Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Ovarian Res ; 14(1): 173, 2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34895279

RESUMEN

INTRODUCTION: The hypothalamic-pituitary-ovarian (HPO) axis is the principal regulator of the reproductive system. The neurons in the arcuate nucleus of the hypothalamus signal the basophilic cells of the anterior pituitary to release luteinizing hormone (LH) and follicle stimulating hormone (FSH), which bind to the granulosa and theca cells of a follicle in the ovary to promote healthy follicular development. Disruption of this process at any time can lead to polycystic ovaries and, if left untreated, can lead to Polycystic Ovarian Syndrome (PCOS), one of the leading causes of infertility. A novel treatment option using 150 kHz Intermediate Frequency (IF) Electromagnetic Radiation (EMR) has been proposed to monitor the effect of this frequency during cystic development. METHODS: To prove this, an experiment was conducted to study the effect of whole-body exposure to 150 kHz EMR for 8 weeks at receptor, cellular, tissue and hormonal levels on the HPO axis of 25 young cyclic female rats. RESULTS: The results showed that 150 kHz EMR did not affect the histoarchitecture of neurons of arcuate nucleus of the hypothalamus of PCO-induced rats. It was also found that the number of basophilic cells of the pituitary gland was increased and the immunoreactivity of LH and FSH secretion increased. This EMR field also decreased the development of follicular cysts in the ovary and possibly increased the immunoreactivity of the LH and FSH receptors as well on the theca and granulosa cells of follicles in the ovary. CONCLUSION: There are still many limitations to this study. If properly evaluated, the results of this experiment could help develop a new non-invasive treatment option for women with PCOS in the near future.


Asunto(s)
Magnetoterapia , Síndrome del Ovario Poliquístico/terapia , Animales , Modelos Animales de Enfermedad , Radiación Electromagnética , Estradiol , Femenino , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante/metabolismo , Hipotálamo/metabolismo , Hipotálamo/patología , Hormona Luteinizante/sangre , Hormona Luteinizante/metabolismo , Neuronas/citología , Ovario/metabolismo , Ovario/patología , Hipófisis/metabolismo , Hipófisis/patología , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Ratas Sprague-Dawley , Receptores de HFE/metabolismo , Receptores de HL/metabolismo
2.
J Reprod Dev ; 66(5): 475-483, 2020 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-32713881

RESUMEN

Iron is important for many cellular functions, including ATP synthesis and cell proliferation. Insufficient of iron in the diet causes iron deficiency anemia (IDA), which often occurs in people living in the world. Since 50% of women with IDA show amenorrhea, the relationship of between iron deficiency and reproductive function was assessed using mice fed a low Fe diet (LFD). The estrous cycle in the LFD mice was blocked at diestrus, which impair follicle development, and fertility. Further, even LFD mice were injected with exogenous pregnant mare serum gonadotropin (PMSG), follicular development was ceased at the secondary follicle stage, and preovulatory follicles were not observed. Amount of ATP decreased in the ovary of the LFD mice, and expression of follicle development markers (Fshr, Cyp19a1, Ccnd2) and estradiol-17ß (E2) was low level compared to levels mice fed a normal diet. Feeding a normal diet with sufficient iron to the LFD mice for an additional 3 weeks completely reversed absence the effects of iron insufficient on the estrous cycle and infertility. Thus, iron restriction depresses ovary functions, especially follicular development from secondary follicle to antral follicles and infertility. These effects are fully reversible by supplementation of a normal diet containing iron.


Asunto(s)
Hierro/química , Folículo Ovárico/metabolismo , Adenosina Trifosfato/metabolismo , Anemia Ferropénica , Animales , Aromatasa/metabolismo , Peso Corporal , Ciclina D2/metabolismo , Estradiol/sangre , Estrógenos/metabolismo , Ciclo Estral/efectos de los fármacos , Femenino , Hormona Folículo Estimulante/metabolismo , Gonadotropinas Equinas/farmacología , Hierro/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Folículo Ovárico/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovulación/efectos de los fármacos , Progesterona/sangre , ARN Mensajero/metabolismo , Receptores de HFE/metabolismo
3.
J Anim Physiol Anim Nutr (Berl) ; 104(1): 371-378, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31724249

RESUMEN

In order to investigate the mechanism of genistein (Gen) in the treatment of climacteric syndrome, an in vivo study was performed to investigate the beneficial effects of genistein on the expression of P450 aromatase (P450 arom) and follicle-stimulating hormone receptor (FSHR) in the mouse ovary and uterus. Fifty female ICR mice (45 ± 5g, n = 50), aged 12 months, were divided into the following five groups with 10 animals in each: blank control group (CG), low-dose genistein group (L-Gen), middle-dose genistein group (M-Gen) and high-dose genistein group (H-Gen) (received 15, 30 and 60 mg/kg of genistein, respectively), and oestrogen group (EG; received 0.5 mg/kg diethylstilbestrol). The expression levels of the FSHR protein were determined by an immunohistochemical staining method. The expression of P450 arom, Cytochrome P450 19 (CYP19) and FSHR was quantified by real-time PCR. Immunohistochemical results showed that the expression levels of the FSHR protein in the M-Gen (average stained area: 20.79) and the H-Gen (average stained area: 21.21) groups were significantly stronger than in the CG (average area was 17.24) group (p < .05). The expression levels of CYP19 mRNA and P450 arom were positively correlated with the dose of genistein. Specifically, the relative expression levels in the H-Gen and EG groups were more than 1.5 times higher than in the CG group (p < .05). Genistein played a significant role in regulating aromatase and FSHR gene expression to improve perimenopausal ovarian and uterine function.


Asunto(s)
Aromatasa/metabolismo , Genisteína/farmacología , Menopausia , Síndrome Metabólico/tratamiento farmacológico , Receptores de HFE/metabolismo , Animales , Aromatasa/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Ovario/efectos de los fármacos , Ovario/metabolismo , Receptores de HFE/genética , Transcriptoma
4.
Domest Anim Endocrinol ; 71: 106391, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31731250

RESUMEN

Corpus luteum (CL), a transient endocrine gland critical for reproductive cyclicity and pregnancy maintenance, is controlled by numerous regulatory factors. Although LH is widely recognized as the major regulator, other factors may also affect luteal functions. It has been demonstrated that FSH receptors (FSHR) are expressed not only in ovarian follicles but also in other tissues within the reproductive tract, including the CL. To evaluate FSHR expression in nontreated (nonsuperovulated; experiment 1) or FSH-treated (superovulated; experiment 2) sheep fed a control (C; maintenance), excess (O; 2 × C), or restricted (U; 0.6 × C) diet, CL were collected at the early, mid and/or late luteal phases (n = 5-7 per group). Protein and messenger RNA (mRNA) expression of FSHR were detected in the CL from all groups using immunohistochemistry followed by image analysis and quantitative RT-PCR, respectively. Follicle-stimulating hormone receptor was immunolocalized to steroidogenic small and large and nonsteroidogenic luteal cells. In both experiments, FSHR protein expression was not affected by stage of luteal development or diet. In experiment 1, expression of mRNA for all FSHR variants was greater (P <0.02 to 0.0003) at the late phase than mid or early luteal phase, and in experiment 2, it was greater (P < 0.001) at the mid than early luteal phase. Plane of nutrition did not affect FSHR mRNA expression. Comparison of FSH-treated with nontreated ewes demonstrated that FSH increased FSHR protein expression by 1.5- to 2-fold (P < 0.0001) in all groups, and mRNA expression by 7- to 30-fold (P < 0.001) for (1) FSHR-1 in all groups except U at the early luteal phase, (2) FSHR-2 in C, O, and U at the mid-phase, but not early luteal phase, and (3) FSHR-3 in U at the mid-luteal phase. Our data demonstrate that (1) FSHRs are expressed in ovine CL at several stages of luteal development, (2) FSHR protein expression does not change during the luteal phase and is not affected by diet, (3) FSHR mRNA expression not only depends on the stage of the estrous cycle but also not affected by diet in nonsuperovulated or superovulated ewes, and (4) in vivo FSH treatment enhanced FSHR protein and/or mRNA expression in the CL depending on diet and phase of the estrous cycle. Presence of FSHR in the CL indicates a regulatory role of FSH in luteal function in sheep. As very little is known about the possible role of FSH and FSHR in luteal functions, further studies should be undertaken to elucidate the endocrine, molecular, and cellular mechanisms of FSH effects on the CL.


Asunto(s)
Cuerpo Lúteo/metabolismo , Hormona Folículo Estimulante/farmacología , Receptores de HFE/metabolismo , Ovinos , Animales , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Estado Nutricional , Receptores de HFE/genética
5.
Pol J Vet Sci ; 22(1): 83-90, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30997768

RESUMEN

The purpose of the study was to study the activity of the phytoestrogen genistein (GEN) act- ing on FSHR and LHR in rat ovaries with polycystic ovary syndrome (PCOS). Sixty rats were di- vided into six groups. Rats in the dose group received genistein at a concentration of either 5 (low genistein dose group, L-gen), 10 (middle genistein dose group, M-Gen) or 20 (high genistein dose group, H-Gen) mg per kg of body weight per day. Estrogen group (EG, received 0.5 mg/kg Dieth- ylstilbestrol). Concentration of sex hormones in serum was quantified by enzyme-linked immuno- sorbent assay (ELISA). Expressions of follicle-stimulating hormone receptor (FSHR) and lutein- izing hormone receptor (LHR) protein were determined by immunohistochemistry. Treatment with genistein resulted in a strong stimulation of the concentration of sex hormone in serum. The concentration of progesterone and FSH was significantly higher in H-Gen when compared to the PCOS model control group (MG) (P ⟨ 0.01). In contrast, the concentration of testosterone, LH and the ratio of LH/FSH decreased in GEN treatment groups compared to MG, the effect was statistically significant, tested by the ANOVA test (p⟨0.01). For hormone receptor activity, treat- ment with genistein resulted in an improvement of ovarian function with LHR protein expression being enhanced and FSHR protein expression being suppressed. Our results demonstrate that Genistein played a significant role in regulating FSH and LH receptor expression to improve perimenopausal ovarian and uterine function.


Asunto(s)
Genisteína/farmacología , Síndrome del Ovario Poliquístico/metabolismo , Receptores de HFE/metabolismo , Receptores de HL/metabolismo , Animales , Femenino , Hormona Folículo Estimulante/sangre , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Hormona Luteinizante , Fitoestrógenos , Síndrome del Ovario Poliquístico/patología , Ratas , Ratas Wistar
6.
Can J Physiol Pharmacol ; 96(4): 395-403, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28977777

RESUMEN

Exhaustive exercises can cause delayed menarche or menstrual cycle irregularities in females. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs) are incorporated into a wide range of benefits in many physiological systems. Our work aimed to assess the role of ω-3 PUFA docosahexaenoic acid (DHA) on the deleterious effects of exhaustive exercise on the female reproductive system in rats. Virgin female rats were randomly divided into 4 groups (12 rats in each): control group, omega-3 group treated with DHA, exhaustive exercise group, and exhaustive exercised rats treated with DHA. Omega-3 was given orally to the rats once daily for 4 estrous cycles. Exhaustive exercises revealed lower levels in progesterone and gonadotropins together with histopathological decrease in number of growing follicles and corpora lutea. Moreover, the exercised rats showed low levels of ovarian antioxidants with high level of caspase-3 and plasma cortisol level that lead to disruption of hypothalamic-pituitary-gonadal axis. ω-3 PUFA DHA has beneficial effects on the number of newly growing follicles in both sedentary and exercised rats with decreasing the level of caspase-3 and increasing the antioxidant activity in ovaries. Exhaustive exercises can cause ovulatory problems in female rats that can be improved by ω-3 supplementation.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Ovulación/efectos de los fármacos , Condicionamiento Físico Animal , Animales , Suplementos Dietéticos , Femenino , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/crecimiento & desarrollo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo
7.
Biol Trace Elem Res ; 183(2): 296-304, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28856574

RESUMEN

The aim of this experiment is to explore the effects of aluminum chloride (AlCl3) on the ATPase enzymes and gonadotropin receptors in the testes. Eighty male Wistar rats were orally exposed to 0 mg/kg body weight (BW) (control group, CG), 64 mg/kg BW (low-dose group, LG), 128 mg/kg BW (mid-dose group, MG), or 256 mg/kg BW (high-dose group, HG) for 120 days. The microstructure and ultrastructure of testes; the activities of Na+-K+-ATPase, Mg2+-ATPase, and Ca2+-ATPase; and the mRNA and protein expressions of follicle-stimulating hormone receptor (FSHR) and luteinizing hormone receptors (LHR) in the testes were examined. The results showed that the testes histological structure were damaged; the activities of Na+-K+-ATPase, Mg2+-ATPase, and Ca2+-ATPase, the mRNA and protein expressions of FSHR and LHR in the testes were all decreased in the rats with AlCl3 exposure. It indicates that AlCl3 causes the dysfunction of testes in rats.


Asunto(s)
Compuestos de Aluminio/toxicidad , ATPasa de Ca(2+) y Mg(2+)/metabolismo , ATPasas Transportadoras de Calcio/metabolismo , Cloruros/toxicidad , Receptores de Gonadotropina/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Testículo/efectos de los fármacos , Testículo/metabolismo , Cloruro de Aluminio , Animales , Masculino , Ratas , Ratas Wistar , Receptores de HFE/metabolismo
8.
Reprod Fertil Dev ; 29(12): 2496-2505, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28641706

RESUMEN

Bisphenol A (BPA) is a synthetic endocrine-disrupting chemical of high prevalence in the environment, which may affect the function of the hypothalamic-pituitary-testis (HPT) axis in adult rats. The aim of the present study was to evaluate whether exposure to BPA during hypothalamic sexual differentiation at doses below the reproductive no observable adverse effect level of the World Health Organization causes changes in the regulation of the HPT axis. For this, 0.5 or 5mgkg-1 BPA was injected subcutaneously to the mothers from gestational day 18 to postnatal day (PND) 5. In adulthood (PND90), the mRNA expression of genes related to HPT axis was evaluated in hypothalamus, pituitary and testis. Hypothalamic expression of gonadotrophin-releasing hormone (Gnrh) and estrogen receptor 2 (Esr2) mRNA was increased in both BPA-treated groups compared to control group. In the pituitary, follicle stimulating hormone beta subunit (Fshb) and androgen receptor (Ar) mRNA expression was increased compared to control group in rats treated with 0.5mgkg-1 of BPA, whereas estrogen receptor 1 (Esr1) mRNA expression was only increased in the group treated with 5mgkg-1of BPA, compared to control group. In the testis, there was increased expression of FSH receptor (Fshr) and inhibin beta B subunit (Inhbb) transcripts only in rats treated with 0.5mgkg-1 of BPA. Serum testosterone and LH concentrations were increased in the group treated with 5mgkg-1of BPA. The results of the present study demonstrate for the first time that perinatal exposure to low doses of BPA during the critical period of hypothalamic sexual differentiation modifies the activity of the HPT axis in the offspring, with consequences for later life in adult rats.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Disruptores Endocrinos/farmacología , Expresión Génica/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Fenoles/farmacología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Maduración Sexual/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Receptor alfa de Estrógeno/metabolismo , Femenino , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Embarazo , Ratas , Receptores de HFE/metabolismo , Maduración Sexual/fisiología , Testículo/metabolismo
9.
Reprod Domest Anim ; 52(3): 477-482, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28181328

RESUMEN

Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) have a central role in follicle growth, maturation and oestrus, but no clear pathway in the seasonal oestrus of yak (Bos grunniens) has been found. To better understand the role of FSH and LH in seasonal oestrus in the yak, six yaks were slaughtered while in oestrus, and the pineal gland, hypothalamus, pituitary gland, and gonads were collected. Using real-time PCR and immunohistochemical assays, we determined the mRNA and protein expression of the FSH and LH receptors (FSHR and LHR) in these organs. The analysis showed that the FSHR mRNA expression level was higher in the pituitary gland tissue compared with LHR (p < .01) during oestrus. By contrast, there was low expression of FSHR and LHR mRNA in the pineal gland and hypothalamus. FSHR mRNA expression was higher than that of LHR (p < .05) in the ovary, whereas LHR mRNA expression was higher than that of FSHR (p < .01) in the uterus. FSHR and LHR proteins were located in the pinealocyte, synaptic ribbon and synaptic spherules of the pineal gland and that FSH and LH interact via nerve fibres. In the hypothalamus, FSHR and LHR proteins were located in the magnocellular neurons and parvocellular neurons. FSHR and LHR proteins were localized in acidophilic cells and basophilic cells in the pituitary gland, and in surface epithelium, stromal cell and gland epithelium in the uterus. In the ovary, FSHR and LHR protein were present in the ovarian follicle. Thus, we concluded that FSHR and LHR are located in the pineal gland, hypothalamus, pituitary and gonad during oestrus in the yak. However, FSHR was mainly expressed in the pituitary gland and ovaries, whereas LHR was mainly expressed in the pituitary gland and uterus.


Asunto(s)
Bovinos/fisiología , Estro/fisiología , ARN Mensajero/genética , Receptores de HFE/genética , Receptores de HL/genética , Animales , Femenino , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Ovario/metabolismo , Glándula Pineal/metabolismo , Hipófisis/metabolismo , ARN Mensajero/metabolismo , Receptores de HFE/metabolismo , Receptores de HL/metabolismo , Estaciones del Año , Útero/metabolismo
10.
Reprod Domest Anim ; 52 Suppl 2: 65-70, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28120353

RESUMEN

Maturation of oocytes is a prerequisite for successful embryo development. The fertilization competence of in vivo derived oocytes is significantly higher than that of oocytes matured in vitro. Commonly evaluated morphological criteria for oocyte maturation do not reflect the complexity and quality of maturation processes. Oocytes and granulosa cells are communicating closely in a bidirectional way during follicular growth and maturation. Assessing the mRNA expression of specific genes in granulosa cells could be a non-invasive way to evaluate the conditions of in vitro oocyte maturation. The objective of this study was to elucidate the influence of two different FSH additives on the in vitro maturation rate and gene expression of cumulus-oocytes complexes in domestic cat. Feline oocytes were matured in a medium, supplemented with LH and 0.02 IU/ml porcine FSH versus 0.02 IU or 1.06 IU/ml human FSH. Granulosa cells were separated from oocytes directly after 24 hr of maturation or after additional 12 hr of in vitro fertilization. Gene expression levels were analysed by quantitative PCR for aromatase, antimullerian hormone, follicle stimulating hormone receptor (FSHR), luteinizing hormone/choriogonadotropin receptor (LHCGR) and prostaglandin E synthase. Neither oocyte maturation rate nor gene expression levels differed after 24 or 36 hr in all three groups. However, variations were discovered in correlations of expression levels, for instance for FSHR and LHCG, indicating differences in the fine-tuning of in vitro maturation processes under varying FSH supplementations. We suppose that correlation between gene expressions of selected genes suggests a superior maturation quality of feline oocytes.


Asunto(s)
Gatos/fisiología , Células de la Granulosa/metabolismo , Técnicas de Maduración In Vitro de los Oocitos , Oocitos/citología , Receptores de HFE/metabolismo , Receptores de HL/metabolismo , Animales , Aromatasa/genética , Femenino , Fertilización In Vitro , Hormona Folículo Estimulante/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hormona Luteinizante/metabolismo , Oogénesis , Receptores de HFE/genética , Receptores de HL/genética
11.
Life Sci ; 173: 150-160, 2017 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-27569590

RESUMEN

AIM: Beta asarone is the major constituent of oil obtained from Acorus calamus, the Indian traditional medicine plant. Several studies have shown that beta asarone causes liver and cardiac damages but the reproductive toxicity is not well understood. The present study was initiated to investigate whether beta asarone has the potential to cause reproductive toxicity by inducing oxidative stress in the testis of male Wistar albino rats. MATERIALS AND METHODS: For this study, the animals were divided into six groups: Group I was treated with saline (normal saline), Group II with DMSO (vehicle control) and Group III with cisplatin (10mg/kgb.wt.). Group IV, V and VI animals were administrated at three dose levels of beta asarone 12.5, 25 and 50mg/kgb.wt. The treatment was carried out for 14days and animals were sacrificed on 29th day and processed for sperm analysis, hormone assay, histopathological, and antioxidant enzymatic assays. We also used molecular docking studies to predict the binding nature of beta asarone with luteinizing hormone receptor (LHR) and follicle-stimulating hormone receptor (FSHR). KEY FINDINGS: Beta asarone administered at a dose of 50mg/kgb.wt. was responsible for inducing certain noticeable degenerative changes in histopathological analysis of the tissue. This was supported by altered sperm morphology and hormonal variations when compared to the control groups. Antioxidant enzyme levels were also found to be decreased. This was further validated by molecular docking studies. SIGNIFICANCE: The present study provides evidence that beta asarone administered at a dose of 50mg/kg b.wt. is capable enough in bringing about moderate amount of degenerative changes in rat testis and altered antioxidant status. Therefore provides a suitable evidence to prove that beta asarone causes reproductive toxicity.


Asunto(s)
Anisoles , Simulación por Computador , Infertilidad Masculina , Simulación del Acoplamiento Molecular , Receptores de HFE , Receptores de HL , Acorus/química , Derivados de Alilbenceno , Animales , Anisoles/química , Anisoles/toxicidad , Antioxidantes/metabolismo , Infertilidad Masculina/inducido químicamente , Infertilidad Masculina/enzimología , Infertilidad Masculina/patología , Masculino , Ratas , Ratas Wistar , Receptores de HFE/química , Receptores de HFE/metabolismo , Receptores de HL/química , Receptores de HL/metabolismo , Espermatozoides/enzimología , Espermatozoides/patología
12.
Pol J Vet Sci ; 20(4): 759-768, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29611661

RESUMEN

The present study aimed to assess LH effects on in vitro maturation (IVM) and apoptosis and also to explore the gene expressions of LHR and FSHR in cumulus-oocyte complexes (COCs) of the sheep. COCs were in vitro matured 24h in the IVM medium supplemented with varying concentrations of LH (0, 5, 10, 20 and 30 µg/mL). They were allocated into LH-1 (control group), LH-2, LH-3, LH-4 and LH-5 groups, respectively. FSH (10 IU/mL) addition was as a positive control (FSH group). COCs apoptosis was assessed by TUNEL. The qPCR and Western blotting were utilized to detect mRNA and protein expressions of FSHR and LHR, respectively. The results showed maturation rates of oocytes improved as LH concentration increased from 0 to 10 µg/mL (IU/mL), reaching a peak value of 44.3% in the LH-3 group. Maturation rate of LH-5 group was lower than that of LH-3 and FSH-treated groups. The lowest apoptosis rate was found in LH-3 group. The germinal vesicle break down (GVBD) rates of LH-2, LH-3 and LH-4 groups were also increased in comparison with that found in LH-1 group (control group). GVBD rate of LH-5 was lower than that in LH-3 group. The germinal vesicle (GV) rates in LH-3 and LH-4 groups were lower than those in LH-1 and LH-5 groups (p<0.05, or p<0.01). The lowest GV rate was found in LH-3 group. GV rates in LH-2, LH-4 and LH-5 groups were higher than that in FSH group (p<0.05). At hours 20, 22 and 24 after oocytes IVM, caspase-3 concentrations in four LH-treated groups were decreased in comparison with that in LH-1 group. At 24h, caspase-3 concentrations of LH-2 and LH-3 groups were lower than that in LH-1 group (p<0.05). Expression levels of FSHR and LHR mRNAs rose when LH concentrations in IVM medium increased. The greatest expressions of FSHR and LHR mRNAs were found in LH-5 and LH-3 groups (p<0.01) in comparison with those in the control group (LH-1). Meanwhile, FSHR mRNA expressions in LH-2, LH-3 and LH-4 groups were lower than that in FSH group (p<0.01 or p<0.05). Expression levels of FSHR proteins revealed no significant differences among all groups. Expression levels in LHR proteins were increased. LHR protein level in LH-2 group was higher than that in LH-1 group. In conclusion, LH treatment could promote the maturation rate and GVBD rate. LH reduced apoptosis rate, GV rate of sheep oocytes, and caspase-3 concentrations in IVM medium fluids and additionally enhanced expressions of FSHR and LHR mRNAs of sheep COCs.


Asunto(s)
Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Hormona Luteinizante/farmacología , Oocitos/fisiología , Receptores de HFE/metabolismo , Receptores de HL/metabolismo , Ovinos , Animales , Apoptosis , Caspasa 3/metabolismo , Medios de Cultivo , Células del Cúmulo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Etiquetado Corte-Fin in Situ , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de HFE/genética , Receptores de HL/genética
13.
J Endocrinol ; 226(3): 167-80, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26264981

RESUMEN

Why IVF pregnancy rates decline sharply after age 43 is unknown. In this study, we compared granulosa cell (GC) function in young oocyte donors (n=31, ages 21-29), middle-aged (n=64, ages 30-37) and older infertile patients (n=41, ages 43-47). Gene expressions related to gonadotropin activity, steroidogenesis, apoptosis and luteinization were examined by real-time PCR and western blot in GCs collected from follicular fluid. FSH receptor (FSHR), aromatase (CYP19A1) and 17ß-hydroxysteroid dehydrogenase (HSD17B) expression were found down regulated with advancing age, while LH receptor (LHCGR), P450scc (CYP11A1) and progesterone receptor (PGR) were up regulated. Upon in vitro culture, GCs were found to exhibit lower proliferation and increased apoptosis with aging. While FSH supplementation stimulated GCs growth and prevented luteinization in vitro. These observations demonstrate age-related functional declines in GCs, consistent with premature luteinization. To avoid premature luteinization in women above age 43, we advanced oocyte retrieval by administering human chorionic gonadotropin at maximal leading follicle size of 16  mm (routine 19-21  mm). Compared to normal cycles in women of similar age, earlier retrieved patients demonstrated only a marginal increase in oocyte prematurity, yet exhibited improved embryo numbers as well as quality and respectable clinical pregnancy rates. Premature follicular luteinization appears to contribute to rapidly declining IVF pregnancy chances after age 43, and can be avoided by earlier oocyte retrieval.


Asunto(s)
Envejecimiento/fisiología , Células de la Granulosa/fisiología , Luteinización/fisiología , Recuperación del Oocito/métodos , 17-Hidroxiesteroide Deshidrogenasas/genética , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Adulto , Factores de Edad , Aromatasa/genética , Aromatasa/metabolismo , Regulación hacia Abajo , Femenino , Regulación de la Expresión Génica , Humanos , Persona de Mediana Edad , Embarazo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Regulación hacia Arriba , Adulto Joven
14.
Sci Rep ; 5: 10179, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-26160710

RESUMEN

A novel protein, designated as DOI, isolated from the Chinese yam (Dioscorea opposita Thunb.) could be the first protein drug for the treatment of menopausal syndrome and an alternative to hormone replacement therapy (HRT), which is known to have undesirable side effects. DOI is an acid- and thermo-stable protein with a distinctive N-terminal sequence Gly-Ile-Gly-Lys-Ile-Thr-Thr-Tyr-Trp-Gly-Gln-Tyr-Ser-Asp-Glu-Pro-Ser-Leu-Thr-Glu. DOI was found to stimulate estradiol biosynthesis in rat ovarian granulosa cells; induce estradiol and progesterone secretion in 16- to 18-month-old female Sprague Dawley rats by upregulating expressions of follicle-stimulating hormone receptor and ovarian aromatase; counteract the progression of osteoporosis and augment bone mineral density; and improve cognitive functioning by upregulating protein expressions of brain-derived neurotrophic factor and TrkB receptors in the prefrontal cortex. Furthermore, DOI did not stimulate the proliferation of breast cancer and ovarian cancer cells, which suggest it could be a more efficacious and safer alternative to HRT.


Asunto(s)
Dioscorea/metabolismo , Estradiol/biosíntesis , Proteínas de Plantas/metabolismo , Secuencia de Aminoácidos , Animales , Aromatasa/genética , Aromatasa/metabolismo , Densidad Ósea , Huesos/diagnóstico por imagen , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Femenino , Menopausia , Datos de Secuencia Molecular , Osteoporosis/prevención & control , Ovario/citología , Péptidos/química , Péptidos/uso terapéutico , Proteínas de Plantas/química , Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor trkB/metabolismo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Rizoma/metabolismo , Microtomografía por Rayos X
15.
Mol Cell Endocrinol ; 400: 1-9, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25462584

RESUMEN

The usefulness of azaline B, a GnRH antagonist, in suppressing gonadotropin secretion in the golden hamster was examined by examining follicular development, steroidogenesis and expression of steroidogenic enzymes. Serum levels of P and E declined significantly, while FSH or LH was undetectable in azaline B-treated hamsters. FSH significantly increased serum E levels, whereas LH upregulated serum P levels. The formation of antral follicles ceased in azaline-treated hamsters, but was reversed by FSH with or without LH supplement. FSH also activated the primordial follicle pool resulting in increased formation of primary and preantral follicles. Further, an increasing trend in the formation of preantral follicles in response to E or E + P, and the formation of antral follicles in response to E + P treatment was evident. The level of Cyp11a1 mRNA increased markedly in LH- or LH + FSH-treated hamsters, whereas FSH with or without LH upregulated Cyp17a1, Cyp19a1 and Fshr mRNA expression. E without or with P also upregulated ovarian Cyp19a1 mRNA expression. The expression of enzyme protein corroborated the mRNA data. In summary, azaline B is an efficient GnRH antagonist in the hamster, and will be useful in studying the selective effect of gonadotropins on ovarian functions without disrupting the physiological functions of other hormones in ovarian cells.


Asunto(s)
Ciclo Estral/efectos de los fármacos , Hormona Folículo Estimulante/metabolismo , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Luteinizante/metabolismo , Folículo Ovárico/efectos de los fármacos , Animales , Aromatasa/genética , Aromatasa/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Estradiol/sangre , Estradiol/farmacología , Ciclo Estral/fisiología , Femenino , Hormona Folículo Estimulante/genética , Hormona Folículo Estimulante/farmacología , Regulación del Desarrollo de la Expresión Génica , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Inyecciones Subcutáneas , Hormona Luteinizante/genética , Hormona Luteinizante/farmacología , Mesocricetus , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Progesterona/sangre , Progesterona/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Transducción de Señal
16.
Artículo en Inglés | MEDLINE | ID: mdl-25395697

RESUMEN

BACKGROUND: Bisphenol-A (BPA) is one of the widespread industrial compounds, which has adverse effects on animal and human health. The study was aimed to explore the effects of Crassostrea gigas extracts (CGE) in alleviating ovarian functional disorders of female rats with exposure to BPA and the underlying possible mechanism. MATERIALS AND METHODS: Eighteen four-week-old female Sprague-Dawley (SD) rats were randomly divided into BPA group (50mg/kg BPA), BPA+CGE group (50mg/kg BPA+50mg/kg CGE), and control group (equivalent dosage of vehicle) with 6 rats in each group. After a 6-week treatment ended, the serum levels of estradiol (E2), follicle stimulating hormone (FSH), luteinizing hormone (LH) were measured by using commercial standard assay kits. The expression levels of FSH receptor (FSHR) in the rat ovarian tissues were respectively detected by immunohistochemistry and Real-time PCR. RESULTS: CGE treatment markedly increased E2 levels and decreased FSH levels in the serum (P<0.05), however, the alterations of serum LH levels were not significant (P>0.05). The protein and mRNA expression levels of FSHR were the lowest in the ovaries of control rats and the highest in BPA rats (P<0.05). CGE treatment markedly decreased the expression levels of FSHR in the ovarian tissues (P<0.05). CONCLUSIONS: Crassostrea gigas successfully alleviates ovarian functional disorders of female rats with exposure to BPA partly through decreasing FSHR expression levels in the ovarian tissues.


Asunto(s)
Compuestos de Bencidrilo/efectos adversos , Productos Biológicos/uso terapéutico , Crassostrea , Hormona Folículo Estimulante/sangre , Enfermedades del Ovario/prevención & control , Ovario/efectos de los fármacos , Fenoles/efectos adversos , Receptores de HFE/metabolismo , Animales , Productos Biológicos/farmacología , Estradiol/sangre , Estrógenos no Esteroides/efectos adversos , Femenino , Hormona Luteinizante/sangre , Enfermedades del Ovario/metabolismo , Ovario/metabolismo , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores de HFE/genética
17.
Gene ; 551(1): 15-25, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25168889

RESUMEN

The hypothalamic-pituitary-gonadal axis (HPG) plays vital roles in reproduction and steroid hormone production in both sexes. The focus of this review is upon gene structures, receptor structures and the signaling pathways of gonadotropin-releasing hormone (GnRH), luteinizing hormone (LH) and follicle-stimulating hormone (FSH). The hormones' functions in reproduction as well as consequences resulting from mutations are also summarized. Specific characteristics of hormones such as the pulsatile secretions of GnRH are also covered. The different regulators of the HPG axis are introduced including kisspeptin, activin, inhibin, follistatin, androgens and estrogen. This review includes not only their basic information, but also their unique function in the HPG axis. Here we view the HPG axis as a whole, so relations between ligands and receptors are well described crossing different levels of the HPG axis. Hormone interactions and transformations are also considered. The major information of this article is depicted in three figures summarizing the current discoveries on the HPG axis. This article systematically introduces the basic knowledge of the HPG axis and provides information of the current advances relating to reproductive hormones.


Asunto(s)
Hormona Liberadora de Gonadotropina/genética , Gonadotropinas/genética , Hipotálamo/metabolismo , Hipófisis/metabolismo , Testículo/fisiología , Animales , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Humanos , Masculino , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Transducción de Señal
18.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 34(3): 312-6, 2014 Mar.
Artículo en Chino | MEDLINE | ID: mdl-24758083

RESUMEN

OBJECTIVE: To observe the effect of Yangjing Zhongyu Decoction (YZD) on mRNA and protein expression of PCNA, StAR, and FSHR in ovarian granulose cells (GCs) cultured by excess androgen. METHODS: Ovarian GCs from porcine follicles were isolated and cultured in vitro. Follicular stimulating hormone (FSH) or YZD was added in the GCs treated by excess testosterone propionate. Totally 48 h later mRNA and protein expression of PCNA, StAR, and FSHR were detected by RT-PCR and Western blot. RESULTS: Excess androgen inhibited mRNA and protein expression of PCNA, StAR, and FSHR of GCs. FSH and YZD could antagonize inhibition of excess androgens, and promote mRNA and protein expression of PCNA, StAR, and FSHR in GCs. CONCLUSION: YZD could antagonize the inhibition of excess androgen on mRNA and protein expression of PCNA, StAR and FSHR in GCs. Thus, we inferred that YZD could improve the follicle dysplasia by promoting mRNA and protein expression of PCNA, StAR and FSHR in GCs.


Asunto(s)
Andrógenos/farmacología , Medicamentos Herbarios Chinos/farmacología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Animales , Células Cultivadas , Femenino , Hormona Folículo Estimulante/farmacología , Células de la Granulosa/citología , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Folículo Ovárico/citología , Folículo Ovárico/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Mensajero/genética , Receptores de HFE/genética , Receptores de HFE/metabolismo , Porcinos
19.
Life Sci ; 100(1): 61-6, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24530740

RESUMEN

AIMS: This experiment investigated the effects of sub-chronic aluminum chloride (AlCl3) exposure on rat ovaries. MAIN METHODS: Eighty female Wistar (5weeks old) rats, weighed 110-120g, were randomly divided into four treatment groups: control group (CG), low-dose group (LG, 64mg/kg BW AlCl3), mid-dose group (MG, 128mg/kg BW AlCl3) and high-dose group (HG, 256mg/kg BW AlCl3). The AlCl3 was administered in drinking water for 120days. The ovarian ultrastructure was observed. The activities of acid phosphatase (ACP), alkaline phosphatase (ALP), succinate dehydrogenase (SDH), Na(+)-K(+)-ATPase, Mg(2+)-ATPase and Ca(2+)-ATPase, the contents of Fe, Cu and Zn, and the protein expression of follicle-stimulating hormone receptor (FSHR) and luteinizing hormone receptor (LHR) in the ovary were determined. KEY FINDINGS: The results showed that the structure of the ovary was disrupted, the activities of ALP, ACP, SDH, Na(+)-K(+)-ATPase, Mg(2+)-ATPase and Ca(2+)-ATPase, the contents of Zn, Fe and the protein expression of FSHR and LHR were lowered, and the content of Cu was increased in AlCl3-treated rats than those in control. SIGNIFICANCE: The results indicate that sub-chronic AlCl3 exposure caused the damage of the ovarian structure, the disturbed metabolism of Fe, Zn and Cu and the decreased activities of Na(+)-K(+)-ATPase, Mg(2+)-ATPase and Ca(2+)-ATPase in the ovary, which could result in suppressed energy supply in the ovary. A combination of suppression of energy supply and reduction of expression of FSHR and LHR could inhibit ovulation and corpus luteum development, leading to infertility in female rats.


Asunto(s)
Compuestos de Aluminio/toxicidad , Cloruros/toxicidad , Ovario/efectos de los fármacos , Fosfatasa Ácida/metabolismo , Fosfatasa Alcalina/metabolismo , Cloruro de Aluminio , Animales , Cobre/metabolismo , Femenino , Infertilidad Femenina/inducido químicamente , Hierro/metabolismo , Ovario/metabolismo , Ovario/patología , Ratas , Ratas Wistar , Receptores de HFE/metabolismo , Receptores de HL/metabolismo , Zinc/metabolismo
20.
Biol Trace Elem Res ; 151(2): 217-24, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23192844

RESUMEN

Manganese (Mn) is an essential element required for normal development and reproduction. However, little is known about the reproductive toxicity of Mn in birds. To investigate the Mn-induced toxicity on testicular trace element levels and crucial hormonal parameters on male reproduction in birds, 50-day-old male Hyline cocks were fed either a commercial diet or a Mn-supplemented diet. The changes in contents of copper (Cu), iron (Fe), zinc (Zn), and calcium (Ca) in testis were detected. Hormonal parameters were evaluated including the levels of testosterone (T), luteinizing hormone (LH), follicle-stimulating hormone (FSH), thyroid-stimulating hormone (TSH), triiodothyronine (T3), and thyroxine (T4) in the serum. The mRNA levels of luteinizing hormone receptor (LHR) and follicle-stimulating hormone receptor (FSHR) were determined in this study. The results showed that Mn was accumulated in testis, and the content of Cu, Fe, Zn, and Ca decreased. Exposure to Mn significantly lowered the content of T, LH, FSH, and the mRNA expression levels of LHR and FSHR. Levels of T3 and T4 appeared with a decreased tendency, and TSH presented no obvious regularity. It indicated that Mn exposure resulted in the disbalance of testicular trace elements and influenced hormone levels in the molecular level, which may be possible underlying reproductive toxicity mechanism induced by Mn.


Asunto(s)
Pollos/metabolismo , Cloruros/farmacología , Compuestos de Manganeso/farmacología , Manganeso/metabolismo , Testículo/metabolismo , Oligoelementos/metabolismo , Alimentación Animal , Animales , Calcio/metabolismo , Cloruros/administración & dosificación , Cobre/metabolismo , Suplementos Dietéticos , Hormona Folículo Estimulante/sangre , Hierro/metabolismo , Hormona Luteinizante/sangre , Masculino , Manganeso/toxicidad , Compuestos de Manganeso/administración & dosificación , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Testículo/efectos de los fármacos , Testosterona/sangre , Tirotropina/sangre , Tiroxina/sangre , Oligoelementos/administración & dosificación , Oligoelementos/farmacología , Triyodotironina/sangre , Zinc/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA