Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 237
Filtrar
1.
J Appl Toxicol ; 44(2): 287-300, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37700462

RESUMO

The HUMMIC skin-liver Chip2 microphysiological system using EpiDerm™ and HepaRG and stellate liver spheroids was used to evaluate the route-specific metabolism and toxicodynamic effects of genistein. Human-relevant exposure levels were compared: 60 nM representing the plasma concentration expected after topical application of a cosmetic product and 1 µM representing measured plasma concentrations after ingesting soya products. Genistein was applied as single and repeated topical and/or systemic doses. The kinetics of genistein and its metabolites were measured over 5 days. Toxicodynamic effects were measured using transcriptional analyses of skin and liver organoids harvested on Days 2 and 5. Route-specific differences in genistein's bioavailability were observed, with first-pass metabolism (sulfation) occurring in the skin after topical application. Only repeated application of 1 µM, resembling daily oral intake of soya products, induced statistically significant changes in gene expression in liver organoids only. This was concomitant with a much higher systemic concentration of genistein which was not reached in any other dosing scenario. This suggests that single or low doses of genistein are rapidly metabolised which limits its toxicodynamic effects on the liver and skin. Therefore, by facilitating longer and/or repeated applications, the Chip2 can support safety assessments by linking relevant gene modulation with systemically available parent or metabolite(s). The rate of metabolism was in accordance with the short half-life observed in in vivo in humans, thus supporting the relevance of the findings. In conclusion, the skin-liver Chip2 provides route-specific information on metabolic fate and toxicodynamics that may be relevant to safety assessment.


Assuntos
Genisteína , Pele , Humanos , Genisteína/toxicidade , Toxicocinética , Fígado
2.
Ecotoxicol Environ Saf ; 275: 116262, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38569320

RESUMO

The aryl hydrocarbon receptor (AHR) is a key ligand-dependent transcription factor that mediates the toxic effects of compounds such as dioxin. Recently, natural ligands of AHR, including flavonoids, have been attracting physiological and toxicological attention as they have been reported to regulate major biological functions such as inflammation and anti-cancer by reducing the toxic effects of dioxin. Additionally, it is known that natural AHR ligands can accumulate in wildlife tissues, such as fish. However, studies in fish have investigated only a few ligands in experimental fish species, and the AHR response of marine fish to natural AHR ligands of various other structures has not been thoroughly investigated. To explore various natural AHR ligands in marine fish, which make up the most fish, it is necessary to develop new screening methods that consider the specificity of marine fish. In this study, we investigated the response of natural ligands by constructing in vitro and in silico experimental systems using red seabream as a model species. We attempted to develop a new predictive model to screen potential ligands that can induce transcriptional activation of red seabream AHR1 and AHR2 (rsAHR1 and rsAHR2). This was achieved through multiple analyses using in silico/ in vitro data and Tox21 big data. First, we constructed an in vitro reporter gene assay of rsAHR1 and rsAHR2 and measured the response of 10 representatives natural AHR ligands in COS-7 cells. The results showed that FICZ, Genistein, Daidzein, I3C, DIM, Quercetin and Baicalin induced the transcriptional activity of rsAHR1 and rsAHR2, while Resveratrol and Retinol did not induce the transcriptional activity of rsAHR isoforms. Comparing the EC50 values of the respective compounds in rsAHR1 and rsAHR2, FICZ, Genistein, and Daidzein exhibited similar isoform responses, but I3C, Baicalin, DIM and Quercetin show the isoform-specific responses. These results suggest that natural AHR ligands have specific profiling and transcriptional activity for each rsAHR isoform. In silico analysis, we constructed homology models of the ligand binding domains (LBDs) of rsAHR1 and rsAHR2 and calculated the docking energies (U_dock values) of natural ligands with measured in vitro transcriptional activity and dioxins reported in previous studies. The results showed a significant correlation (R2=0.74(rsAHR1), R2=0.83(rsAHR2)) between docking energy and transcriptional activity (EC50) value, suggesting that the homology model of rsAHR1 and rsAHR2 can be utilized to predict the potential transactivation of ligands. To broaden the applicability of the homology model to diverse compound structures and validate the correlation with transcriptional activity, we conducted additional analyses utilizing Tox21 big data. We calculated the docking energy values for 1860 chemicals in both rsAHR1 and rsAHR2, which were tested for transcriptional activation in Tox21 data against human AHR. By comparing the U_dock energy values between 775 active compounds and 1085 inactive compounds, a significant difference (p<0.001) was observed between the U_dock energy values in the two groups, suggesting that the U_dock value can be applied to distinguish the activation of compounds. Furthermore, we observed a significant correlation (R2=0.45) between the AC50 of Tox21 database and U_dock values of human AHR model. In conclusion, we calculated equations to translate the results of an in silico prediction model for ligand screening of rsAHR1 and rsAHR2 transactivation. This ligand screening model can be a powerful tool to quantitatively estimate AHR transactivation of major marine agents to which red seabream may be exposed. The study introduces a new screening approach for potential natural AHR ligands in marine fish, based on homology model-docking energy values of rsAHR1 and rsAHR2, with implications for future agonist development and applications bridging in silico and in vitro data.


Assuntos
Dioxinas , Dibenzodioxinas Policloradas , Dourada , Animais , Humanos , Dourada/genética , Dourada/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Dioxinas/metabolismo , Ligantes , Quercetina , Genisteína/toxicidade , Genisteína/metabolismo , Dibenzodioxinas Policloradas/metabolismo , Isoformas de Proteínas/genética
3.
Arch Toxicol ; 97(6): 1547-1575, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37087486

RESUMO

In next generation risk assessment (NGRA), the Dietary Comparator Ratio (DCR) can be used to assess the safety of chemical exposures to humans in a 3R compliant approach. The DCR compares the Exposure Activity Ratio (EAR) for exposure to a compound of interest (EARtest) to the EAR for an established safe exposure level to a comparator compound (EARcomparator), acting by the same mode of action. It can be concluded that the exposure to a test compound is safe at a corresponding DCR ≤ 1. In this study, genistein (GEN) was selected as a comparator compound by comparison of reported safe internal exposures to GEN to its BMCL05, as no effect level, the latter determined in the in vitro estrogenic MCF7/Bos proliferation, T47D ER-CALUX, and U2OS ERα-CALUX assay. The EARcomparator was defined using the BMCL05 and EC50 values from the 3 in vitro assays and subsequently used to calculate the DCRs for exposures to 14 test compounds, predicting the (absence of) estrogenicity. The predictions were evaluated by comparison to reported in vivo estrogenicity in humans for these exposures. The results obtained support in the DCR approach as an important animal-free new approach methodology (NAM) in NGRA and show how in vitro assays can be used to define DCR values.


Assuntos
Estrogênios , Receptores de Estrogênio , Humanos , Estrogênios/toxicidade , Linhagem Celular Tumoral , Genisteína/toxicidade , Medição de Risco
4.
J Appl Toxicol ; 43(12): 1916-1925, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37551860

RESUMO

The isoflavones genistein and daidzein are flavonoid compounds mainly found in legumes, especially in soybeans and their derived products. These flavonoids can be present in agricultural, domestic and industrial wastewater effluents as a result of anthropogenic activities and may be discharged in the environment. Due to the large growth of the aquaculture sector in recent decades, new and cost-effective fish feeds are being sought, but there is also a particular need to determine the effects of exposed flavonoids on fish in the aquatic environment, as this is the main route of exposure of organisms to endocrine disruptors. This study evaluated the possible effects of these isoflavones on juveniles of Solea senegalensis and Solea solea. After 48-96 h of exposure, the acetylcholinesterase activity in the sole head tissues was measured, and the cholinesterase activity in juveniles of common sole (S. solea) was determined. Experiments were carried out to determine the optimal pH, investigate the specificity of three substrates (acetylthiocholine, butyrylthiocholine, propionylthiocholine) on cholinesterase activity and determine the kinetic parameters (Vmax and Km ). The results obtained showed that neither genistein nor daidzein exposure to S. senegalensis and S. solea inhibited the activity of acetylcholinesterase at the tested concentrations (genistein: 1.25, 2.5, 5, 10 and 20 mg/L; daidzein: 0.625, 1.25, 2.5, 5 and 10 mg/L).


Assuntos
Linguados , Isoflavonas , Animais , Genisteína/toxicidade , Acetilcolinesterase , Isoflavonas/farmacologia , Metamorfose Biológica
5.
Int J Mol Sci ; 24(2)2023 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-36674726

RESUMO

Perinatal exposure to endocrine disrupting chemicals (EDCs) has been shown to affect male reproductive functions. However, the effects on male reproduction of exposure to EDC mixtures at doses relevant to humans have not been fully characterized. In previous studies, we found that in utero exposure to mixtures of the plasticizer di(2-ethylhexyl) phthalate (DEHP) and the soy-based phytoestrogen genistein (Gen) induced abnormal testis development in rats. In the present study, we investigated the molecular basis of these effects in adult testes from the offspring of pregnant SD rats gavaged with corn oil or Gen + DEHP mixtures at 0.1 or 10 mg/kg/day. Testicular transcriptomes were determined by microarray and RNA-seq analyses. A protein analysis was performed on paraffin and frozen testis sections, mainly by immunofluorescence. The transcription factor forkhead box protein 3 (FOXA3), a key regulator of Leydig cell function, was identified as the most significantly downregulated gene in testes from rats exposed in utero to Gen + DEHP mixtures. FOXA3 protein levels were decreased in testicular interstitium at a dose previously found to reduce testosterone levels, suggesting a primary effect of fetal exposure to Gen + DEHP on adult Leydig cells, rather than on spermatids and Sertoli cells, also expressing FOXA3. Thus, FOXA3 downregulation in adult testes following fetal exposure to Gen + DEHP may contribute to adverse male reproductive outcomes.


Assuntos
Dietilexilftalato , Disruptores Endócrinos , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Feminino , Humanos , Ratos , Masculino , Animais , Testículo/metabolismo , Disruptores Endócrinos/efeitos adversos , Dietilexilftalato/toxicidade , Dietilexilftalato/metabolismo , Ratos Sprague-Dawley , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Genisteína/toxicidade , Fator 3-gama Nuclear de Hepatócito/metabolismo
6.
Biol Reprod ; 107(5): 1360-1373, 2022 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-35948002

RESUMO

It is controversial whether exposure to isoflavones exerts male reproductive toxicity. The aim of this study was to investigate whether isoflavone exposure during adulthood could have deleterious impacts on male reproductive health by the cross-sectional study, animal experiments, and in vitro tests. In the cross-sectional study, we observed that urinary isoflavones were not significantly associated with semen quality including sperm concentrations, sperm count, progressive motility, and total motility, respectively. However, negative associations were found between plasma testosterone and urinary Σisoflavones, genistein, glycitein, and dihydrodaidzein. In the animal experiments, serum and intratesticular testosterone levels were decreased in mice exposed to several dosages of genistein. Genistein administration caused upregulation of estrogen receptor alpha and downregulation of cytochrome P45017A1 protein levels in testes of mice. In vitro tests showed that genistein caused a concentration-dependent inhibition of testosterone production by TM3 Leydig cells. Elevated protein expression of estrogen receptor alpha and decreased messenger RNA/protein level of cytochrome P45017A1 were also observed in genistein-treated cells. Protein level of cytochrome P45017A1 and testosterone concentration were significantly restored in the estrogen receptor alpha small interferring RNA-transfected cells, compared to cells that treated with genistein alone. The results demonstrate that exposure to isoflavones during adulthood may be associated with alterations of reproductive hormones. Particularly, genistein, which inhibits testosterone biosynthesis through upregulation of estrogen receptor alpha in Leydig cells of mice, might induce the disruption of testosterone production in human. The present study provides novel perspective into potential targets for male reproductive compromise induced by isoflavone exposure.


Assuntos
Genisteína , Isoflavonas , Humanos , Adulto , Masculino , Camundongos , Animais , Genisteína/toxicidade , Receptor alfa de Estrogênio , Análise do Sêmen , Estudos Transversais , Sêmen , Isoflavonas/efeitos adversos , Testosterona , Citocromos
7.
Arch Toxicol ; 96(12): 3385-3402, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35986755

RESUMO

Humans and animals are exposed to multiple substances in their food and feed that might have a negative health impact. Among these substances, the Fusarium mycoestrogen zearalenone (ZEN) and its metabolites α-zearalenol (α-ZEL) and α-zearalanol (α-ZAL) are known to possess endocrine disruptive properties. In a mixed diet or especially animal feed, these potential contaminants might be ingested together with naturally occurring phytoestrogens such as soy isoflavones. So far, risk assessment of potential endocrine disruptors is usually based on adverse effects of single compounds whereas studies investigating combinatorial effects are scarce. In the present study, we investigated the estrogenic potential of mycoestrogens and the isoflavones genistein (GEN), daidzein (DAI) and glycitein (GLY) as well as equol (EQ), the gut microbial metabolite of DAI, in vitro alone or in combination, using the alkaline phosphatase (ALP) assay in Ishikawa cells. In the case of mycoestrogens, the tested concentration range included 0.001 to 10 nM with multiplication steps of 10 in between, while for the isoflavones 1000 times higher concentrations were investigated. For the individual substances the following order of estrogenicity was obtained: α-ZEL > α-ZAL > ZEN > GEN > EQ > DAI > GLY. Most combinations of isoflavones with mycoestrogens enhanced the estrogenic response in the investigated concentrations. Especially lower concentrations of ZEN, α-ZEL and α-ZAL (0.001-0.01 nM) in combination with low concentrations of GEN, DAI and EQ (0.001-0.1 µM) strongly increased the estrogenic response compared to the single substances.


Assuntos
Disruptores Endócrinos , Isoflavonas , Zearalenona , Zeranol , Humanos , Animais , Zearalenona/toxicidade , Zearalenona/metabolismo , Equol , Fitoestrógenos/toxicidade , Genisteína/toxicidade , Disruptores Endócrinos/toxicidade , Fosfatase Alcalina , Estrona
8.
Int J Mol Sci ; 22(12)2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-34208060

RESUMO

Mycotoxins are toxic metabolites of filamentous fungi. Previous studies demonstrated the co-occurrence of Fusarium and Alternaria toxins, including zearalenone (ZEN), ZEN metabolites, and alternariol (AOH). These xenoestrogenic mycotoxins appear in soy-based meals and dietary supplements, resulting in the co-exposure to ZEN and AOH with the phytoestrogen genistein (GEN). In this study, the cytotoxic and estrogenic effects of ZEN, reduced ZEN metabolites, AOH, and GEN are examined to evaluate their individual and combined impacts. Our results demonstrate that reduced ZEN metabolites, AOH, and GEN can aggravate ZEN-induced toxicity; in addition, the compounds tested exerted mostly synergism or additive combined effects regarding cytotoxicity and/or estrogenicity. Therefore, these observations underline the importance and the considerable risk of mycotoxin co-exposure and the combined effects of mycoestrogens with phytoestrogens.


Assuntos
Estrogênios/toxicidade , Genisteína/toxicidade , Lactonas/toxicidade , Zearalenona/metabolismo , Zearalenona/toxicidade , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Genisteína/química , Células HeLa , Humanos , Lactonas/química , Micotoxinas/toxicidade , Oxirredução , Zearalenona/química
9.
Int J Mol Sci ; 22(9)2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-34062716

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is considered the most common liver disorder, affecting around 25% of the population worldwide. It is a complex disease spectrum, closely linked with other conditions such as obesity, insulin resistance, type 2 diabetes mellitus, and metabolic syndrome, which may increase liver-related mortality. In light of this, numerous efforts have been carried out in recent years in order to clarify its pathogenesis and create new prevention strategies. Currently, the essential role of environmental pollutants in NAFLD development is recognized. Particularly, endocrine-disrupting chemicals (EDCs) have a notable influence. EDCs can be classified as natural (phytoestrogens, genistein, and coumestrol) or synthetic, and the latter ones can be further subdivided into industrial (dioxins, polychlorinated biphenyls, and alkylphenols), agricultural (pesticides, insecticides, herbicides, and fungicides), residential (phthalates, polybrominated biphenyls, and bisphenol A), and pharmaceutical (parabens). Several experimental models have proposed a mechanism involving this group of substances with the disruption of hepatic metabolism, which promotes NAFLD. These include an imbalance between lipid influx/efflux in the liver, mitochondrial dysfunction, liver inflammation, and epigenetic reprogramming. It can be concluded that exposure to EDCs might play a crucial role in NAFLD initiation and evolution. However, further investigations supporting these effects in humans are required.


Assuntos
Disruptores Endócrinos/toxicidade , Poluentes Ambientais/toxicidade , Metabolismo dos Lipídeos/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Compostos Benzidrílicos/toxicidade , Cumestrol/toxicidade , Dioxinas/toxicidade , Disruptores Endócrinos/classificação , Genisteína/toxicidade , Humanos , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/patologia , Fenóis/toxicidade , Fitoestrógenos/toxicidade , Bifenilos Policlorados/toxicidade
10.
Environ Res ; 190: 109975, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32827888

RESUMO

Living species including humans are continuously exposed to low levels of a myriad of endocrine active compounds that may affect their reproductive function. In contrast, experimental designs scrutinizing this question mostly consider the gestational/lactational period, select high unrealistic doses and, have rarely investigated the possible reproductive consequences in the progeny. The present study aimed at assessing comparatively a set of male reproductive endpoints according to exposure windows, gestational/lactational versus pre-pubertal to adulthood, using low doses of endocrine active substances in male rats as well as their unexposed male progeny. Animals were orally exposed to 1 mg/kg bw/d of genistein and/or vinclozolin, from conception to weaning or from prepuberty to young adulthood. A number of reproductive endpoints were assessed as well as testicular mRNA expression profiles, in the exposed rats and their unexposed progeny. Overall, the low dosage used only affected weakly most of classical reproductive endpoints. However, the gestational/lactational exposure to vinclozolin alone or combined to genistein significantly delayed the puberty onset. Contrasting with the gestational/lactational exposure, a decreased sperm production was found in the animals exposed to genistein and vinclozolin from the pre-pubertal period but also in their progeny for vinclozolin and the mixture. The expression level of several genes involved in meiosis, apoptosis and steroidogenesis was also affected differentially as a function of the exposure window in both exposed rats and unexposed offspring. We also provide further evidence that doses of endocrine active substances relevant with human exposure may affect the male reproductive phenotype and testicular transcriptome in the exposed generation as well as in the indirectly exposed offspring.


Assuntos
Genisteína , Efeitos Tardios da Exposição Pré-Natal , Adulto , Animais , Feminino , Expressão Gênica , Genisteína/toxicidade , Humanos , Masculino , Oxazóis/toxicidade , Gravidez , Ratos , Testículo , Adulto Jovem
11.
Arch Toxicol ; 94(5): 1625-1635, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32185415

RESUMO

P-glycoprotein (P-gp) is an ABC transporter exhibiting high pharmacotoxicological relevance by extruding a wide range of cytotoxic compounds out of the cells. Previously, we demonstrated that the phytoestrogen genistein (GNT) modulates P-gp expression in hepatocellular carcinoma in vitro. Although several beneficial effects (e.g., antioxidant, antimutagenic, anticancer) have been attributed to GNT, the molecular mechanisms have not been totally elucidated. In the present work, we evaluated the effect of GNT on P-gp expression in rat liver, kidney and ileum. We found that GNT (5 mg/kg daily s.c. 3 days) increased hepatic P-gp expression and also Mdr1a (one of the genes encoding P-gp) mRNA levels. Renal and intestinal P-gp remained unchanged after GNT treatment. Hepatic P-gp activity measured with rhodamine-123 and digoxin, both well-known P-gp substrates, was also increased. In vitro experiments using hepatocyte primary cell culture demonstrated that inhibition of ER-α with ICI182/780 did not prevent Mdr1a mRNA up-regulation by GNT (10 µM). In contrast, Mdr1a induction was suppressed after pregnane X receptor (PXR) inhibition by sulforaphane and knockdown of this nuclear receptor. These findings were confirmed in vivo by using the PXR antagonist ketoconazole. In conclusion, we demonstrated the induction of hepatic P-gp expression and activity by GNT in vivo, with PXR being a likely mediator. This suggests that GNT, at concentrations observed in plasma of individuals consuming the phytoestrogen in the diet or through supplements, could affect the clearance of relevant P-gp substrates of therapeutic use as well as toxicity of environmental and food toxicants.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Anticarcinógenos/toxicidade , Genisteína/toxicidade , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Carcinoma Hepatocelular/metabolismo , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , RNA Mensageiro/metabolismo , Ratos
12.
Int J Mol Sci ; 21(16)2020 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-32824312

RESUMO

Soybeans are one of the most used alternative dietary ingredients in aquafeeds. However, they contain phytoestrogens like genistein (GE), which can have an impact on fish metabolism and health. This study aimed to investigate the in vitro and in vivo effects of GE on lipid metabolism, apoptosis, and autophagy in rainbow trout (Oncorhynchus mykiss). Primary cultured preadipocytes were incubated with GE at different concentrations, 10 or 100 µM, and 1 µM 17ß-estradiol (E2). Furthermore, juveniles received an intraperitoneal injection of GE at 5 or 50 µg/g body weight, or E2 at 5 µg/g. In vitro, GE 100 µM increased lipid accumulation and reduced cell viability, apparently involving an autophagic process, indicated by the higher LC3-II protein levels, and higher lc3b and cathepsin d transcript levels achieved after GE 10 µM. In vivo, GE 50 µg/g upregulated the gene expression of fatty acid synthase (fas) and glyceraldehyde-3-phosphate dehydrogenase in adipose tissue, suggesting enhanced lipogenesis, whereas it increased hormone-sensitive lipase in liver, indicating a lipolytic response. Besides, autophagy-related genes increased in the tissues analyzed mainly after GE 50 µg/g treatment. Overall, these findings suggest that an elevated GE administration could lead to impaired adipocyte viability and lipid metabolism dysregulation in rainbow trout.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia , Autofagia , Genisteína/farmacologia , Fitoestrógenos/farmacologia , Truta/metabolismo , Adipócitos/metabolismo , Animais , Catepsina D/genética , Catepsina D/metabolismo , Sobrevivência Celular , Células Cultivadas , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Genisteína/toxicidade , Gliceraldeído-3-Fosfato Desidrogenases/genética , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Metabolismo dos Lipídeos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Fitoestrógenos/toxicidade
13.
Toxicol Appl Pharmacol ; 367: 12-22, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30684530

RESUMO

Catechol-O-methyltransferase (COMT) acts as a 'gate-keeper' to prevent DNA damage during estrogen metabolism. Both experimental and epidemiological studies suggest the role of COMT in pathogenesis of human breast cancer (BCa). It was previously reported that inhibition of COMT enzyme activity in estradiol-treated human breast epithelial carcinoma-derived MCF-7 cells caused increased oxidative DNA damage and formation of mutagenic depurinating adducts. To improve our understanding of factors influencing estrogen metabolism in BCa, it requires a mechanistic study illustrating the regulation of this 'gate-keeper'. We investigated the epigenetic mechanisms underlying decreased COMT transcription in MCF-7 cells exposed to 17ß-estradiol (E2) and the phytoestrogen, genistein (GEN). CpG site-specific methylation at promoters for both soluble (S) and membrane-bound (MB) COMT transcripts were assessed. Both E2 and GEN induced CpG site-specific methylation within the distal promoter of MB-COMT. In addition, ChIP analysis showed that there was increased binding of DNMT3B, MBD2 and HDAC1 within this promoter. These epigenetic changes were associated with decreased COMT transcript levels. Interestingly, sulforaphane, an antioxidant commonly found in cruciferous vegetables, was able to reverse the estrogen-induced epigenetic changes and gene silencing of COMT. Our data provide a new insight in epigenetically targeting COMT transcription. Since reactive estrogen metabolites may contribute to breast cancer, our findings may help in developing prevention and/or intervention strategies for human BCa.


Assuntos
Neoplasias da Mama/genética , Catecol O-Metiltransferase/genética , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Estradiol/toxicidade , Genisteína/toxicidade , Regiões Promotoras Genéticas , Transcrição Gênica/efeitos dos fármacos , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Catecol O-Metiltransferase/metabolismo , Ilhas de CpG , Regulação para Baixo , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7
14.
J Reprod Dev ; 65(1): 7-17, 2019 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-30333376

RESUMO

Soy-based formula contains high concentrations of the isoflavone genistein. Genistein possesses estrogenic and tyrosine kinase inhibitory activity and interferes with cellular proliferation and development. To date, the acute and chronic effects of genistein on ovarian and uterine development have not been fully elucidated. In this study, mice at postnatal day 1 were subcutaneously injected with 100 mg/kg genistein for 10 consecutive days, and then their ovaries and uteri were collected on days 10, 21, and 90. Histological evaluation was performed after hematoxylin and eosin staining. The proliferating activity was indicated by the proliferating indicator protein Ki67. Results showed that the subcutaneous injection of genistein to neonatal mice induced the formation of multi-oocyte follicles and delayed the primordial follicle assembly in the ovaries. Genistein significantly enlarged the cross-sectional area of the uterine cavity and wall and disrupted the regularity between the uterine stroma and myometrium. Genistein exposure inhibited proliferative activity because fewer Ki67-positive nuclei were detected in ovarian and uterine cell populations than in the control. Furthermore, most ovaries from adult mice given neonatal genistein were without corpora lutea, and there appeared to be cystic follicles and hypertrophy of the theca, and cortical and medullary layers. Considering the high concentration of isoflavone in soy-based infant formulas and livestock feed, we suggest that the use of isoflavone-rich diets in humans and livestock receive closer examination.


Assuntos
Animais Recém-Nascidos , Proliferação de Células/efeitos dos fármacos , Genisteína/toxicidade , Ovário/efeitos dos fármacos , Ovário/crescimento & desenvolvimento , Útero/efeitos dos fármacos , Útero/crescimento & desenvolvimento , Animais , Feminino , Genisteína/administração & dosagem , Genisteína/análise , Antígeno Ki-67/análise , Camundongos , Ovário/citologia , Alimentos de Soja/análise , Útero/citologia
15.
Pharm Biol ; 57(1): 226-230, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30946631

RESUMO

CONTEXT: We identified an active prenylated derivative of genistein, 8-prenylgenistein (8PG) from Erythrina variegata L. (Leguminosae) and found that 8PG increased osteoprotective effects of genistein in oestrogen-deficient mice. OBJECTIVE: This study investigated and compared the oestrogenic effects of genistein and 8PG on uterus and vagina of immature mice. MATERIALS AND METHODS: Immature female CD-1 mice were orally treated with vehicle (Control, n = 10) or genistein (75 mg/kg, n = 10) or 8PG with low (8PG-L, 75 mg/kg, n = 10) and high dose (8PG-H, 150 mg/kg, n = 10) for 7 consecutive days by intragastric gavage. The uterus and vagina were harvested for histological and molecular measurements. RESULTS: Treatment with genistein and 8PG-H significantly increased uterus index (1.98 ± 0.21 & 1.49 ± 0.16 mg/g) and vagina index (3.83 ± 0.11 & 3.13 ± 0.25 mg/g) as compared to untreated control (uterus, 1.12 ± 0.13 mg/g; vagina, 2.32 ± 0.18 mg/g). Accordingly, both genistein and 8PG-H made vaginal cells keratinized and induced uterine and vaginal hypertrophy associated with the endometrial proliferation. 8PG-L did not affect oestrus cycle and histology of uterus and vagina. Treatment of immature mice with genistein or 8PG-H upregulated protein expression of oestrogen receptor-α (ER-α) and proliferating cell nuclear antigen (PCNA), but 8PG-L did not alter ER-α and PCNA expression in uterus and vagina. CONCLUSION: This study indicated that 8-prenylgenistein exerted oestrogenic effects in immature female mice. The efficacy and safety of 8-prenylgenistein when applied in improving oestrogen deficiency-induced syndrome requires further elucidation.


Assuntos
Estrogênios/farmacologia , Genisteína/análogos & derivados , Genisteína/farmacocinética , Útero/efeitos dos fármacos , Vagina/efeitos dos fármacos , Administração Oral , Animais , Relação Dose-Resposta a Droga , Receptor alfa de Estrogênio/metabolismo , Estrogênios/administração & dosagem , Estrogênios/toxicidade , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genisteína/administração & dosagem , Genisteína/farmacologia , Genisteína/toxicidade , Camundongos , Regulação para Cima/efeitos dos fármacos , Útero/metabolismo , Vagina/metabolismo
16.
Toxicol Pathol ; 46(5): 574-596, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29895210

RESUMO

Developmental exposure to estrogenic chemicals is an established risk factor for cancer of the female reproductive tract. This increase in risk has been associated with disruption of normal patterns of cellular differentiation during critical stages of morphogenesis. The goal of this study was to document uterine epithelial phenotypes over time following neonatal treatment with the synthetic estrogen diethylstilbestrol (DES) or the soy phytoestrogen genistein (GEN) in female CD-1 mice. Both DES and GEN induced three distinct populations of abnormal endometrial epithelial cells: luminal (SIX1+/P63-/CK14-/CK18+), basal (SIX1+/P63+/CK14+/CK18-), and mixed/bipotential (SIX1+/P63-/CK14+/CK18+), which were all established by early adulthood. In older animals, DES and GEN resulted in uterine carcinomas with mixed glandular, basal, and squamous cell elements. All carcinomas were composed largely of the three abnormal cell types. These findings identify novel epithelial differentiation patterns in the uterus and support the idea that disruption of cellular programming in early development can influence cancer risk later in life.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Neoplasias do Endométrio/induzido quimicamente , Endométrio/efeitos dos fármacos , Estrogênios/toxicidade , Morfogênese/efeitos dos fármacos , Lesões Pré-Cancerosas/induzido quimicamente , Animais , Animais Recém-Nascidos , Dietilestilbestrol/toxicidade , Neoplasias do Endométrio/patologia , Endométrio/crescimento & desenvolvimento , Endométrio/patologia , Feminino , Genisteína/toxicidade , Imuno-Histoquímica , Camundongos Endogâmicos , Lesões Pré-Cancerosas/patologia
17.
Can J Physiol Pharmacol ; 95(10): 1298-1305, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28732172

RESUMO

Our previous work showed the presence of endothelin-1 (ET-1) receptors, ETA and ETB, in human vascular endothelial cells (hVECs). In this study, we wanted to verify whether ET-1 plays a role in the survival of hVECs via the activation of its receptors ETA and (or) ETB (ETAR and ETBR, respectively). Our results showed that treatment of hVECs with ET-1 prevented apoptosis induced by genistein, an effect that was mimicked by treatment with ETBR-specific agonist IRL1620. Furthermore, blockade of ETBR with the selective ETBR antagonist A-192621 prevented the anti-apoptotic effect of ET-1 in hVECs. However, activation of ETA receptor alone did not seem to contribute to the anti-apoptotic effect of ET-1. In addition, the anti-apoptotic effect of ETBR was found to be associated with caspase 3 inhibition and does not depend on the density of this type of receptor. In conclusion, our results showed that ET-1 possesses an anti-apoptotic effect in hVECs and that this effect is mediated, to a great extent, via the activation of ETBR. This study revealed a new role for ETBR in the survival of hVECs.


Assuntos
Apoptose/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Endotelina-1/farmacologia , Receptor de Endotelina A/agonistas , Receptor de Endotelina B/agonistas , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Antagonistas do Receptor de Endotelina A/farmacologia , Antagonistas do Receptor de Endotelina B/farmacologia , Genisteína/toxicidade , Humanos , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Transdução de Sinais/efeitos dos fármacos
18.
Environ Toxicol ; 32(3): 832-844, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27235753

RESUMO

We hypothesized that genistein can interfere with the regulation of uterine fluid volume, secretion rate and expression of aquaporin in the uterus by female sex-steroids, i.e., estrogen and progesterone. Therefore, the aims of this study were to investigate changes in these parameters in the presence of genistein and female sex-steroids. METHODS: Female Sprague-Dawley rats were ovariectomized and received 3-days estradiol-17ß benzoate (E2) plus genistein (25, 50, or 100 mg kg-1  day-1 ) or 3-days E2 followed by 3-days E2 plus progesterone with genistein (25, 50, or 100 mg kg-1  day-1 ). A day after last treatment, uterine fluid secretion rate was determined by in vivo uterine perfusion with rats under anesthesia. Animals were sacrificed and uteri were harvested and subjected for histological analyses. Luminal/outer uterine circumference was determined and distribution of AQP-1, 2, 5, and 7 in endometrium was visualized by immunofluorescence. Expression of AQP-1, 2, 5, and 7 proteins and mRNAs were determined by Western blotting and Real-time PCR respectively. RESULTS: Combined treatment of E2 with high dose genistein (50 and 100 mg kg-1  day-1 ) resulted in significant decrease in uterine fluid volume, secretion rate and expression of AQP-1, 2, 5, and 7 proteins and mRNAs in uterus (p < 0.05). No significant changes in these parameters were observed when 25 mg kg-1  day-1 genistein was given with E2 or when genistein was given with E2 followed by E2 plus progesterone Conclusions: Decreased in uterine fluid volume, secretion rate and AQP-1, 2, 5, and 7 expression in the uterus by high dose genistein in the presence of E2 could potentially affect female fertility. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 832-844, 2017.


Assuntos
Aquaporinas/metabolismo , Estradiol/farmacologia , Genisteína/toxicidade , Progesterona/farmacologia , Útero/efeitos dos fármacos , Animais , Aquaporina 1/genética , Aquaporina 1/metabolismo , Aquaporina 2/genética , Aquaporina 2/metabolismo , Aquaporina 5/genética , Aquaporina 5/metabolismo , Aquaporinas/genética , Western Blotting , Endométrio/metabolismo , Endométrio/patologia , Feminino , Microscopia de Fluorescência , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Útero/metabolismo , Útero/patologia
19.
Toxicol Appl Pharmacol ; 293: 53-62, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26792615

RESUMO

Genistein is a naturally occurring isoflavone phytoestrogen commonly found in plant products such as soybeans, lentils, and chickpeas. Genistein, like other phytoestrogens, has the potential to mimic, enhance, or impair the estradiol biosynthesis pathway, thereby potentially altering ovarian follicle growth. Previous studies have inconsistently indicated that genistein exposure may alter granulosa cell proliferation and hormone production, but no studies have examined the effects of genistein on intact antral follicles. Thus, this study was designed to test the hypothesis that genistein exposure inhibits follicle growth and steroidogenesis in intact antral follicles. To test this hypothesis, antral follicles isolated from CD-1 mice were cultured with vehicle (dimethyl sulfoxide; DMSO) or genistein (6.0 and 36µM) for 18-96h. Every 24h, follicle diameters were measured to assess growth. At the end of each culture period, the media were pooled to measure hormone levels, and the cultured follicles were collected to measure expression of cell cycle regulators and steroidogenic enzymes. The results indicate that genistein (36µM) inhibits growth of mouse antral follicles. Additionally, genistein (6.0 and 36µM) increases progesterone, testosterone, and dehydroepiandrosterone (DHEA) levels, but decreases estrone and estradiol levels. The results also indicate that genistein alters the expression of steroidogenic enzymes at 24, 72 and 96h, and the expression of cell cycle regulators at 18h. These data indicate that genistein exposure inhibits antral follicle growth by inhibiting the cell cycle, alters sex steroid hormone levels, and dysregulates steroidogenic enzymes in cultured mouse antral follicles.


Assuntos
Genisteína/toxicidade , Folículo Ovariano/efeitos dos fármacos , Fitoestrógenos/toxicidade , 17-Hidroxiesteroide Desidrogenases/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas de Ciclo Celular/genética , Sistema Enzimático do Citocromo P-450/genética , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Feminino , Hormônios Esteroides Gonadais/metabolismo , Camundongos , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Fosfoproteínas/genética
20.
Proc Natl Acad Sci U S A ; 110(2): 537-42, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23267115

RESUMO

Reports that maternal diet influences coat color in mouse offspring carrying the agouti A(vy) allele have received considerable attention because the range, from pseudoagouti (brown) to yellow, predicts adult health outcomes, especially disposition toward obesity and diabetes, in yellower mice. Bisphenol A (BPA), an endocrine-disrupting compound with estrogenic properties, fed to a/a dams harboring A(vy)/a conceptuses has been reported to induce a significant shift toward yellower mice, whereas consumption of either genistein (G) alone or in combination with BPA led to greater numbers of healthy, brown offspring. Groups of C57/B6 a/a females, which are nonagouti, were fed either a phytoestrogen-free control diet or one of six experimental diets: diets 1-3 contained BPA (50 mg, 5 mg, and 50 µg BPA/kg food, respectively); diet 4 contained G (250 mg/kg food); diet 5 contained G plus BPA (250 and 50 mg/kg food, respectively); and diet 6 contained 0.1 µg of ethinyl estradiol (EE)/kg food. Mice were bred to A(vy)/a males over multiple parities. In all, 2,824 pups from 426 litters were born. None of the diets provided any significant differences in relative numbers of brown, yellow, or intermediate coat color A(vy)/a offspring. However, BPA plus G (P < 0.0001) and EE diets (P = 0.005), but not the four others, decreased the percentage of black (a/a) to A(vy)/a offspring from the expected Mendelian ratio of 1:1. Data suggest that A(vy)/a conceptuses, which may possess a so-called "thrifty genotype," are at a competitive advantage over a/a conceptuses in certain uterine environments.


Assuntos
Compostos Benzidrílicos/toxicidade , Disruptores Endócrinos/toxicidade , Genisteína/toxicidade , Cor de Cabelo/efeitos dos fármacos , Cor de Cabelo/genética , Exposição Materna , Fenóis/toxicidade , Efeitos Tardios da Exposição Pré-Natal/genética , Animais , Cruzamentos Genéticos , Feminino , Modelos Lineares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Gravidez , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA