulus hamsters, the Armenian hamster (Cricetulus migratorius) and the Chinese hamster (Cricetulus griseus). Nevertheless, the pathophysiologic process responsible for this estrogen induced icterus may be different in women and hamsters. The present study compares various facets of estrogen-induced icterus in these two closely related hamsters. METHODS: Hamsters were injected with various estrogens and the acute and chronic effects on liver were monitored by measuring changes in serum constituents and by observing changes in hepatic structure as seen grossly and by light and electron microscopy. RESULTS: In previous studies, hepatic tumors developed in most Armenian hamsters after chronic estrogen treatment, but in the present study, the livers of Chinese hamsters were remarkably free of neoplastic change under similar conditions. Also, when compared with the responses in the Armenian hamsters, signs of hepatic destruction and regeneration were less prevalent in estrogen-treated Chinese hamsters, and they were less susceptible to the effects of estrogen (because larger doses of estrogen were required to produce icterus and the bilirubin levels were lower and of shorter duration). In contrast to the findings in Armenian hamsters, bile canaliculi were severely affected in livers of estrogen-treated Chinese hamsters, and hepatic microvesicular steatosis, indicative of an unusual lipodystrophy caused by estrogen, was prominent. An additional lesion peculiar to the Chinese hamster was striking sinusoidal dilatation, which may be analogous to the oral contraceptive-induced sinusoidal dilatation in humans. CONCLUSIONS: Although these two hamster species are genetically similar, the genes activated by the estrogen receptor show remarkable heterogeneity when their respective livers are examined. Comparisons within these species may provide information about the specific gene activation responsible for particular pathologic events.


Asunto(s)
Adenoma de Células Hepáticas/inducido químicamente , Carcinoma Hepatocelular/inducido químicamente , Cricetulus , Estrógenos/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Hígado/efectos de los fármacos , Adenoma de Células Hepáticas/patología , Animales , Bilirrubina/sangre , Carcinoma Hepatocelular/patología , Cricetinae , Dietilestilbestrol/toxicidad , Femenino , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/ultraestructura , Hígado/patología , Neoplasias Hepáticas Experimentales/patología , Masculino , Acetato de Medroxiprogesterona/toxicidad , Mifepristona/toxicidad , Especificidad de la Especie , Tamoxifeno/toxicidad , Zeranol/toxicidad
20.
Int J Oncol ; 12(5): 1171-6, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9538145

RESUMEN

The present study was undertaken to evaluate the efficacy of using steroid hormone antagonists tamoxifen and Ru486 for chemotherapy or chemoprevention of choriocarcinoma or other less malignant gestational trophoblastic diseases (GTDs) such as invasive mole. Using 4 trophoblast cell lines, we have shown that tamoxifen (>/= 2 microM) has anti-growth activity on the choriocarcinoma cell line BeWo but not on the other cell lines in a time and dose dependent manner while Ru486 invariably had no detectable effect. Based on a radioimmunoassay, we have been able to detect low levels of estrogen receptors on BeWo (6 +/- 0.4 fm/mg; Kd=438+/- 73 pM) and JEG-3 (6.55 +/- 1.2 fm/mg; Kd=710 +/- 42 pM) cells and progesterone receptors on HT (48.62 fm/mg; Kd=1,690 +/- 182 pM) and TL (8.46 fm/mg; Kd=1,540 +/- 115 pM) cells. However, there is no definite correlation between steroid responsiveness and the presence of the receptors. The mechanism of our observed tamoxifen-mediated anti-cellular effect is uncertain and characteristics commonly associated with apoptotic cell death were not observed. The level of neither wild-type nor mutant forms of the p53 protein correlated with sensitivity to tamoxifen. Our results suggest that estrogen may be a growth hormone for some trophoblasts and tamoxifen may be potentially useful for the treatment of selected cases of choriocarcinoma or other trophoblastic diseases.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Tamoxifeno/toxicidad , Línea Celular , Coriocarcinoma , Antagonistas de Estrógenos/toxicidad , Femenino , Antagonistas de Hormonas/toxicidad , Humanos , Cinética , Mifepristona/toxicidad , Embarazo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Neoplasias Uterinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA