Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biochem Pharmacol ; 210: 115490, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36893816

RESUMO

Maternal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) causes developmental and reproductive disorders in pups due to the attenuated luteinizing hormone (LH) production during the perinatal stage; however, the administration of α-lipoic acid (LA) to TCDD-exposed pregnant rats reversed the attenuated LH production. Therefore, reproductive disorders in pups are expected to be ameliorated with LA supplementation. To address this issue, pregnant rats orally received low dose TCDD at gestational day 15 (GD15) and proceeded to parturition. The control received a corn oil vehicle. To examine the preventive effects of LA, supplementation with LA was provided until postnatal day 21. In this study, we demonstrated that maternal administration of LA restored the sexually dimorphic behavior of male and female offspring. TCDD-induced LA insufficiency is likely a direct cause of TCDD reproductive toxicity. In the analysis to clarify the mechanism of the decrease in LA, we found evidence suggesting that TCDD inhibits the synthesis and increases the utilization of S-adenosylmethionine (SAM), a cofactor for LA synthesis, resulting in a decrease in the SAM level. Furthermore, folate metabolism, which is involved in SAM synthesis, is disrupted by TCDD, which may adversely affect infant growth. Maternal supplementation of LA restored SAM to its original level in the fetal hypothalamus; in turn, SAM ameliorated abnormal folate consumption and suppressed aryl hydrocarbon receptor activation induced by TCDD. The study demonstrates that the application of LA could prevent and recover next-generation dioxin reproductive toxicity, which provides the potential to establish effective protective measures against dioxin toxicity.


Assuntos
Ácido Fólico , Exposição Materna , Dibenzodioxinas Policloradas , Efeitos Tardios da Exposição Pré-Natal , Caracteres Sexuais , Desenvolvimento Sexual , Ácido Tióctico , Animais , Feminino , Masculino , Gravidez , Ratos , Feto/efeitos dos fármacos , Feto/metabolismo , Ácido Fólico/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Exposição Materna/efeitos adversos , Dibenzodioxinas Policloradas/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , S-Adenosilmetionina/metabolismo , Desenvolvimento Sexual/efeitos dos fármacos , Ácido Tióctico/administração & dosagem , Ácido Tióctico/farmacologia , Ácido Tióctico/uso terapêutico , Reprodução/efeitos dos fármacos
2.
Andrology ; 8(6): 1895-1906, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32598553

RESUMO

BACKGROUND: Recent studies have shown that epigenetic alterations, such as those involving lysine-specific demethylase 1 (LSD1), lead to oncogenic activation and highlight such alterations as therapeutic targets. However, studies evaluating the effect of LSD1 inhibitors on male fertility are lacking. OBJECTIVES: We analyzed the potential toxicity of a new selective LSD1 inhibitor, N-[(1S)-3-[3-(trans-2-aminocyclopropyl)phenoxy]-1-(benzylcarbamoyl)propyl] benzamide (NCL1), in testes. MATERIALS AND METHODS: Human testicular samples were immunohistochemically analyzed. Six-week-old male C57BL/6J mice were injected intraperitoneally with dimethyl sulfoxide vehicle (n = 15), or 1.0 (n = 15) or 3.0 (n = 15) mg/kg NCL1 biweekly. After five weeks, toxicity and gene expression were analyzed in testicular samples by ingenuity pathway analysis (IPA) using RNA sequence data and quantitative reverse transcriptase (qRT)-PCR; hormonal damage was analyzed in blood samples. NCL1 treated GC-1, TM3, and TM4 cell lines were analyzed by cell viability, chromatin immunoprecipitation, flow cytometry, and Western blot assays. RESULTS: LSD1 was mainly expressed in human Sertoli and germ cells, with LSD1 levels significantly decreased in a progressive meiosis-dependent manner; germ cells showed similar expression patterns in normal spermatogenesis and early/late maturation arrest. Histological examination revealed significantly increased levels of abnormal seminiferous tubules in 3.0 mg/kg NCL1-treated mice compared to control, with increased cellular detachment, sloughing, vacuolization, eosinophilic changes, and TUNEL-positive cells. IPA and qRT-PCR revealed NCL1 treatment down-regulated LSD1 activity. NCL1 also reduced total serum testosterone levels. Western blots of mouse testicular samples revealed NCL1 induced a marked elevation in cleaved caspases 3, 7, and 8, and connexin 43 proteins. NCL1 treatment significantly reduced GC-1, but not TM3 and TM4, cell viability in a dose-dependent manner. In flow cytometry analysis, NCL1 induced apoptosis in GC-1 cells. CONCLUSIONS: High-dose NCL1 treatment targeting LSD1 caused dysfunctional spermatogenesis and induced caspase-dependent apoptosis. This suggests the LSD1 inhibitor may cause testicular toxicity via the regulation of apoptosis.


Assuntos
Antineoplásicos/efeitos adversos , Apoptose/efeitos dos fármacos , Infertilidade Masculina/induzido quimicamente , Espermatogênese/efeitos dos fármacos , Testículo/patologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Neoplasias Hematológicas/tratamento farmacológico , Histona Desmetilases/antagonistas & inibidores , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias da Próstata/tratamento farmacológico , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Testículo/efeitos dos fármacos , Testosterona/sangue
3.
Yakugaku Zasshi ; 137(11): 1373-1379, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-29093374

RESUMO

Dioxins, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are responsible for producing serious toxic effects in the next generation, such as sexual immaturity. Our laboratory found that treating pregnant rats on gestational day 15 with TCDD (1 µg/kg orally) targets pituitary luteinizing hormone (LH) to attenuate testicular steroidogenesis in fetuses. Because sex steroids during a short window ("the critical period") in the perinatal stage stimulate brain differentiation closely linked to sexual maturation, it is likely that TCDD imprints sexual immaturity on the offspring due to the lowered expression of LH during the fetal period. To address this hypothesis, we first investigated the effect of supplementation of equine chorionic gonadotropin (eCG), an LH-mimicking hormone, in fetuses exposed to TCDD. The result showed that eCG ameliorated defects in sexual behavior in adulthood as well as in steroidogenesis during the fetal stage. We also found that maternal exposure to TCDD induced the pituitary expression of histone deacetylases (HDACs) in fetuses. In agreement with this, TCDD deacetylated the histones wrapped around the LHß gene, and valproic acid, an HDAC inhibitor, blocked the reduced level of LHß caused by TCDD. These observations strongly suggest that TCDD induces the expression of HDACs to attenuate fetal LH production. Finally, such a transient reduction in steroidogenesis of the pituitary-gonadal axis causes a decrease in the expression of hypothalamic gonadotropin-releasing hormone, resulting in defects in sexual behavior in adulthood. This review increases our understanding of the developmental toxicities caused by endocrine disruptors including dioxins.


Assuntos
Dioxinas/efeitos adversos , Disruptores Endócrinos/efeitos adversos , Exposição Materna/efeitos adversos , Troca Materno-Fetal , Efeitos Tardios da Exposição Pré-Natal , Maturidade Sexual/efeitos dos fármacos , Maturidade Sexual/genética , Animais , Dioxinas/toxicidade , Disruptores Endócrinos/toxicidade , Feminino , Idade Gestacional , Hormônios Esteroides Gonadais/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Histona Desacetilases/metabolismo , Humanos , Hipotálamo/metabolismo , Recém-Nascido , Hormônio Luteinizante/metabolismo , Masculino , Hipófise/metabolismo , Gravidez , Ratos , Testículo/metabolismo
4.
J Toxicol Sci ; 42(1): 13-23, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28070105

RESUMO

Our previous studies demonstrated that treating pregnant rats with dioxins, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), targets the pituitary expression of luteinizing hormone (LH) to attenuate testicular steroidogenesis in fetuses, resulting in the imprinting of sexual immaturity of the offspring after reaching maturity. Furthermore, we found that although TCDD disturbs the tricarboxylic acid (TCA) cycle in the fetal hypothalamus, maternal co-treatment with α-lipoic acid (α-LA), a cofactor of the TCA cycle, restores a TCDD-produced reduction in the LH-evoked steroidogenesis as well as the TCA cycle activity in fetuses. However, the mechanism underlying the beneficial effect of α-LA remains to be fully elucidated. To address this issue, we compared the effect of α-LA with that of thiamine, another cofactor of the TCA cycle. As with α-LA, supplying thiamine to dams exposed to TCDD alleviates the reduced level of not only hypothalamic ATP but also pituitary LH and testicular steroidogenic protein in fetuses. However, thiamine had a much weaker effect than α-LA. In agreement with ATP attenuation, TCDD activated AMP-activated protein kinase (AMPK), a negative regulator of LH production, whereas the supplementation of α-LA allowed recovery from this defect. Furthermore, α-LA restored the TCDD-produced reduction in the pituitary expression of the receptor for gonadotropin-releasing hormone (GnRH), an upstream regulator of LH synthesis. These results suggest that α-LA rescues TCDD-produced attenuation during fetal steroidogenesis due not only to facilitation of energy production through the TCA cycle but also through suppression of AMPK activation, and the pituitary GnRH receptor may serve as a mediator of these effects.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Encéfalo/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Receptores LHRH/genética , Ácido Tióctico/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Encéfalo/metabolismo , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Hormônio Liberador de Gonadotropina/genética , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Hormônio Luteinizante/sangue , Hormônio Luteinizante/genética , Masculino , Miocárdio/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos Wistar , Testículo/efeitos dos fármacos , Testículo/metabolismo
5.
Mol Pharmacol ; 85(1): 74-82, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24132183

RESUMO

Our previous studies have shown that treatment of pregnant rats with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 1 µg/kg) at gestational day (GD) 15 reduces the pituitary synthesis of luteinizing hormone (LH) during the late fetal and early postnatal period, leading to the imprinting of defects in sexual behaviors at adulthood. However, it remains unclear how the attenuation of pituitary LH is linked to sexual immaturity. To address this issue, we performed a DNA microarray analysis to identify the gene(s) responsible for dioxin-induced sexual immaturity on the pituitary and hypothalamus of male pups, born of TCDD-treated dams, at the age of postnatal day (PND) 70. Among the reduced genes, we focused on gonadotropin-releasing hormone (GnRH) in the hypothalamus because of published evidence that it has a role in sexual behaviors. An attenuation by TCDD of GnRH expression emerged at PND4, and no subsequent return to the control level was seen. A change in neither DNA methylation nor histone acetylation accounted for the reduced expression of GnRH. Intracerebroventricular infusion of GnRH to the TCDD-exposed pups after reaching maturity restored the impairment of sexual behaviors. Supplying equine chorionic gonadotropin, an LH-mimicking hormone, to the TCDD-exposed fetuses at GD15 resulted in a recovery from the reduced expression of GnRH, as well as from the defects in sexual behavior. These results strongly suggest that maternal exposure to TCDD fixes the status of the lowered expression of GnRH in the offspring by reducing the LH-assisted steroidogenesis at the perinatal stage, and this mechanism imprints defects in sexual behaviors at adulthood.


Assuntos
Poluentes Ambientais/toxicidade , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Exposição Materna/efeitos adversos , Dibenzodioxinas Policloradas/toxicidade , Efeitos Tardios da Exposição Pré-Natal/psicologia , Comportamento Sexual Animal , Animais , Animais Recém-Nascidos , Gonadotropina Coriônica/uso terapêutico , Metilação de DNA , Embrião de Mamíferos , Feminino , Impressão Genômica , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/uso terapêutico , Cavalos , Masculino , Troca Materno-Fetal , Hipófise/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/etiologia , Ratos , Ratos Wistar , Comportamento Sexual Animal/efeitos dos fármacos , Testículo/metabolismo , Fatores de Tempo
6.
J Toxicol Sci ; 35(3): 365-73, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20519845

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) reduces the synthesis of pituitary gonadotropins in a fetal age-specific manner. The pituitary synthesis of gonadotropins is regulated by the hypothalamus and, thus, needs the differentiation and development of the hypothalamus requiring a number of factors including energy supply and neurotransmitters. To investigate the mechanism whereby TCDD reduces fetal gonadotropins, we carried out a comparative study on the metabolomes of the hypothalamus and pituitary using fetal and mature Wistar rats. Male fetuses at gestational day (GD)20 were removed from dams treated orally with TCDD (1 microg/kg) at GD15, and the metabolome profiles were analyzed by gas chromatography-mass spectrometry (GC-MS). The principal component analysis of GC-MS data revealed that TCDD caused a change in the profile of fetal metabolome more markedly in the hypothalamus than in the pituitary. In sharp contrast, TCDD did not cause any marked alteration in hypothalamic as well as pituitary metabolomes in male rats born of untreated dams and treated with TCDD at postnatal day 49. It was also demonstrated that a number of fetal hypothalamic components, including glutamine and gamma-aminobutyric acid, are reduced by TCDD. These results demonstrate a possibility that TCDD may reduce the metabolic activity of the hypothalamus in a fetus-specific fashion, resulting in the reduced synthesis of gonadotropins.


Assuntos
Gonadotropinas/biossíntese , Hipotálamo/metabolismo , Exposição Materna/efeitos adversos , Troca Materno-Fetal , Metaboloma/efeitos dos fármacos , Dibenzodioxinas Policloradas/efeitos adversos , Animais , Depressão Química , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Glutamina/metabolismo , Masculino , Hipófise/metabolismo , Gravidez , Ratos , Ratos Wistar , Ácido gama-Aminobutírico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA