Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gut Microbes ; 16(1): 2359729, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38816999

RESUMO

Early life environment influences mammalian brain development, a growing area of research within the Developmental Origins of Health and Disease framework, necessitating a deeper understanding of early life factors on children's brain development. This study introduces a mouse model, LAO1 knockout mice, to investigate the relationship between breast milk, the gut microbiome, and brain development. The results reveal that breast milk's reactive oxygen species (ROS) are vital in shaping the neonatal gut microbiota. Decreased hydrogen peroxide (H2O2) levels in milk disrupt the gut microbiome and lead to abnormal metabolite production, including D-glucaric acid. This metabolite inhibits hippocampal myelin formation during infancy, potentially contributing to behavioral abnormalities observed in adulthood. These findings suggest that H2O2 in breast milk is crucial for normal gut microbiota formation and brain development, with implications for understanding and potentially treating neurodevelopmental disorders in humans.


Assuntos
Animais Recém-Nascidos , Microbioma Gastrointestinal , Peróxido de Hidrogênio , Camundongos Knockout , Leite Humano , Bainha de Mielina , Animais , Feminino , Masculino , Camundongos , Animais Recém-Nascidos/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/crescimento & desenvolvimento , Hipocampo/metabolismo , Hipocampo/crescimento & desenvolvimento , Peróxido de Hidrogênio/metabolismo , Camundongos Endogâmicos C57BL , Leite Humano/química , Leite Humano/metabolismo , Bainha de Mielina/metabolismo , Espécies Reativas de Oxigênio/metabolismo
2.
Neurosci Res ; 168: 76-82, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31022413

RESUMO

Depression and anxiety, which are severe symptoms during menopause, are caused by ceased ovarian activity and declined serum progesterone levels. Studies have demonstrated that gut microbiota can regulate brain function and change the microbiota composition during the perimenopause period. This study investigated whether progesterone affects depressant and anxious behaviors via gut microbiota. In ovariectomized (OVX) mice, treatment with progesterone improved depressive and anxious behaviors, and gut microbiota composition was significantly changed. In particular, increased Lactobacillus spp. were observed in these mice. Reduction of microbiota by antibiotic treatment abolished the effect of progesterone on depression and anxiety. In addition, administration of Lactobacillus (L.) reuteri that was increased by progesterone also reduced the depressant behavior in OVX mice, and BDNF gene expression was elevated by progesterone treatment and L. reuteri administration in the hippocampus. Moreover, we found that progesterone stimulated the growth of L. reuteri in vitro. In summary, our findings indicate that progesterone reduces depression and anxiety through changes in gut microbiota composition, particularly by increasing the Lactobacillus spp. population.


Assuntos
Microbioma Gastrointestinal , Animais , Ansiedade , Depressão , Feminino , Lactobacillus , Camundongos , Camundongos Endogâmicos C57BL , Progesterona
3.
Poult Sci ; 99(9): 4586-4597, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32868003

RESUMO

Reproductive failure associated with heat stress is a well-known phenomenon in poultry. High temperatures also induce various metabolic disturbances in many animals. Because the liver plays a central role in metabolism, the present study aimed to clarify the relationship between liver and reproduction in Japanese quails exposed to high temperatures. In the consecutive 20-D experimental period, quails were treated with 25°C (control) or 34°C (heat) from 12:00 to 16:00. Eggs were collected for hatching. On completion of the experimental period, quails were humanely euthanized for hormone analyses (e.g., serum and ovarian follicles). Serum metabolites were analyzed using GC/MS. Liver and ovary samples were collected for mRNA levels, histomorphology, and metabolic analysis. Ovary and oviduct weights significantly decreased after daily heat exposure. The number and weight of hierarchical follicles also decreased. Consequently, egg weight decreased. Although there was no difference in fertilization rate, chick birth weight significantly decreased in the heated group. Corticosterone and 17ß-estradiol in the serum significantly increased in the heated group. Yolk corticosterone and 17ß-estradiol concentration and content were higher in the heated group. Ovary sterologenic enzymes gene P450scc and estrogen receptor expression level increased. The FSH receptor decreased in heat-stressed quails. MetaboAnalyst analysis indicated that high temperature affects propanoate metabolism, beta-alanine metabolism, aspartate metabolism, and histidine metabolism. Triglyceride and cholesterol levels in the liver increased in the heated group. The heated group also had an increased mRNA expression of AGPAT5, apoptosis gene caspase3, and the immunocytokine genes IL-6 and TLR4. However, NF-κB gene expression decreased. These results suggest that high temperatures affect lipid metabolism and apoptosis and cause inflammation in the liver. High temperature induced ovarian dysfunction, which resulted in the decline of hierarchical follicle number and weight, egg weight, and chick birth weight. The increased level of 17ß-estradiol suggests liver damage. Protecting liver function from damage may assist quails cope in summer.


Assuntos
Coturnix , Temperatura Alta , Fígado , Reprodução , Animais , Coturnix/fisiologia , Exposição Ambiental , Feminino , Fígado/patologia
4.
FASEB J ; 33(3): 3343-3352, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30433825

RESUMO

The development of gut microbiota during infancy is an important event that affects the health status of the host; however, the mechanism governing it is not fully understood. l-Amino acid oxidase 1 (LAO1) is a flavoprotein that catalyzes the oxidative deamination of particular l-amino acids and converts them into keto acids, ammonia, and H2O2. Our previous study showed that LAO1 is present in mouse milk and exerts protection against bacteria by its production of H2O2. The data led us to consider whether LAO1, H2O2, or both could impact infant gut microbiota development via mother's milk consumption in mice. Different gut microbiota profiles were observed in the wild-type (WT) and LAO1-knockout mouse pups. The WT pups' microbiota was relatively simple and composed of only a few dominant bacteria, such as Lactobacillus, whereas the lactating knockout pups had high microbiota diversity. Cross-fostering experiments indicated that WT milk (containing LAO1) has the ability to suppress the diversity of microbiota in pups. We observed that the stomach content of pups fed WT milk had LAO1 proteins and the ability to produce H2O2. Moreover, culture experiments showed that Lactobacillus was abundant in the feces of pups fed WT milk and that Lactobacillus was more resistant to H2O2 than Bifidobacterium and Escherichia. Human breast milk produces very little H2O2, which could be the reason for Lactobacillus not being dominant in the feces of breast-fed human infants. In mouse mother's milk, H2O2 is generated from the process of free amino acid metabolism, and H2O2 may be a key player in regulating the initial acquisition and development of gut microbiota, especially growth of Lactobacillus, during infancy.-Shigeno, Y., Zhang, H., Banno, T., Usuda, K., Nochi, T., Inoue, R., Watanabe, G., Jin, W., Benno, Y., Nagaoka, K. Gut microbiota development in mice is affected by hydrogen peroxide produced from amino acid metabolism during lactation.


Assuntos
Aminoácidos/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Lactação/efeitos dos fármacos , Microbiota/efeitos dos fármacos , Animais , Bifidobacterium/efeitos dos fármacos , Fezes/microbiologia , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Lactação/metabolismo , Lactobacillus/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Leite/microbiologia , Probióticos/administração & dosagem
5.
J Vet Med Sci ; 81(1): 77-83, 2019 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-30464091

RESUMO

High temperature induces various metabolic disturbances in animals. However, no comprehensive information is currently available on the metabolic pathway affected by high environmental temperature. The present study examined metabolite content in the serum of heat challenged quails using metabolomic analysis. In the present study, female quails with normal laying rate at 20 weeks kept in standard condition (control group) or exposed to 34°C 4 hr per day (12:00 to 16:00 hr)(heat group) for 10 consecutive days. The metabolomic analysis identified 165 metabolites in the serum, and significant differences were observed in the serum for 7 metabolites between two groups. An analysis by MetaboAnalyst, a web-based metabolome data tool, indicate that high temperature affect ketone body metabolism, butyrate metabolism, arginine and proline metabolism. Furthermore, histological examination of liver indicates a heat challenge induced abnormal lipid metabolism. Triglyceride and cholesterol level in the liver increased, however cholesterol level decreased in the serum. Genes related to lipid metabolism significantly increased in the liver after heat challenge. The present study demonstrated that high temperature cause liver damage, thus lipid metabolic was affected. Protect liver under high temperature could be one solution for coping with high temperatures in summer.


Assuntos
Temperatura Alta/efeitos adversos , Metabolismo dos Lipídeos , Fígado/metabolismo , Metaboloma , Animais , Coturnix , Feminino , Cromatografia Gasosa-Espectrometria de Massas/veterinária , Expressão Gênica , Fígado/patologia , Redes e Vias Metabólicas , Peso Molecular
6.
Sci Rep ; 8(1): 11073, 2018 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-30038322

RESUMO

Amino acids participate directly and indirectly in many important biochemical functions in the brain. We focused on one amino acid metabolic enzyme, L-amino acid oxidase (LAO), and investigated the importance of LAO in brain function using LAO1 knockout (KO) mice. Compared to wild-type mice, LAO1 KO mice exhibited impaired fear learning and memory function in a passive avoidance test. This impairment in LAO1 KO mice coincided with significantly reduced hippocampal acetylcholine levels compared to wild-type mice, while treatment with donepezil, a reversible acetylcholine esterase inhibitor, inhibited this reduction. Metabolomic analysis revealed that knocking out LAO1 affected amino acid metabolism (mainly of phenylalanine [Phe]) in the hippocampus. Specifically, Phe levels were elevated in LAO1 KO mice, while phenylpyruvic acid (metabolite of Phe produced largely by LAO) levels were reduced. Moreover, knocking out LAO1 decreased hippocampal mRNA levels of pyruvate kinase, the enzymatic activity of which is known to be inhibited by Phe. Based on our findings, we propose that LAO1 KO mice exhibited impaired fear learning and memory owing to low hippocampal acetylcholine levels. Furthermore, we speculate that hippocampal Phe metabolism is an important physiological mechanism related to glycolysis and may underlie cognitive impairments, including those observed in Alzheimer's disease.


Assuntos
Aminoácidos/metabolismo , Medo , Hipocampo/metabolismo , L-Aminoácido Oxidase/metabolismo , Memória , Aminoácidos/sangue , Animais , Regulação Enzimológica da Expressão Gênica , Hipocampo/enzimologia , L-Aminoácido Oxidase/genética , Masculino , Metaboloma , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurotransmissores/metabolismo , Piruvato Quinase/genética , Piruvato Quinase/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
Neurosci Lett ; 670: 36-40, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29355696

RESUMO

Diverse commercially available feeds are used in animal studies according to the purpose of the studies. We sought to understand the relationship between feed ingredients and their effects on animal physiology and behaviors. Here, we investigated how male laboratory mice (C57BL/6J ("B6") mice) were affected by chronic feeding with two commercially available diets, a non-purified diet (MF) and a semi-purified diet (AIN-93G). In B6 mice, both diets similarly induced spontaneous activities in the home cage and the open field box, anxiety in the elevated plus maze test, and depressive-like behaviors in tail-suspension and forced-swimming tests, and with both diets, similar data were obtained on calorie intake, water intake, body weight gain, and plasma corticosterone levels. By contrast, liver weight was significantly higher in MF-fed B6 mice than in AIN-93G-fed B6 mice. Furthermore, the cecum microbiome was drastically affected by the diets, and, specifically, Allobaculum was the major genus (43.4%) in the cecum microbiota of AIN-93G-fed mice but its abundance was reduced (to 3.8%) in the case of MF-fed mice. Future studies should address whether the differences in diet purity and cecum microbiota influence brain functions and behaviors in B6 mice.


Assuntos
Ração Animal , Comportamento Animal/fisiologia , Ceco/microbiologia , Corticosterona/sangue , Microbiota , Animais , Dieta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/fisiologia
8.
J Reprod Dev ; 63(3): 325-332, 2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28381667

RESUMO

During mammary gland involution, the epithelial mesenchymal transition (EMT) process plays an important role in tissue remodelling and in the termination of milk production. Transforming growth factor ß (TGFß) has been known as a central inducer to EMT and contributor to the mammary gland involution. However, the whole mechanism has accomplished the EMT process in mammary gland is still unclear. Here, we show that arachidonic acid, one of the major products in milk, is new player to control the EMT together with TGFß during mammary gland involution. Firstly, we observed decrease in CDH1 (epithelial marker gene) expression and increases in VIM and TWIST1 (mesenchymal marker genes), TGFB1, and PLCG2 (arachidonic acid synthesis gene) at involution. In epithelial cells culture experiments, depletion of lactogenic hormones to mimic the involution induced TGFß1 and PLCG2 expressions. Treatment with arachidonic acid in epithelial cells increased VIM and TWIST1 expressions without decrease of CDH1 expression, while TGFß1 decreased CDH1 and increased VIM and TWIST1; more importantly, TGFß1 induced the expression of PLCG2, but arachidonic acid did not induce the expression of TGFB1. Finally, arachidonic acid accelerated the TGFß1 increasing VIM and TWIST1 expressions, meanwhile arachidonic acid synthase inhibitor partially blocked the TGFß1 increasing VIM and TWIST1 expressions. In conclusion, TGFß1 stimulates arachidonic acid synthesis and the arachidonic acid has a function to postulate the EMT process together with TGFß1 during mammary gland involution.


Assuntos
Ácido Araquidônico/metabolismo , Transição Epitelial-Mesenquimal , Glândulas Mamárias Animais/fisiologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Feminino , Camundongos Endogâmicos ICR
9.
Biol Reprod ; 95(4): 78, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27512153

RESUMO

Exposure to endocrine-disrupting chemicals (EDCs) during fetal and neonatal periods can have toxic effects that are irreversible and last a lifetime. However, the mechanism underlying this phenomenon is still unknown. Here, we show the effect of 17alpha-ethynyl estradiol (EE) on the development of the primordial follicle during early ovarian development in female rats. Microarray analysis revealed the down-regulation of Hrk, an activator of apoptosis, in neonatal ovaries exposed to EE. Real-time PCR analysis also showed a decrease of Hrk mRNA expression in ovaries treated with EE both in vitro and in neonatal rats. An immunostaining assay showed that HRK protein and cleaved caspase 3 colocalize in the oocytes at Postnatal Day 1 (PND1). The EE-exposed ovaries had a reduced number of oocytes positive for TUNEL staining compared to control ovaries at PND1. Abnormal follicle formation of EE-exposed ovaries was observed at PND7 and PND21. A TUNEL staining assay revealed that Hrk depletion reduced the number of apoptotic oocytes. In addition, down-regulation of Hrk mRNA expression was observed in ovaries treated with other estrogenic chemicals. We propose a model in which EE inhibits oocyte apoptosis in the neonatal ovary by suppressing the expression of Hrk, thereby disrupting follicle formation and ovary function.


Assuntos
Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Estrogênios/toxicidade , Neuropeptídeos/antagonistas & inibidores , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Ovário/efeitos dos fármacos , Ovário/fisiologia , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Caspase 3/metabolismo , Regulação para Baixo/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Etinilestradiol/toxicidade , Feminino , Técnicas de Silenciamento de Genes , Masculino , Modelos Biológicos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oócitos/patologia , Folículo Ovariano/patologia , Ovário/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Transcriptoma/efeitos dos fármacos
10.
Neurotoxicology ; 56: 64-75, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27395752

RESUMO

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


Assuntos
Deficiências do Desenvolvimento/induzido quimicamente , Doenças do Sistema Endócrino/induzido quimicamente , Hipotálamo/patologia , Kisspeptinas/metabolismo , Neurônios/metabolismo , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Fatores Etários , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Deficiências do Desenvolvimento/patologia , Modelos Animais de Doenças , Doenças do Sistema Endócrino/patologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Ciclo Estral/efeitos dos fármacos , Feminino , Hormônios/metabolismo , Hipotálamo/efeitos dos fármacos , Kisspeptinas/genética , Neurônios/efeitos dos fármacos , Ovariectomia , Gravidez , Progesterona/farmacologia , Cloridrato de Raloxifeno/farmacologia , Ratos , Tamoxifeno/farmacologia
11.
Biol Reprod ; 93(2): 32, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26134866

RESUMO

Neonatal exposure to estrogens is known to cause delayed effects, a late-occurring adverse effect on adult female reproductive functions, such as early onset of age-matched abnormal estrous cycling. However, the critical period in which neonates are sensitive to delayed effects inducible by exogenous estrogen exposure has not been clearly identified. To clarify this window, we examined the intensity and timing of delayed effects using rats exposed to ethynylestradiol (EE) at various postnatal ages. After subcutaneous administration of a single dose of EE (20 µg/kg, which induces delayed effects) on Postnatal Day (PND) 0, 5, 10, or 14 in Wistar rats, hypothalamic and hormonal alterations in young adults and long-term estrous cycling status were investigated as indicators of delayed effects. In young adults, peak luteinizing hormone concentrations at the time of the luteinizing hormone surge showed a decreasing trend, and KiSS1 mRNA expression of the anterior hypothalamus and number of KiSS1-positive cells in the anteroventral periventricular nucleus were significantly decreased in the PND 0, 5, and 10 groups. The reduction in KiSS1 mRNA and KiSS1-postive cells was inversely correlated with age at time of exposure. These groups also exhibited early onset of abnormal estrous cycling, starting from 17 wk of age in the PND0 group and 19 wk of age in the PND5 and 10 groups. These indicators were not apparent in the PND14 group. Our results suggest that PND0-PND10 is the critical window of susceptibility for delayed effects, and PND14 is presumed to be the provisional endpoint of the window.


Assuntos
Etinilestradiol/toxicidade , Envelhecimento , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Ciclo Estral/efeitos dos fármacos , Etinilestradiol/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Hipotálamo/efeitos dos fármacos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo Anterior/metabolismo , Kisspeptinas/biossíntese , Kisspeptinas/genética , Hormônio Luteinizante/sangue , Gravidez , Efeitos Tardios da Exposição Pré-Natal , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Diferenciação Sexual/efeitos dos fármacos , Vagina/efeitos dos fármacos , Vagina/crescimento & desenvolvimento , Doenças Vaginais/induzido quimicamente , Doenças Vaginais/patologia
12.
J Cell Physiol ; 230(11): 2755-62, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25820447

RESUMO

L-amino acid oxidase (LAO), a classic flavoprotein, shows antibacterial activity by producing hydrogen peroxide. LAO exists in many tissues such as salivary gland, thymus, spleen, small intestine and testis. In particular, LAO was highly expressed in mice milk and plays an important factor in innate immunity of mammary glands. However, the mechanism which LAO expression is regulated spatially and temporally in lactating mammary glands has been unclear. In this study, we showed the contribution of lactogenic hormone and epigenetic control on LAO gene expression. In monolayer of mammary epithelial cells, treatment of lactogenic hormone mixture, dexamethasone, insulin and prolactin, did not induce LAO mRNA expression and its promoter activity, even though one of milk protein ß-casein expression was stimulated. However, increase of LAO expression was observed when the cells were treated with lactogenic hormones in a 3-dimensional culture. The results of chromatin immunoprecipitation analysis revealed that histone H3K18 acetylation increased and histone H3K27 tri-methylation decreased with lactation, which is associated with a period of high LAO expression. Moreover, the treatment of histone methylation inhibitor (DZNep) as well as histone deacetylation inhibitor (Trichostatine A) induced LAO expression in monolayer of mammary cells. Taken together, this is the first demonstration showing that LAO expression is induced in cell culture, and stimulation of lactogenic hormone and change of histone modification are promising signals to show highly expression of LAO in lactating mammary glands.


Assuntos
L-Aminoácido Oxidase/biossíntese , Lactação/genética , Glândulas Mamárias Animais/metabolismo , Prolactina/biossíntese , Animais , Epigênese Genética , Feminino , Regulação da Expressão Gênica , Histonas/antagonistas & inibidores , Histonas/metabolismo , Ácidos Hidroxâmicos/administração & dosagem , Imunidade Inata/genética , L-Aminoácido Oxidase/metabolismo , Metilação , Camundongos , RNA Mensageiro/biossíntese
13.
Reprod Toxicol ; 51: 145-56, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25615539

RESUMO

Neonatal exposure to 17alpha-ethynylestradiol (EE) causes delayed effect, a late-occurring irreversible damage to reproductive functions characterized by the early onset of age-matched abnormal estrous cycling. To clarify the involvement of a hypothalamic key cycling regulator KiSS1/GPR54 in the delayed effect, we investigated artificially induced LH surges and KiSS1 mRNA expression in the anteroventral periventricular nucleus (AVPV) of cycling young adult rats neonatally exposed to EE, and compared these parameters to those in about 5 months old middle-aged rats. KiSS1 mRNA expression, the number of KiSS1-positive cells and KiSS1/ERα co-expressing cells in the AVPV decreased in both EE-exposed and middle-aged rats. The peak area and levels of LH surge dose-dependently decreased in EE-exposed rats, and reduction was more evident in middle-aged rats. These results indicate that the prior attenuation of KiSS1 and consequent depression of LH surges plays a key role in the onset of abnormal estrous cycling in the delayed effect.


Assuntos
Etinilestradiol/toxicidade , Hipotálamo Anterior/efeitos dos fármacos , Kisspeptinas/genética , Hormônio Luteinizante/sangue , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Feminino , Hormônio Foliculoestimulante/sangue , Hipotálamo Anterior/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Transdução de Sinais/efeitos dos fármacos
14.
J Vet Med Sci ; 76(8): 1105-10, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24784441

RESUMO

Contamination of estrogenic compounds disrupts endocrinological and neurological reproductive systems in animals. Neonatal exposure to 17α-ethinyl estradiol (EE) induced an abnormal estrous cycle at postnatal day (PND) 180, but not at PND90. We found that serum level of luteinizing hormone (LH) at the latter half of proestrus in EE-treated rats was lower than in the controls at PND90 when there was no significant difference on estrous cyclicity. Additionally, kiss1 mRNA levels in the anteroventral periventricular nucleus-preoptic area (AVPV/POA) were lower in EE-treated rats than in the controls. The expression of GnRH precursor (GNRH1) mRNA in the AVPV/POA and that of LH beta subunit (LHb) mRNA in the pituitary were similar in the control- and EE-treated groups. Our results indicated that neonatal exposure to EE leads to reduced expression of kiss1 mRNA in AVPV/POA and LH-surge, which is likely related to the delayed reproductive dysfunction seen in adult female rats.


Assuntos
Animais Recém-Nascidos , Etinilestradiol/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Kisspeptinas/metabolismo , Hormônio Luteinizante/sangue , Reprodução/efeitos dos fármacos , Análise de Variância , Animais , Feminino , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
15.
Reprod Toxicol ; 46: 77-84, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24632129

RESUMO

Neonatal exposure to synthetic estrogen causes delayed reproductive dysfunction in female rats. Exposure to 17α-ethynyl estradiol (EE, low: 20 and high: 2000 µg/kg) induced an abnormal estrous cycle during PND171-190 in low-dose and PND126-145 in high-dose group. At PND90 within normal estrous cycle, high-dose animals showed lack of LH surge and low of ovarian hormones in serum level. Gene expression analysis demonstrated that level of mRNA encoding luteinizing hormone/chorionic gonadotropin receptor (LHCGR) was higher in EE-treated ovaries than in control ovaries, and LHCGR protein colocalized with apoptosis-related proteins in the interstitial area of the ovary. At PND1, ovarian LHCGR mRNA levels were higher in EE-treated rats than in control rats, and direct induction of LHCGR expression by EE was observed in vitro. Our results indicate that neonatal exposure to EE induces irregular LHCGR expression in the immature ovary, which may influence the occurrence of delayed reproductive dysfunction in adult animals.


Assuntos
Ciclo Estral/efeitos dos fármacos , Etinilestradiol/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Ovário/metabolismo , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Tamanho do Órgão/efeitos dos fármacos , Ovário/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...