Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Environ Toxicol ; 36(4): 598-606, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33247482

RESUMO

As a natural androgen, androstenedione (AED) may pose potential risks to aquatic organisms due to its ubiquitousness in aquatic environments. Here we assessed the adverse effects of AED on histology of gonads, as well as mRNA expression levels of 34 genes concerned with hypothalamic-pituitary-gonadal (HPG) axis, germ-cell differentiation and sex differentiation in zebrafish (Danio rerio). Adult zebrafish were exposed to solvent control and three measured concentrations of 0.2, 2.3 and 23.7 µg/L AED for 60 days. The results showed that AED did not induce any obvious histological effects in the ovaries and testes. Of the investigated genes, transcriptional expression levels of amh and cyp11c1 genes in the ovaries of females were significantly increased by AED at 2.3 or 23.7 µg/L. However, different exposure concentrations of AED significantly inhibited mRNA expression of gnrh3, atf4b1 and cyp19a1b in the brain of males. In the testes of males, AED at 2.3 µg/L led to a significant induction of sox9b gene, but it at 23.7 µg/L down-regulated nr5a1b gene. These observed transcriptional changes indicated that AED could pose potential androgenic effects in zebrafish.


Assuntos
Androstenodiona/toxicidade , Ovário/efeitos dos fármacos , Testículo/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Peixe-Zebra , Animais , Relação Dose-Resposta a Droga , Regulação para Baixo , Feminino , Células Germinativas/efeitos dos fármacos , Células Germinativas/patologia , Masculino , Ovário/metabolismo , Ovário/patologia , Diferenciação Sexual/efeitos dos fármacos , Diferenciação Sexual/genética , Testículo/metabolismo , Testículo/patologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
2.
Molecules ; 26(20)2021 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-34684800

RESUMO

Androstenedione is a steroidal hormone produced in male and female gonads, as well as in the adrenal glands, and it is known for its key role in the production of estrogen and testosterone. Androstenedione is also sold as an oral supplement, that is being utilized to increase testosterone levels. Simply known as "andro" by athletes, it is commonly touted as a natural alternative to anabolic steroids. By boosting testosterone levels, it is thought to be an enhancer for athletic performance, build body muscles, reduce fats, increase energy, maintain healthy RBCs, and increase sexual performance. Nevertheless, several of these effects are not yet scientifically proven. Though commonly used as a supplement for body building, it is listed among performance-enhancing drugs (PEDs) which is banned by the World Anti-Doping Agency, as well as the International Olympic Committee. This review focuses on the action mechanism behind androstenedione's health effects, and further side effects including clinical features, populations at risk, pharmacokinetics, metabolism, and toxicokinetics. A review of androstenedione regulation in drug doping is also presented.


Assuntos
Androstenodiona/farmacologia , Anabolizantes/farmacologia , Androstenodiona/metabolismo , Androstenodiona/toxicidade , Animais , Atletas , Desempenho Atlético , Suplementos Nutricionais/toxicidade , Dopagem Esportivo , Feminino , Humanos , Masculino , Fatores Sexuais , Testosterona/metabolismo
3.
Ecotoxicol Environ Saf ; 147: 509-515, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28915398

RESUMO

Androstenedione (AED) is a naturally occurring steroid hormone. It is metabolized to potent androgens, which may induce androgenic effects in fish. However, little is known whether and how the androgens interfere with the fish gonadal development and reproduction. This study aimed at demonstrating the effects of long-term AED exposure on reproduction and development in mosquitofish (Gambusia affinis). The growth, development and several morphological endpoints, including the segment number and length of anal fin, histological changes of gonads and liver, were evaluated in mosquitofish during development from fertilized embryo to adulthood (180 days) after exposure of AED at environmentally relevant concentrations. We found that the growth (length, body weight and condition factor) of fish was negatively correlated with AED concentration in females, but not in males. The significant elongation of the ray and increment of segment numbers in the anal fin, were detected in all mosquitofish after exposure. Moreover, AED exposure (0.4gµ/L) caused damages in gonads and reduced the number of pregnant females. These findings indicate that AED has adverse effects on the growth and development of the western mosquitofish after long-term exposure (180d). Long-term exposure (180d) to AED, including environmentally relevant concentration (0.4µg/L and 4µg/L), induced masculinization in female mosquitofish under the experimental conditions.


Assuntos
Androstenodiona/toxicidade , Ciprinodontiformes/crescimento & desenvolvimento , Gônadas/efeitos dos fármacos , Caracteres Sexuais , Poluentes Químicos da Água/toxicidade , Androgênios/metabolismo , Androstenodiona/análise , Animais , Tamanho Corporal/efeitos dos fármacos , Ciprinodontiformes/metabolismo , Relação Dose-Resposta a Droga , Feminino , Gônadas/crescimento & desenvolvimento , Fígado/efeitos dos fármacos , Fígado/crescimento & desenvolvimento , Masculino , Reprodução/efeitos dos fármacos , Fatores de Tempo , Poluentes Químicos da Água/análise
4.
Mar Environ Res ; 189: 106062, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37390515

RESUMO

An increasing number of microplastics have been detected in aquatic environments, causing various damage to organisms. The size of microplastics affects the toxicity once they enter the organisms. Meanwhile, there is an increasing variety of Endocrine-disrupting chemicals (EDCs) present in aquatic environments. Androstenedione (AED) is a typical EDC. In this study, we used polystyrene microspheres of 80 nm (NPs) and 8 µm (MPs) as materials to simulate environmental contaminants in the aquatic environment with AED. We used female mosquitofish (Gambusia affinis) as the research object to investigate the effects of microplastics on fish in waters containing AED. We compared different sizes of particles accumulation in some tissues of fish and variation of enzyme activities (SOD, LDH, CAT), and the content of MDA in the gut. MPs, NPs, and AED combined exposure test investigated mRNA profiles of immune-related genes (IL-1ß, IL-6, IL-8, IL-10) and hormone receptor genes (ARα, ARß, ERα, ERß) in the liver of fish. Our results indicated that MPs emerged in various tissues (gill, gut, and liver) of mosquitofish. Besides, NPs and MPs caused enteric abnormal enzyme activity after 48 h of exposure, which was particularly pronounced in the MPs-AED group. MPs induced significant upregulation of inflammatory factors and gonadal factor genes after 96 h of exposure, which was more pronounced when co-exposed with AED. In conclusion, NPs and MPs caused mechanisms of immune damage and inflammatory response. MPs were found to be more likely to cause adverse reactions than NPs, and these responses were enhanced by the combined effects of AED. This study demonstrated that AED can exacerbate the negative effects of MPs and NPs on mosquitofish. It provided an important basis for the effective assessment of MPs and NPs on bioaccumulation and biochemical status of mosquitofish. Additionally, it serves as a foundation to investigate the interactive effects of microplastics and EDCs in living organisms.


Assuntos
Androstenodiona , Ciprinodontiformes , Disruptores Endócrinos , Microplásticos , Poluentes Químicos da Água , Animais , Feminino , Androstenodiona/toxicidade , Ciprinodontiformes/fisiologia , Disruptores Endócrinos/toxicidade , Microplásticos/toxicidade , Plásticos , Poliestirenos/toxicidade , Poluentes Químicos da Água/toxicidade , Poluentes Químicos da Água/análise
5.
Chem Pharm Bull (Tokyo) ; 59(3): 327-31, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21372413

RESUMO

Taking into consideration the structural requirements for cytotoxicity and aromatase inhibition, several new 16E-arylidenosteroidal derivatives have been prepared and evaluated for their cytotoxic and aromatase inhibitory activity. The new steroidal analogues 3, 5-8 and 11 exhibited significant cytotoxic effects when screened against three cancer cell lines, MCF-7 (breast), NCl-H460 (lung) and SF-268 central nervous system (CNS) at 100 µM and sensible cytotoxic effects subsequently in sixty cancer cell lines derived from nine cancers types (leukemia, lung, colon, CNS, melanoma, ovarian, renal, prostate and breast cancers). The imidazolyl substituted steroidal derivatives 5 and 7 exhibited strong inhibition of the aromatase enzyme with 16-[4-{3-(imidazol-1-yl)propoxy}-3-methoxybenzylidene]-5-androstene-3ß,17ß-diol (7) displaying 13 times more potency in comparison to aminoglutethimide.


Assuntos
Androstenodióis/síntese química , Androstenodiona/análogos & derivados , Antineoplásicos/síntese química , Inibidores da Aromatase/síntese química , Aromatase/química , Esteroides/química , Androstenodióis/química , Androstenodióis/toxicidade , Androstenodiona/síntese química , Androstenodiona/química , Androstenodiona/toxicidade , Antineoplásicos/química , Antineoplásicos/toxicidade , Aromatase/metabolismo , Inibidores da Aromatase/química , Inibidores da Aromatase/toxicidade , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Esteroides/síntese química , Esteroides/toxicidade
6.
Environ Toxicol ; 25(3): 261-71, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19489062

RESUMO

Brain estrogen production, performed by the enzyme aromatase, can be disrupted/affected in teleost fish exposed to endocrine disruptors found in polluted aquatic environments. The guppy (Poecilia reticulata) was previously studied and confirmed to suffer negative effects on reproductive behaviors following inhibition of the brain aromatase reaction. Here adult guppies (Poecilia reticulata) of both genders were subjected to known endocrine disruptors: the androgen androstenedione (A), the synthetic estrogen 17alpha-ethinylestradiol (EE(2)), and the estrogenic surfactant 4-nonylphenol (NP), at high (50 microg/L) and at environmentally relevant concentrations (10 ng/L EE(2), 5 microg/L NP, and 0.7 microg/L A) for 2 weeks followed by measurements of brain aromatase activity (bAA). In the adult males, bAA was stimulated by A and EE(2) at 50 microg/L. Female activity was also stimulated by the higher estrogenic treatment. At environmentally relevant concentrations only the EE(2) treatment affected bAA, and only in males. The alkylphenolic substance NP produced no effect in either of the experiments, not on males nor females. The results indicate that short-term steroid treatments have stimulatory effects on guppy brain aromatase even at concentrations that can be found in the environment. We thus suggest bAA of adult guppies to be a suitable bioindicator of endocrine disruptors.


Assuntos
Aromatase/metabolismo , Encéfalo/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Fenóis/toxicidade , Poecilia/crescimento & desenvolvimento , Esteroides/toxicidade , Androstenodiona/toxicidade , Animais , Comportamento Animal/efeitos dos fármacos , Biomarcadores/análise , Encéfalo/enzimologia , Relação Dose-Resposta a Droga , Monitoramento Ambiental , Etinilestradiol/toxicidade , Feminino , Masculino , Fatores Sexuais , Fatores de Tempo
7.
Natl Toxicol Program Tech Rep Ser ; (560): 1, 7-31,33-171 passim, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21037592

RESUMO

UNLABELLED: Androstenedione is an androgen steroid that is normally synthesized within men and women and may be metabolized to a more potent androgen or estrogen hormone. It was nominated to the National Toxicology Program for study due to concern for adverse health effects associated with its chronic use as a dietary supplement by athletes (prior to the banning of its over the counter sales). In order to evaluate its subchronic and chronic toxicity, male and female F344/N rats and B6C3F1 mice were administered androstenedione (98% pure) by gavage for 2 weeks, 3 months, or 2 years. Genetic toxicology studies were conducted in Salmonella typhimurium, Escherichia coli, rat bone marrow cells, and mouse peripheral blood erythrocytes. 2-WEEK STUDY IN RATS: groups of five male and five female rats were administered 0, 1, 5, 10, 20, or 50 mg androstenedione/kg body weight in a 0.5% aqueous methylcellulose solution by gavage, 5 days per week for 12 days. All rats survived to the end of the study, and the mean body weights of dosed groups were similar to those of the vehicle control groups. The development of cytoplasmic vacuoles within centrilobular hepatocytes in male rats was the only treatment-related effect observed. 2-WEEK STUDY IN MICE: groups of five male and five female mice were administered 0, 1, 5, 10, 20, or 50 mg androstenedione/kg body weight in a 0.5% aqueous methylcellulose solution by gavage, 5 days per week for 12 days. One vehicle control female, one 20 mg/kg female, and one 50 mg/kg female died early due to gavage accidents. There were no significant chemical-related histopathological or mean body weight changes. 3-MONTH STUDY IN RATS: groups of 10 male and 10 female core study rats were administered 0, 1, 5, 10, 20, or 50 mg androstenedione/kg body weight in a 0.5% aqueous methylcellulose solution by gavage, 5 days per week for 14 weeks; additional groups of 10 male and 10 female clinical pathology study rats received the same doses for 23 days. All rats survived to the end of the study. The mean body weights of the 20 mg/kg female group was significantly greater than those of the vehicle control group and there was significant increased weight gain in the 1, 20, and 50 mg/kg female groups. Female thymus weights were significantly increased in the 20 and 50 mg/kg groups, which may be related to the increase in mean body weight. The numbers of sperm per mg cauda epididymis in the 10, 20, and 50 mg/kg male groups and the total number of sperm per cauda epididymis in 50 mg/kg males were significantly less than those of the vehicle controls. No treatment-related histological lesions were observed in males or females. 3-MONTH STUDY IN MICE: groups of 10 male and 10 female mice were administered 0, 1, 5, 10, 20, or 50 mg androstenedione/kg body weight in a 0.5% aqueous methylcellulose solution by gavage, 5 days per week for 14 weeks. Except for one 10 mg/kg female that died early due to a dosing accident, all mice survived to the end of the study. The mean body weights of dosed groups were similar to those of the vehicle control groups. The number of spermatids per mg testis and the total number of spermatids per testis in 20 mg/kg males were significantly greater than those of the vehicle controls. Sperm motility in 50 mg/kg males was significantly lower than that in the vehicle controls. The incidences of x-zone atrophy of the adrenal cortex, an androgen-sensitive endpoint, were significantly increased in females administered 5 mg/kg or greater. There were also significant decreases in the incidences of x-zone cytoplasmic vacuolization in 20 and 50 mg/kg females. The incidences of bone marrow hyperplasia were significantly increased in 5 and 50 mg/kg males. 2-YEAR STUDY IN RATS: groups of 50 male and 50 female rats were administered 0, 10, 20, or 50 mg androstenedione/kg body weight in a 0.5% aqueous methylcellulose solution by gavage, 5 days per week for at least 104 weeks. Survival of 10 mg/kg males was significantly greater than that of the vehicle controls. The mean body weights of 20 and 50 mg/kg females were greater than those of the vehicle controls after weeks 17 and 9, respectively. The incidences of mononuclear cell leukemia were significantly increased in 20 and 50 mg/kg females and significantly decreased in 20 and 50 mg/kg males. Incidences of alveolar/bronchiolar adenoma and alveolar/bronchiolar adenoma or carcinoma (combined) were significantly increased in 20 mg/kg males. The incidence of testicular interstitial cell adenoma (including bilateral) was significantly decreased in 50 mg/kg males. In females, the incidences of mammary gland fibroadenoma were significantly decreased in the 20 and 50 mg/kg groups, the incidences of mammary gland hyperplasia were significantly decreased in all dosed groups, and the incidences of mammary gland cyst were significantly decreased in the 10 and 50 mg/kg groups. In the liver of males, the incidences of basophilic focus in all dosed groups, the incidence of clear cell focus in the 20 mg/kg group, and the incidence of eosinophilic focus in the 50 mg/kg group were significantly increased. The incidences of pancreatic islet hyperplasia and atrophy of the exocrine pancreas were significantly increased in 50 mg/kg females. 2-YEAR STUDY IN MICE: groups of 50 male and 50 female mice were administered 0, 2 (females only), 10, 20 (males only), or 50 mg androstenedione/kg body weight in a 0.5% aqueous methylcellulose solution by gavage, 5 days per week for at least 104 weeks. Survival of dosed groups was similar to that of the vehicle control groups. Mean body weights of 10 and 50 mg/kg females were generally less than those of the vehicle controls after weeks 81 and 17, respectively. The incidences of hepatocellular adenoma in males and females were significantly increased in the 50 mg/kg groups. In females, the incidences of hepatocellular carcinoma were significantly increased in all dosed groups. Incidences of hepatocellular adenoma or carcinoma (combined) in males and females were significantly increased in the 50 mg/kg groups. Incidences of hepatoblastoma were marginally increased in dosed males. Incidences of multiple hepatocellular adenomas and carcinomas were significantly increased in 10 and 50 mg/kg males, and there was an increased incidence of multiple hepatocellular adenomas in 50 mg/kg females. The incidence of eosinophilic focus was significantly increased in 50 mg/kg males, and the incidences of mixed cell focus and cytoplasmic vacuolization were significantly increased in 50 mg/kg females. There was a marginally increased incidence of pancreatic islet adenoma in 50 mg/kg males and in 10 and 50 mg/kg females, with an earlier day of first incidence in males. The incidences of clitoral gland hyperplasia and clitoral gland duct dilatation were significantly increased in 10 and 50 mg/kg females. The incidence of glomerular metaplasia of the kidney was significantly increased in 50 mg/kg females, and the incidences of cytoplasmic alteration of the submandibular salivary gland were significantly increased in all dosed female groups. The increased incidences of cytoplasmic alteration of the submandibular salivary gland and glomerular metaplasia of the kidney in female mice indicated a masculinizing effect from androstenedione treatment. In 50 mg/kg females, the incidence of malignant lymphoma was significantly decreased. GENETIC TOXICOLOGY: androstenedione was not mutagenic in either of two independent bacterial mutation assays conducted with and without exogenous metabolic activation. No significant increases in the frequencies of micronucleated polychromatic erythrocytes, indicators of chromosomal damage, were observed in bone marrow of male rats administered androstenedione by gavage once daily for 3 consecutive days. Results of a peripheral blood erythrocyte micronucleus test in mice, in which androstenedione was administered by gavage for 3 months, were negative in males but judged to be equivocal in females due to a small increase (twofold over background) in micronucleated normochromatic erythrocytes observed at the highest dose administered (50 mg/kg). CONCLUSIONS: under the conditions of these 2-year gavage studies, there was equivocal evidence of carcinogenic activity of androstenedione in male F344/N rats based on increased incidences of alveolar/bronchiolar adenoma and alveolar/bronchiolar adenoma or carcinoma (combined). There was equivocal evidence of carcinogenic activity of androstenedione in female F344/N rats based on increased incidences of mononuclear cell leukemia. There was clear evidence of carcinogenic activity of androstenedione in male B6C3F1 mice based on increased incidences of multiple hepatocellular adenoma and hepatocellular carcinoma and increased incidence of hepatoblastoma. There was clear evidence of carcinogenic activity of androstenedione in female B6C3F1 mice based on increased incidences of hepatocellular adenoma and hepatocellular carcinoma. Increased incidences of pancreatic islet adenoma in male and female mice were also considered chemical related. Androstenedione administration caused increased incidences in nonneoplastic lesions of the liver in male and female rats and mice; pancreatic islets and exocrine pancreas of female rats; and clitoral gland, kidney, and submandibular salivary gland of female mice. Decreases in the incidences of testicular interstitial cell adenoma in male rats, mammary gland fibroadenoma, cysts, and hyperplasia in female rats, and malignant lymphoma in female mice were considered related to androstenedione administration. Synonyms: Andro; androst-4-ene-3,17-dione; 4-androstene-3,17-dione; delta-4-androstene-3,17-dione; delta-4-androstenedione; 3,17-dioxoandrost-4-ene; 17-ketotestosterone; SKF 2170 Trade names: Androtex, Fecundin.


Assuntos
Androstenodiona/toxicidade , Androstenodiona/farmacologia , Ração Animal , Animais , Biomarcadores , Peso Corporal/efeitos dos fármacos , Testes de Carcinogenicidade , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Química Farmacêutica , Relação Dose-Resposta a Droga , Ciclo Estral , Feminino , Genitália/efeitos dos fármacos , Intubação Gastrointestinal , Masculino , Camundongos , Camundongos Endogâmicos , Testes para Micronúcleos , Mutagênicos/toxicidade , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344
8.
Environ Toxicol Pharmacol ; 68: 133-140, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30889543

RESUMO

Acute developmental exposure to pharmaceuticals or environmental contaminants can have deleterious, long lasting effects. Many of these compounds are endocrine disruptors (EDCs) that target estrogen signaling, with effects on reproductive and non-reproductive tissues. We recently reported that zebrafish larvae transiently exposed to the pharmaceutical EDC 4-OH-A display visual deficits as adults. Here, we examine whether these long-term effects are due to compound-induced morphological and/or cellular changes. Zebrafish aged 24 h, 48 h, 72 h, or 7 days post-fertilization (larvae) or 3-4mos (adults) were exposed to either 4-OH-A or PCB1254 for 24 h. After that time, notochord length, eye diameter, inter-eye distance, and heart rate were measured from larvae; and aromatase (estrogen synthase) activity was measured in homogenates of adult brain tissue. In general, indices of larval growth and development were not altered by 24 h exposure to either compound. 4-OH-A potently inhibited aromatase activity, while PCB1254 did not, with inhibition continuing even after removal from treatment. These results support differential function of EDCs and indicate that developmental exposure to 4-OH-A causes sustained inhibition of aromatase, which could be associated with altered adult behaviors.


Assuntos
Androstenodiona/análogos & derivados , Inibidores da Aromatase/toxicidade , Disruptores Endócrinos/toxicidade , Peixe-Zebra , Androstenodiona/toxicidade , Animais , Aromatase/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Larva/crescimento & desenvolvimento , Larva/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
9.
Toxicol Lett ; 177(3): 198-204, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18325697

RESUMO

One of the most frequently misused steroid precursors (prohormones) is 19-norandrostenedione (4-estrene-3,17-dione, NOR), which is, after oral administration, readily metabolised to nortestosterone, also known as nandrolone (durabolin). In this study we have characterised molecular mechanisms of its action determined its tissue specific androgenic and anabolic potency after subcutaneous (s.c.) administration and investigated potential adverse effects. Receptor binding tests demonstrate that NOR binds with high selectivity to the AR. The potency of NOR to transactivate androgen receptor (AR) dependent reporter gene expression was 10 times lower as compared to dihydrotestosterone (DHT). In vivo experiments in orchiectomised rats demonstrated that s.c. treatment with NOR resulted only in a stimulation of the weight of the levator ani muscle; the prostate and seminal vesicle weights remained completely unaffected. Like testosterone, administration of NOR resulted in a stimulation of AR and myostatin mRNA expression in the gastrocnemius muscle. NOR does not affect prostate proliferation, the liver weight and the expression of the tyrosine aminotransferase gene (TAT) in the liver. Summarizing these data it is obvious that NOR, if administrated s.c. and in contrast to its metabolite nandrolone, highly selectively stimulates the growth of the skeletal muscle but has only weak androgenic properties. This observation may have relevance with respect to therapeutic aspects but also doping prevention.


Assuntos
Anabolizantes/toxicidade , Androstenodiona/análogos & derivados , Receptores Androgênicos/efeitos dos fármacos , Androgênios , Androstenodiona/administração & dosagem , Androstenodiona/metabolismo , Androstenodiona/toxicidade , Animais , Injeções Subcutâneas , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Ratos , Ratos Wistar , Propionato de Testosterona/farmacologia
10.
Toxicol Lett ; 183(1-3): 58-64, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18951961

RESUMO

The abuse of anabolic steroids for doping raises concerns. Many of these compounds have never been examined for their toxicological properties. Aside from hormonal (androgenic) activity, anabolic steroids may also exert genotoxic effects. In the present study, we determined the potencies of the "designer steroid" madol (MAD) and the anabolic prohormone 19-norandrostenedione (NA) to induce micronuclei in V79 cells in vitro. CREST analysis was used to differentiate between aneugenic and clastogenic mechanisms of micronucleus induction. Cytotoxicity of the steroids and their influence on the cell cycle were assessed in parallel. In addition, the ability of MAD and NA to increase production of reactive oxygen species and to induce apoptosis were studied. Both agents caused a concentration-dependent increase in the rates of micronuclei in V79 cells, exceeding a doubling of the background micronucleus rates of untreated controls, which was evident at 27microM and 29microM for MAD and NA, respectively. The steroid-induced micronuclei were predominantly kinetochor (CREST)-negative, pointing to a clastogenic mode of action. As cytotoxicity of both compounds is weak (IC(20) value of 300microM for NA and IC(10) of 100microM for MAD), cytotoxicity was unlikely to contribute to their genotoxicity. The observed genotoxicity of both compounds was due neither to apoptosis induction nor to production of reactive oxygen species. However, the ability of both steroids to induce micronuclei appears related to their lipophilicity. Therefore, a "non-specific" chromosomal genotoxicity of MAD and NA, based on hydrophobic interactions, appears likely. This could well result in biologically relevant increases in chromosomal damage as soon as critical concentrations of the agents are reached in vivo. Regarding the current misuse of the steroids for doping, the uncontrolled administration of very high doses must be considered. Therefore it cannot be ruled out that MAD and NA present genotoxic hazards under current misuse conditions by athletes in sports or in body building.


Assuntos
Anabolizantes/toxicidade , Androstenodiona/análogos & derivados , Androstenóis/toxicidade , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Anabolizantes/química , Androstenodiona/química , Androstenodiona/toxicidade , Androstenóis/química , Animais , Apoptose/efeitos dos fármacos , Camptotecina/química , Camptotecina/toxicidade , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Dopagem Esportivo , Relação Dose-Resposta a Droga , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fase G2/efeitos dos fármacos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/toxicidade , Metanossulfonato de Metila/química , Metanossulfonato de Metila/toxicidade , Testes para Micronúcleos/métodos , Microscopia de Fluorescência/métodos , Estrutura Molecular , Mutagênicos/química , Mutagênicos/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Fase S/efeitos dos fármacos
11.
Ecotoxicol Environ Saf ; 71(3): 757-64, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18395257

RESUMO

Steroid compounds have been globally detected in surface waters. The ecological impacts of these biologically active chemicals are largely unknown. Toxicity of testosterone and 4-hydroxyandrostenedione was assessed for the freshwater cladoceran Daphnia magna. Acute toxicity tests showed that 6.20 mg L(-1) of testosterone, the highest concentration tested, did not have effect on the daphnids, whereas 4-hydroxyandrostenedione had an EC(50) of 4.26 mg L(-1). Chronic toxicity tests were carried out using survival, body length, fecundity, and fertility as endpoints. Long-term testosterone exposure reduced D. magna fecundity and fertility at concentrations ranging from 0.31 to 2.48 mg L(-1). The significant decrease in fecundity was associated with an increase in aborted eggs. Long-term 4-hydroxyandrostenedione exposure at 0.84 mg L(-1) increased the mortality of the neonates. The chronic toxicity effects were observed at concentrations higher than the measured environmental concentrations of these compounds. Nevertheless, the reproductive impairment of the daphnids is likely to occur at environmental levels as an ultimate response to long-term exposure.


Assuntos
Androstenodiona/análogos & derivados , Daphnia/efeitos dos fármacos , Testosterona/toxicidade , Poluentes Químicos da Água/toxicidade , Androstenodiona/toxicidade , Animais , Daphnia/fisiologia , Disruptores Endócrinos/toxicidade , Fertilidade/efeitos dos fármacos , Água Doce/química , Estágios do Ciclo de Vida/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Testes de Toxicidade Aguda , Testes de Toxicidade Crônica
12.
Aquat Toxicol ; 202: 1-5, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29960009

RESUMO

Steroid hormones in the aquatic environment may pose a risk to fish health. Here we evaluated effects of two different class steroids that frequently occur in the aquatic environment, the androgen androstenedione (A4) and the progestin 17α-hydroxyprogesterone (17-OHP4). Zebrafish embryos were exposed to four concentrations of A4 and the positive control testosterone and to 17-OHP4, and transcriptional changes were determined at 96 h post fertilization (hpf) and 120 hpf. Transcriptional changes of 18 selected genes were assessed upon exposure to measured concentrations of 0.004, 0.046, 0.62 and 6.56 µg/L A4. Significant induction of the genes encoding sulfotransferase (sult2st3) and aromatase (cyp19b) occurred in 120 hpf embryos at 6.56 µg/L A4 and 1 µg/L testosterone. Additionally, cyp2k7 was significantly induced in two of three independent experiments. 17-OHP4 did not induce physiological effects (muscle contraction, heart rate, hatching success, swimming activity) at concentrations between 0.01 and 10 µg/L. Of the analyzed 15 genes, slight transcriptional alterations occurred for the genes encoding progesterone receptor, aromatases (cyp19a) and (cyp19b) and cyp2k7 at 10 µg/L. Our study highlights sult2st3, cyp19b and cyp2k7 as potential markers of androgen exposure in fish and indicates that 17-OHP4 is not likely to pose a risk for fish at environmental concentrations.


Assuntos
Androstenodiona/toxicidade , Progesterona/análogos & derivados , Transcrição Gênica/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Aromatase/genética , Aromatase/metabolismo , Análise por Conglomerados , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Progesterona/toxicidade , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Sulfotransferases/genética , Sulfotransferases/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética
13.
Toxicol Lett ; 169(2): 129-36, 2007 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-17267145

RESUMO

The aim of this study is to investigate the effects in vitro induced by androgenic anabolic steroids (AAS) (testosterone, nandrolone, androstenedione, norandrostenedione, and norandrostenediol) used illicitly in sport competitions, on the proliferation ability, apoptosis and the intracellular calcium concentration ([Ca2+]i) in human umbilical vein endothelial cells (HUVECs), selected as a prototype of a biological target system whose structure and function can be affected by steroids. For this purpose, we evaluated the proliferation inhibition by cytotoxic assay expressed as the concentration of drug inducing a 50% decrease in growth (IC50). The IC50 was reached for testosterone at 100 microM, androstenedione at 375 microM, nandrolone at 9 microM, norandrostenedione at 500 microM. The IC50 value for norandrostenediol was not reached until a concentration of 6000 microM. The apoptotic effect was evaluated by flow cytometry at IC50 for each drug. We observed that testosterone induced 31% of apoptotic cells, norandrostenedione 25%, androstenedione 15% and nandrolone 18%. We have analyzed the effects of these drugs on [Ca2+]i both in the immediate and long-term continuous presence of each compound. Our data show a statistically significant increase of [Ca2+]i in the acute condition and in long-term treated cultures, suggesting that androgen steroids modulate intracellular levels of calcium independent of incubation time or compound identity. As a whole, this study demonstrates that AAS might alter endothelial homeostasis, predisposing to the early endothelial cell activation that is responsible for vascular complications observed frequently in AAS users.


Assuntos
Anabolizantes/toxicidade , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Esteroides/toxicidade , Androstenodiona/toxicidade , Cálcio/metabolismo , Dopagem Esportivo , Endotélio Vascular/metabolismo , Citometria de Fluxo , Fluorometria , Humanos , Concentração Inibidora 50 , Nandrolona/toxicidade , Testosterona/toxicidade
14.
Environ Toxicol Chem ; 26(5): 920-6, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17521138

RESUMO

In this study, we investigated the capacity of androstenedione to masculinize female mosquitofish. Previous studies have identified androstenedione in the water and sediment of the Fenholloway River, a Florida, USA, coastal river that receives paper mill effluent and contains masculinized eastern mosquitofish (Gambusia holbrooki). Females of the closely related western mosquitofish, Gambusia affinis, were exposed to androstenedione through both dietary and static renewal treatments. Morphological masculinization of female mosquitofish is characterized by the development of a male secondary sexual trait: an elongated and modified anal fin (gonopodium). Dietary exposure to 0.7, 7, 70, and 700 microg of androstenedione per gram of food failed to induce gonopodial development at any concentration within the six-week exposure period. Static renewal treatments used androstenedione concentrations of 0.14, 1.4, 14, 140, and 350 nM. Significant anal fin ray elongation was observed in all but the lowest exposure group. Fish growth during the static renewal exposure experiment was negatively correlated with androstenedione concentration. No significant effects were observed for gonadosomatic index, vitellogenin expression, or ovarian area in fish exposed to androstenedione via either the dietary or static renewal methods. These results indicate that exposure to androstenedione via water can cause masculinization of adult female mosquitofish in a relatively short period of time and that acute dietary exposure to androstenedione at the concentrations used is not sufficient to induce masculinization.


Assuntos
Androstenodiona/toxicidade , Ciprinodontiformes/fisiologia , Dieta/veterinária , Caracteres Sexuais , Poluentes Químicos da Água/toxicidade , Animais , Feminino , Florida , Resíduos Industriais , Masculino , Medição de Risco , Rios , Processos de Determinação Sexual , Estados Unidos
15.
Toxicol Sci ; 160(1): 15-29, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973534

RESUMO

Current strategies for predicting carcinogenic mode of action for nongenotoxic chemicals are based on identification of early key events in toxicity pathways. The goal of this study was to evaluate short-term key event indicators resulting from exposure to androstenedione (A4), an androgen receptor agonist and known liver carcinogen in mice. Liver cancer is more prevalent in men compared with women, but androgen-related pathways underlying this sex difference have not been clearly identified. Short-term hepatic effects of A4 were compared with reference agonists of the estrogen receptor (ethinyl estradiol, EE) and glucocorticoid receptor (prednisone, PRED). Male B6C3F1 mice were exposed for 7 or 28 days to A4, EE, or PRED. EE increased and PRED suppressed hepatocyte proliferation, while A4 had no detectable effects. In a microarray analysis, EE and PRED altered >3000 and >670 genes, respectively, in a dose-dependent manner, whereas A4 did not significantly alter any genes. Gene expression was subsequently examined in archival liver samples from male and female B6C3F1 mice exposed to A4 for 90 days. A4 altered more genes in females than males and did not alter expression of genes linked to activation of the mitogenic xenobiotic receptors AhR, CAR, and PPARα in either sex. A gene expression biomarker was used to show that in female mice, the high dose of A4 activated the growth hormone-regulated transcription factor STAT5b, which controls sexually dimorphic gene expression in the liver. These findings suggest that A4 induces subtle age-related effects on STAT5b signaling that may contribute to the higher risk of liver cancer in males compared with females.


Assuntos
Androstenodiona/toxicidade , Biomarcadores Tumorais/genética , Transformação Celular Neoplásica/química , Transformação Celular Neoplásica/genética , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/genética , Fígado/efeitos dos fármacos , Animais , Biomarcadores Tumorais/metabolismo , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Relação Dose-Resposta a Droga , Etinilestradiol/toxicidade , Feminino , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Fenótipo , Prednisona/toxicidade , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Fatores Sexuais , Fatores de Tempo , Transcriptoma
16.
J Steroid Biochem Mol Biol ; 100(1-3): 87-94, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16713252

RESUMO

Methyl protodioscin (1), a natural furostanol biglycoside steroid, was a preclinical anticancer drug, which showed potent activity against most cell lines from leukemia and solid tumors in the National Cancer Institute's (NCI) human cancer panel. Metabolism of methyl protodioscin by Aspergillus niger was investigated. Seven metabolites were isolated and identified. Two main metabolites were pregnane glycosides and four were furostanol glycosides, together with the aglycone. It was found that steroidal saponin skeleton could be converted to pregnenolone skeleton only using microbial methods, which must have chemical procedures in the reported literatures. The proposed biosynthetic pathways of the microbial conversion products of methyl protodioscin were drawn. The found enriched the reaction types of microbial bioconversion and provided a new producing way of androstenedione from steroid. Most metabolites showed strong cytotoxic activities against HepG2, NCI-H460, HeLa, and MCF-7 cell lines.


Assuntos
Androstenodiona/metabolismo , Aspergillus niger/metabolismo , Diosgenina/análogos & derivados , Saponinas/metabolismo , Esteroides/metabolismo , Adenocarcinoma/tratamento farmacológico , Androstenodiona/química , Androstenodiona/toxicidade , Aspergillus niger/genética , Biotransformação , Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Diosgenina/química , Diosgenina/metabolismo , Diosgenina/toxicidade , Relação Dose-Resposta a Droga , Feminino , Células HeLa , Humanos , Concentração Inibidora 50 , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Estrutura Molecular , Saponinas/química , Saponinas/toxicidade , Neoplasias do Colo do Útero/tratamento farmacológico
17.
Food Chem Toxicol ; 44(4): 579-87, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16289525

RESUMO

Androstenedione, a steroidal dietary supplement taken to enhance athletic performance, could affect serum and liver lipid metabolism, induce liver toxicity or alter inflammatory response depending on dose and duration of exposure. Pregnancy could further exaggerate these effects. To examine this, mature female rats were gavaged with 0, 5, 30 or 60 mg/kg/day androstenedione beginning two weeks prior to mating and continuing through gestation day 19. Non-pregnant female rats were gavaged over the same time frame with 0 or 60 mg/kg/day androstenedione. Serum was collected and livers were removed from dams on gestation day 20 and from non-pregnant rats after 5 weeks of treatment. Androstenedione had no effect on serum total cholesterol, triglycerides or HDL-cholesterol, but significantly decreased C-reactive protein in pregnant rats and prostaglandin E(2) in serum of both pregnant and non-pregnant rats. There were treatment related decreases in liver ATP and, to a lesser degree, caspase-3 and no change in alkaline phosphatase of pregnant female rats. Androstenedione decreased docosahexaenoic acid in both serum and liver phospholipids of pregnant female rats. In conclusion, oral androstenedione did not result in overt hepatotoxicity in pregnant female rats, but produced modest changes in lipid metabolism and may impair regeneration of injured hepatic cells or tissue.


Assuntos
Androstenodiona/toxicidade , Trifosfato de Adenosina/sangue , Trifosfato de Adenosina/metabolismo , Administração Oral , Androstenodiona/administração & dosagem , Animais , Proteína C-Reativa/efeitos dos fármacos , Proteína C-Reativa/metabolismo , Caspase 3 , Caspases/sangue , Caspases/efeitos dos fármacos , Caspases/metabolismo , Dinoprostona/sangue , Dinoprostona/metabolismo , Relação Dose-Resposta a Droga , Ácidos Graxos/sangue , Ácidos Graxos/metabolismo , Feminino , Fígado/efeitos dos fármacos , Fígado/enzimologia , Gravidez , Ratos
18.
Aquat Toxicol ; 80(3): 237-48, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16996625

RESUMO

This study combined bioassay-derived and direct chemical analysis of steroidal compounds in pulp and paper and municipal sewage effluent in order to determine the cause of masculinization of female mosquitofish. The bioassays used in this study consisted of androgen and estrogen receptor binding, and aromatase inhibition using tissues from rainbow trout. This study observed no masculinization of female mosquitofish from a pulp and paper mill effluent that was previously observed to cause this effect. Mosquitofish sampled from the receiving environment of the same mill verified that masculinization was not occurring in the wild. The municipal sewage effluent also had no masculinizing effect. In vitro bioassays indicated significant sources of both androgens and estrogens in the effluents tested with sewage effluent having both the highest estradiol (41 ng/L) and testosterone (182 ng/L) equivalent concentration. These results could not be attributed to any particular compounds measured in the effluents. Two compounds implicated in the masculinization of mosquitofish by pulp and paper effluent, androstenedione and androstadienedione required relatively large (10-100 microg/L) waterborne concentrations to elicit a masculinizing effect and neither of these compounds are likely to occur at levels this high in the natural environment. The potent aromatase inhibitor, 4-hydroxyandrostenedione also did not cause masculinization at 100 microg/L indicating that masculinization did not occur through this mechanism. The mammalian anti-androgen, cyproterone acetate was only partially effective in mosquitofish and reduced the severity of masculinization in the presence of methyl testosterone. While neither effluent masculinized mosquitofish, there was a significant reduction of in vitro ovarian steroid production with the most severe effects observed with the sewage effluent. Overall, this study found the disappearance of a masculinizing effect that had been previously observed; concluded that based on 21 days aqueous exposures androstenedione and androstadienedione are not likely candidates for mosquitofish masculinization, and showed that masculinization and in vitro steroid production are unrelated biological endpoints.


Assuntos
Androgênios/análise , Ciprinodontiformes/fisiologia , Monitoramento Ambiental/métodos , Oncorhynchus mykiss/fisiologia , Poluentes Químicos da Água/análise , Androgênios/toxicidade , Androstadienos/análise , Androstadienos/toxicidade , Androstenodiona/análise , Androstenodiona/toxicidade , Animais , Inibidores da Aromatase/análise , Bioensaio/métodos , Relação Dose-Resposta a Droga , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Resíduos Industriais/análise , Ovário/efeitos dos fármacos , Esgotos/análise , Esteroides/análise , Esteroides/biossíntese , Poluentes Químicos da Água/toxicidade
19.
Clin Cancer Res ; 4(3): 697-711, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9533540

RESUMO

Although the antiestrogen tamoxifen has been the mainstay of therapy for estrogen receptor (ER)-positive breast cancer, successful treatment of responsive tumors is often followed by the acquisition of tamoxifen resistance. Subsequently, only 30-40% of patients have a positive response to second hormonal therapies. This lack of response might be explained by mechanisms for tamoxifen resistance that sensitize ER pathways to small amounts of estrogenic activity present in tamoxifen or that bypass ER pathways completely. To elucidate one possible mechanism of tamoxifen resistance, we treated ovariectomized tumor-bearing mice injected with fibroblast growth factor (FGF)-transfected MCF-7 breast carcinoma cells with the steroidal antiestrogen ICI 182,780 or one of two aromatase inhibitors, 4-OHA or letrozole. These treatments did not slow estrogen-independent growth or prevent metastasis of tumors produced by FGF-transfected MCF-7 cells in ovariectomized nude mice. FGF-transfected cells had diminished responses to ICI 182,780 in vitro, suggesting that autocrine activity of the transfected FGF may be replacing estrogen as a mitogenic stimulus for tumor growth. ER levels in FGF transfectants were not down-regulated, and basal levels of transcripts for estrogen-induced genes or of ER-mediated transcription of estrogen response element (ERE) luciferase reporter constructs in the FGF expressing cells were not higher than parental cells, implying that altered hormonal responses are not due to down-regulation of ER or to FGF-mediated activation of ER. These studies indicate that estrogen independence may be achieved through FGF signaling pathways independent of ER pathways. If so, therapies directed at the operative mechanism might produce a therapeutic response or allow a response to a second course of antiestrogen treatment.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores da Aromatase , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Estradiol/análogos & derivados , Fatores de Crescimento de Fibroblastos/fisiologia , Tamoxifeno/uso terapêutico , Androstenodiona/análogos & derivados , Androstenodiona/uso terapêutico , Androstenodiona/toxicidade , Animais , Antineoplásicos/toxicidade , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Cloranfenicol O-Acetiltransferase/biossíntese , Inibidores Enzimáticos/uso terapêutico , Inibidores Enzimáticos/toxicidade , Estradiol/uso terapêutico , Estradiol/toxicidade , Antagonistas de Estrogênios/uso terapêutico , Antagonistas de Estrogênios/toxicidade , Feminino , Fatores de Crescimento de Fibroblastos/biossíntese , Fulvestranto , Humanos , Letrozol , Luciferases/biossíntese , Camundongos , Camundongos Nus , Nitrilas/uso terapêutico , Nitrilas/toxicidade , Ovariectomia , Reação em Cadeia da Polimerase , RNA Mensageiro/biossíntese , Receptores de Progesterona/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Tamoxifeno/toxicidade , Transcrição Gênica , Transfecção , Transplante Heterólogo , Triazóis/uso terapêutico , Triazóis/toxicidade , Células Tumorais Cultivadas
20.
Food Chem Toxicol ; 43(4): 505-13, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15721196

RESUMO

Thirty-day old female rats received corn oil or androstenedione (in corn oil) at one of four concentrations (5.0, 10.0, 30.0 or 60.0 mg/kg body weight) by gavage for two weeks prior to mating, during the mating period and until gestation day (GD) 19. Caesarean sections were performed on GD 20. No dose related changes were observed in serum androstenedione, estradiol, LH, FSH, testosterone or progesterone. A statistically significant decrease in estrous cycle length was observed in the 60.0 mg/kg dose group only. Feed and fluid consumption, mean body weight gain, organ weight and fetal parameters were not affected by androstenedione treatment. At the doses given, androstenedione had no specific effect on the development of individual bones or soft tissues.


Assuntos
Androstenodiona/toxicidade , Estro/efeitos dos fármacos , Desenvolvimento Fetal/efeitos dos fármacos , Troca Materno-Fetal , Administração Oral , Animais , Osso e Ossos/embriologia , Relação Dose-Resposta a Droga , Estro/fisiologia , Feminino , Masculino , Gravidez , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA