Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 259
Filtrar
Más filtros













Intervalo de año de publicación
1.
Behav Brain Res ; 397: 112952, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33017640

RESUMEN

Low concentrations of ovarian hormones, among other factors, are associated with greater vulnerability to negative effects of environmental stressors and may trigger anxiety symptoms in females. The flavonoid chrysin (5,7-dihydroxyflavone) exerts anxiolytic-like effects in male and ovariectomized female rats, but it is unknown if chrysin could reduce anxiety-like behavior that naturally occurs through the ovarian cycle phases. The present study evaluated the effect of chrysin on anxiety-like behavior associated with the ovarian cycle phases in rats and the participation of γ-aminobutyric acid-A (GABAA) receptors in these actions. The acute effects of chrysin (2 mg/kg) were investigated in female cycling Wistar rats in the elevated plus maze, locomotor activity test, and light/dark test. Diazepam (2 mg/kg) was used as reference anxiolytic drug. The participation of GABAA receptor in the anxiolytic actions of chrysin was explored by pretreating the rats with the noncompetitive GABAA chloride ion channel antagonist picrotoxin (1 mg/kg). Chrysin and diazepam prevented anxiety-like behavior that was associated with the metestrus-diestrus phase in both the elevated plus maze and light/dark test, and these effects were reversed by picrotoxin, with no significant changes in spontaneous locomotor activity. No significant motor effects of chrysin were detected in either behavioral test during proestrus-estrus or metestrus-diestrus phases, whereas diazepam produced motor hypoactivity in the locomotor activity test during proestrus-estrus phase. These results indicate that the flavonoid chrysin prevents anxiety-like behavior that naturally occurs during metestrus-diestrus in two unconditioned models that are used to evaluate anxiety-like behavior, and these effects were mediated by actions on GABAA receptors.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Diazepam/farmacología , Ciclo Estral/efectos de los fármacos , Flavonoides/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Animales , Ansiolíticos/administración & dosificación , Diazepam/administración & dosificación , Diestro/efectos de los fármacos , Estro/efectos de los fármacos , Femenino , Flavonoides/administración & dosificación , Antagonistas de Receptores de GABA-A/administración & dosificación , Metestro/efectos de los fármacos , Picrotoxina/farmacología , Proestro/efectos de los fármacos , Ratas , Ratas Wistar
2.
Acta Histochem ; 122(8): 151630, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32992122

RESUMEN

Visfatin is a crucial adipokine, which also regulates ovarian functions in many animals. Mice estrous cycle is characterized by a dynamic complex physiological process in the reproductive system. Expression of various factors changes during the estrous cycle in the ovary. To the best of our knowledge, no previous study has been conducted on the expression of visfatin in mice ovaries during the estrous cycle. Therefore, we investigated the localization and expression of visfatin protein in the ovary of mice during the estrous cycle. Western blot analysis showed the elevated expression of visfatin in proestrus and lowest in diestrus. Immunohistochemical localization of visfatin showed intense staining in the corpus luteum of proestrus and diestrus ovaries. Thecal cells, granulosa cells, and oocytes also showed the presence of visfatin. Expression of ovarian visfatin was correlated to BCL2 and active caspase3 expression and exhibited a significant positive correlation. Furthermore, in vivo inhibition of visfatin by FK866 in the proestrus ovary down-regulated active caspase3 and PCNA expression, and up-regulated the BCL2 expression. These results suggest the role of visfatin in the proliferation and apoptosis of the follicles and specific localization of visfatin in the corpus luteum also indicate its role in corpus luteum function, which may be in progesterone biosynthesis and regression of old corpus luteum. However, further study is required to support these findings. In conclusion, visfatin may also be regulating follicular growth during the estrous cycle by regulating proliferation and apoptosis.


Asunto(s)
Cuerpo Lúteo/metabolismo , Citocinas/genética , Células de la Granulosa/metabolismo , Nicotinamida Fosforribosiltransferasa/genética , Oocitos/metabolismo , Células Tecales/metabolismo , Acrilamidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Caspasa 3/genética , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Cuerpo Lúteo/citología , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/crecimiento & desarrollo , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Diestro/efectos de los fármacos , Diestro/genética , Inhibidores Enzimáticos/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Inmunohistoquímica , Ratones , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/metabolismo , Oocitos/citología , Oocitos/efectos de los fármacos , Piperidinas/farmacología , Proestro/efectos de los fármacos , Proestro/genética , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células Tecales/citología , Células Tecales/efectos de los fármacos
3.
Biol Reprod ; 103(3): 643-653, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32352507

RESUMEN

Changes in circulating progesterone (P4) and estradiol (E2) during proestrus produce dynamic changes in endometrial function and pituitary release of gonadotropins. Independent and combined effects of P4 and E2 on endometrium and pituitary were evaluated. In a preliminary study, an exogenous hormone model of proestrus was created by removal of corpus luteum and follicles ≥5 mm followed by gradual removal of intravaginal P4 implants during 18 h and treatment with increasing doses of estradiol benzoate during 48 h to mimic proestrus using high E2 (n = 9) or low E2 (n = 9). Decreased P4, increased E2, and increased endometrial area (EA) simulated proestrus in high-E2 cows and this was used subsequently. The main experiment used a 2 × 2 factorial design with: high E2 and low P4 (n = 11); high E2 and high P4 (n = 11); low E2 and high P4 (n = 11); low E2 and low P4 (n = 10). At 48 h, gonadotropin-releasing hormone (GnRH)-induced luteinizing hormone (LH) and follicle stimulating hormone (FSH) release was determined. Variables were analyzed using PROCMIXED of Statistical Analysis System. The EA increased dramatically during 48 h only in high-E2 and low-P4 cows. For FSH, high-E2 cows had greater area under the curve (AUC) and FSH peak after GnRH than low E2, with mild negative effects of high P4. For LH, concentration at peak and AUC were 2-fold greater in high E2 compared to low-E2 groups, with low P4 also 2-fold greater than high-P4 groups. Thus, maximal changes in uterus and pituitary during proestrus depend on both low P4 and high E2, but different physiologic responses are regulated differently by E2 and P4. Changes in endometrium depend on low P4 and high E2, whereas GnRH-induced FSH secretion primarily depends on high E2, and GnRH-induced LH secretion is independently increased by high E2 or reduced by high P4.


Asunto(s)
Endometrio/efectos de los fármacos , Endometrio/metabolismo , Estradiol/farmacología , Hormona Liberadora de Gonadotropina/farmacología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Progesterona/farmacología , Animales , Área Bajo la Curva , Bovinos , Cuerpo Lúteo/efectos de los fármacos , Estradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Hormona Luteinizante/sangre , Proestro/efectos de los fármacos , Progesterona/sangre , Útero/efectos de los fármacos
4.
J Neuroendocrinol ; 32(3): e12836, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32062869

RESUMEN

The present study aimed to determine whether an i.c.v. administration of allopregnanolone (ALLO) rapidly modifies the hypothalamic and ovarian 3ß-hydroxysteroid dehydrogenase (3ß-HSD) enzymatic activity and gene expression in in vivo and ex vivo systems in pro-oestrus (PE) and dioestrus I (DI) rats. Animals were injected with vehicle, ALLO, bicuculline or bicuculline plus ALLO and were then killed. In the in vivo experiment, the hypothalamus, ovaries and serum were extracted and analysed. In the ex vivo experiment, the superior mesenteric ganglion - ovarian nerve plexus - ovary system was extracted and incubated during 120 minutes at 37 ºC. The serum and ovarian compartment fluids were used to determine progesterone by radioimmunoanalysis. In the in vivo experiments, ALLO caused a decrease in hypothalamic and ovarian 3ß-HSD enzymatic activity during PE. During DI, ALLO increased hypothalamic and ovarian 3ß-HSD activity and gene expression. The ovarian 3ß-HSD activity increased in both stages in the ex vivo system; gene expression increased only during DI. ALLO induced an increase in serum progesterone only in D1 and in the ovarian incubation liquids in both stages. All findings were reversed by an injection of bicuculline before ALLO. Ovarian steroidogenic changes could be attributed to signals coming from ganglion neurones, which are affected by the acute central neurosteroid stimulation. The i.c.v. administration of ALLO via the GABAergic system altered 3ß-HSD activity and gene expression, modulating the neuroendocrine axis. The present study reveals the action that ALLO exerts on the GABAA receptor in both the central and peripheral nervous system and its relationship with hormonal variations. ALLO is involved in the "fine tuning" of neurosecretory functions as a potent modulator of reproductive processes in female rats.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/metabolismo , Hipotálamo/efectos de los fármacos , Neuroesteroides/administración & dosificación , Ovario/efectos de los fármacos , Pregnanolona/administración & dosificación , Animales , Diestro/efectos de los fármacos , Diestro/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Hipotálamo/enzimología , Inyecciones Intraventriculares , Ovario/metabolismo , Proestro/efectos de los fármacos , Proestro/metabolismo , Progesterona/sangre , Ratas
5.
Physiol Behav ; 203: 113-119, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29273454

RESUMEN

Drug use influences sexual behavior, performance, and can be associated with increased sexual risk-taking. Our prior results using an animal model indicate that progestogens contribute to hormonally-mediated changes in sexual behavior of female rodents during acute cocaine exposure. Androgens, such as testosterone, and its metabolite 3ɑ-androstanediol (3α-diol), and estradiol, are known to influence male sexual behavior, but can also alter the expression of sexual behavior of female rodents. As such, we investigated the influence of endogenous androgen and estradiol fluctuations on cocaine-mediated changes in motor behavior and sexual receptivity of rats during diestrous or proestrous phases of the estrous cycle. Female rats were administered saline or cocaine (5, 10, or 20mg/kg, i.p.). Motor behavior was observed in the first 30min following drug administration, and then sexual responding was assessed for 15min. Cocaine decreased aggressive behavior in response to attempted mounts by a male among non-receptive (diestrous) rats and inhibited sexual behavior among sexually receptive (proestrous) rats. Cocaine dose-dependently altered concentrations of testosterone metabolites (estradiol and 3α-diol), but not testosterone, which correlated to motor and sexual behaviors of diestrous and proestrous rats, respectively. These data suggest that actions of 3α-diol may be involved in female sexual and motor behavior in response to cocaine, in a cycle-dependent manner.


Asunto(s)
Andrógenos/sangre , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Postura , Proestro/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos , Agresión/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Estradiol/sangre , Femenino , Ratas , Testosterona/sangre
6.
Reprod Fertil Dev ; 30(11): 1541-1552, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29778102

RESUMEN

The aim of the present study was to investigate the effects of a strategy for extending pro-oestrus (the interval between luteolysis and ovulation) in an oestrus synchronisation protocol (named J-Synch) in beef heifers on follicular growth, sexual steroid concentrations, the oestrogen receptor ERα and progesterone receptors (PR) in the uterus, insulin-like growth factor (IGF) 1 and pregnancy rates. In Experiment 1, heifers treated with the new J-Synch protocol had a longer pro-oestrus period than those treated with the conventional protocol (mean (±s.e.m.) 93.7±12.9 vs 65.0±13.7h respectively; P<0.05). The rate of dominant follicle growth from the time of progesterone device removal to ovulation was greater in heifers in the J-Synch than conventional group (P<0.05). Luteal area and serum progesterone concentrations were greater in the J-Synch Group (P<0.05) for the 12 days after ovulation. Progesterone receptor (PGR) staining on Day 6 after ovulation in the uterine stroma was lower in the J-Synch than conventional group (P<0.05), and the expression of PR gene (PGR) and IGF1 gene tended to be lower in J-Synch-treated heifers (P<0.1). In Experiment 2 (n=2349), the pregnancy rate 30-35 days after fixed-time AI (FTAI) was greater for heifers in the J-Synch than conventional group (56.1% vs 50.7% respectively). In conclusion, our strategy for extending pro-oestrus (i.e. the J-Synch protocol) significantly improves pregnancy establishment in beef heifers. This improvement was related to an increased rate of growth of the dominant ovulatory follicle, greater progesterone concentrations during the ensuing luteal phase and different uterine patterns of PGR and IGF1, which may have favoured embryo development and pregnancy establishment.


Asunto(s)
Estradiol/análogos & derivados , Sincronización del Estro/fisiología , Ovario/fisiología , Proestro/fisiología , Progesterona/administración & dosificación , Útero/fisiología , Animales , Bovinos , Estradiol/administración & dosificación , Estradiol/sangre , Sincronización del Estro/efectos de los fármacos , Femenino , Folículo Ovárico/diagnóstico por imagen , Folículo Ovárico/efectos de los fármacos , Ovario/diagnóstico por imagen , Ovario/efectos de los fármacos , Embarazo , Proestro/efectos de los fármacos , Progesterona/sangre , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Útero/diagnóstico por imagen , Útero/efectos de los fármacos
7.
Reprod Sci ; 24(6): 844-855, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27688242

RESUMEN

The aims of the present study were to analyze if the superior ovarian nerve (SON) plays a role in the neural signals from suprachiasmatic nucleus (SCN) that lead to ovulation and ovarian steroids secretion on proestrus day. Rats on proestrus day were treated at 11.00 to 11.30 or 17.00 to 17.30 hours with 1 of the 3 experimental procedures (1) unilateral or bilateral SON sectioning, (2) unilateral or bilateral injury to the SCN, or (3) unilateral injury to the SCN followed by unilateral sectioning of the SON ipsilateral to the treated SCN. Treatments were evaluated 24 hours after surgical procedures. Compared to laparotomized animals, right or bilateral SON sectioning treatment at 17.00 hours resulted in lower ovulation rates and number of ova shed by the right ovary. The ovaries of nonovulating animals showed early follicular luteinization signs and trapped ova. Bilateral SCN injury treatment at 11.00 hours resulted in anovulation; whereas right SCN injury treatment, with or without right SON sectioning, resulted in a lower number of ova shed. Injecting luteinizing hormone-releasing hormone to animals with bilateral SCN injury restored ovulation. In rats with unilateral or bilateral SON sectioning, or with injury to the SCN with or without unilateral sectioning of the SON, the effects on hormone levels depended of the hormone studied and the time of day treatment was performed. The present results suggest that on proestrus day, the role of the right or both SON in ovulation and steroid hormone secretion regulation takes place through different neuroendocrine mechanisms from SCN.


Asunto(s)
Estradiol/sangre , Ovario/inervación , Ovario/patología , Ovulación/fisiología , Proestro/fisiología , Progesterona/sangre , Núcleo Supraquiasmático/fisiología , Animales , Femenino , Hormona Liberadora de Gonadotropina/farmacología , Ovario/efectos de los fármacos , Ovulación/efectos de los fármacos , Proestro/efectos de los fármacos , Ratas , Testosterona/sangre
8.
Reprod Biol Endocrinol ; 14(1): 75, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27809846

RESUMEN

BACKGROUND: Muscarinic receptors (mAChRs) of the preoptic and anterior hypothalamus areas (POA-AHA) regulate ovulation in an asymmetric manner during the estrous cycle. The aims of the present study were to analyze the effects of a temporal blockade of mAChRs on either side of the POA-AHA performed in diestrus-2 rats on ovulation, the levels of estradiol, follicle stimulating hormone (FSH) and luteinizing hormone (LH) and the mechanisms involved in changes in ovulation. METHODS: Cyclic rats on diestrus-2 day were anesthetized and randomly assigned to the following groups: 1) microinjection of 1 µl of saline or atropine solution (62.5 ng) in the left or right POA-AHA; 2) removal (unilateral ovariectomty, ULO) of the left (L-ULO) or right (R-ULO) ovary, and 3) rats microinjected with atropine into the left or right POA-AHA plus L-ULO or R-ULO. The ovulation rate and the number of ova shed were measured during the predicted estrus, as well as the levels of estradiol, FSH and LH during the predicted proestrus and the effects of injecting synthetic LH-releasing hormone (LHRH) or estradiol benzoate (EB). RESULTS: Atropine in the left POA-AHA decreased both the ovulation rate and estradiol and LH levels on the afternoon of proestrus, also LHRH or EB injection restored ovulation. L- or R-ULO resulted in a lower ovulation rate and smaller number of ova shed, and only injection of LHRH restored ovulation. EB injection at diestrus-2 restored ovulation in animals with L-ULO only. The levels of estradiol, FSH and LH in rats with L-ULO were higher than in animals with unilateral laparotomy. In the group microinjected with atropine in the left POA-AHA, ovulation was similar to that in ULO rats. In contrast, atropine in the right POA-AHA of ULO rats blocked ovulation, an action that was restored by either LHRH or EB injection. CONCLUSIONS: These results indicated that the removal of a single ovary at noon on diestrus-2 day perturbed the neuronal pathways regulating LH secretion, which was mediated by the muscarinic system connecting the right POA-AHA and the ovaries.


Asunto(s)
Núcleo Hipotalámico Anterior/metabolismo , Diestro/metabolismo , Estradiol/metabolismo , Hormona Folículo Estimulante/metabolismo , Hormona Luteinizante/metabolismo , Ovulación/metabolismo , Área Preóptica/metabolismo , Receptores Muscarínicos/metabolismo , Animales , Núcleo Hipotalámico Anterior/efectos de los fármacos , Atropina/farmacología , Anticonceptivos/farmacología , Diestro/efectos de los fármacos , Estradiol/análogos & derivados , Estradiol/farmacología , Femenino , Hormona Liberadora de Gonadotropina/farmacología , Hormona Luteinizante/efectos de los fármacos , Antagonistas Muscarínicos/farmacología , Ovariectomía , Ovario/efectos de los fármacos , Ovulación/efectos de los fármacos , Área Preóptica/efectos de los fármacos , Proestro/efectos de los fármacos , Proestro/metabolismo , Ratas , Receptores Muscarínicos/efectos de los fármacos
9.
Reprod Biol Endocrinol ; 14(1): 34, 2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27306649

RESUMEN

BACKGROUND: The suprachiasmatic nucleus (SCN) and the cholinergic system of various regions of the hypothalamus participate in the regulation of gonadotropin-releasing hormone (GnRH) and gonadotropin secretion, which are necessary for the occurrence of ovulation. In the present study, our goal was to analyse the effects of unilaterally blocking the muscarinic receptors in the SCN on ovulation and steroid secretion. METHODS: Cyclic rats were randomly allotted to one of the experimental groups. Groups of 8-14 rats were anaesthetized and microinjected with 0.3 µl of saline or a solution of atropine (62.5 ng in 0.3 µl of saline) into the left or right SCN at 09.00 or 19.00 h during diestrus-1 or on the proestrus day. The rats were euthanized on the predicted day of oestrus, and evaluated ovulation and levels of progesterone and oestradiol. Other groups of 10 rats were microinjected with atropine into the left or right SCNs at 09.00 h on the proestrus day, were euthanized eight h later, and luteinizing hormone (LH) was measured. RESULTS: At 09.00 or 19.00 h during diestrus-1, atropine microinjections into the SCNs on either side did not modify ovulation. The atropine microinjections performed at 09.00 h of proestrus into either side of the SCN blocked ovulation (right SCN: 1/9 ovulated vs. 9/10 in the saline group; left SCN: 8/14 ovulated vs. 10/10 in the saline group). The LH levels at 17.00 h in the rats that were microinjected with atropine at 09.00 h of proestrus were lower than those of the controls. In the non-ovulating atropine-treated rats, the injection of synthetic LH-releasing hormone (LHRH) restored ovulation. Atropine treatment at 19.00 h of proestrus on either side of the SCN did not modify ovulation, while the progesterone and oestradiol levels were lower. CONCLUSION: Based on the present results, we suggest that the cholinergic neural information arriving on either side of the SCN is necessary for the pre-ovulatory secretion of LH to induce ovulation. Additionally, the regulation of progesterone and oestradiol secretion by the cholinergic innervation of the SCN varies with the time of day, the day of the cycle, and the affected SCN.


Asunto(s)
Atropina/farmacología , Hormona Luteinizante/sangre , Antagonistas Muscarínicos/farmacología , Ovulación/efectos de los fármacos , Proestro/efectos de los fármacos , Núcleo Supraquiasmático/efectos de los fármacos , Animales , Femenino , Ovario/efectos de los fármacos , Proestro/metabolismo , Ratas , Núcleo Supraquiasmático/metabolismo
10.
Nitric Oxide ; 53: 45-53, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26778278

RESUMEN

An ex-vivo Coeliac Ganglion-Superior Ovarian Nerve-Ovary (CG-SON-O) system from virgin rats in the first proestrous was used to test whether cholinergic stimulation of CG affects oxidative status and steroidogenesis in the ovary. The CG and the O were placed in separate buffered-compartments, connected by the SON, and the CG was stimulated by acetylcholine (Ach). To test a possible role of nitric oxide (NO) in the ovarian response to cholinergic stimulation of CG, aminoguanidine (AG) - an inhibitor of inducible-NO synthase was added to the O compartment. After 180 min incubation, the oxidative status was assessed in O whereas nitrite and steroidogenesis were assessed at 30, 120 and 180 min. Ach in CG decreased the total antioxidant capacity, but increased NO production and protein carbonization in O. Ach stimulation of CG increased estradiol, but decreased progesterone release in O by reducing the mRNAs related to their synthesis and degradation. The addition of AG to the O compartment caused an opposite effect, which was more pronounced in the presence of Ach in the CG compartment than in its absence. These results show that the stimulation of the extrinsic-cholinergic innervation of the O increases the concentration of NO, causes oxidative stress and modulates steroidogenesis in the first rat proestrous.


Asunto(s)
Colinérgicos/farmacología , Ganglios Simpáticos/efectos de los fármacos , Óxido Nítrico/metabolismo , Ovario/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Proestro , Progesterona/biosíntesis , Animales , Femenino , Ganglios Simpáticos/fisiología , Óxido Nítrico/biosíntesis , Ovario/inervación , Ovario/metabolismo , Proestro/efectos de los fármacos , Progesterona/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Physiol Behav ; 152(Pt A): 41-6, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26335038

RESUMEN

Life history theory predicts that selectivity for mates generally declines as females age. We previously demonstrated this phenomenon in Syrian hamsters (Mesocricetus auratus), in that older females showed reduced preference for dominant over subordinate males. To test the hypothesis that decreased reproductive quality due to aging reduces mate preference, we decoupled reproductive and chronological age by treating young female hamsters with 4-vinylcyclohexene diepoxide (VCD), which destroys ovarian follicles and functionally accelerates ovarian follicle depletion without compromising the general health of rodents. In this study, VCD effectively reduced follicle numbers in young Syrian hamsters. VCD-treated and control females were allowed to choose between a dominant and a subordinate male in a Y-maze on the day of proestrus. Both VCD-treated and control females demonstrated preference for the dominant male by leaving a greater proportion of vaginal scent marks near him, which is a behavior that females display when soliciting prospective mates. However, there was no effect of treatment on the proportion of vaginal scent marks left for the dominant male. Furthermore, ovarian follicle numbers were not significantly correlated with any behaviors in either group. We conclude that accelerated ovarian follicle depletion does not reduce mate preference in young female hamsters.


Asunto(s)
Dominación-Subordinación , Preferencia en el Apareamiento Animal/fisiología , Mesocricetus/fisiología , Mesocricetus/psicología , Folículo Ovárico/fisiología , Envejecimiento/fisiología , Envejecimiento/psicología , Animales , Femenino , Masculino , Preferencia en el Apareamiento Animal/efectos de los fármacos , Aprendizaje por Laberinto , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Folículo Ovárico/efectos de los fármacos , Proestro/efectos de los fármacos , Proestro/fisiología , Pruebas Psicológicas
12.
Biol Reprod ; 93(1): 15, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25995272

RESUMEN

Variations in mRNA levels and sources of metastin/kisspeptin, neurokinin B (NKB), dynorphin, and kisspeptin receptor GPR54 were examined in the ovaries of cycling rats. Kisspeptin and dynorphin mRNAs dramatically increased at 2000 h of the proestrous day. NKB mRNA also increased, but the peak was delayed by 6 h. GPR54 mRNA declined inversely with kisspeptin. Whole-ovary expressions of kisspeptin and dynorphin mRNAs, but not of NKB mRNA, were augmented by the administration of human chorionic gonadotropin (hCG). By means of laser-capture microdissection, kisspeptin mRNA was shown mostly in follicles at 2000 h of proestrus, whereas NKB and dynorphin were expressed mainly in interstitial tissues. GPR54 mRNA was detected equally in follicles, corpora lutea, and interstitial tissues. The hCG stimulated the follicular expression of kisspeptin and interstitial tissue expression of dynorphin mRNA. In primary cultures of granulosa cells prepared from equine chorionic gonadotropin-pretreated immature rats, hCG stimulated the expression of kisspeptin, dynorphin, and NKB mRNAs. Distortion of the corpus luteum and surrounding tissue borders was sometimes seen after intra-ovarian bursa administration of kisspeptin antagonist p234 for 3 days from proestrus. Progesterone production stimulated by hCG in granulosa cell culture was suppressed by p234. These data demonstrate that significant amounts of kisspeptin are synthesized in granulosa cells and dynorphin in interstitial tissues, in response to the proestrous luteinizing hormone surge, whereas granulosa cells also contain dynorphin and NKB, suggesting at least a role for kisspeptin in the luteinization of granulosa cells.


Asunto(s)
Regulación de la Expresión Génica , Células de la Granulosa/metabolismo , Kisspeptinas/metabolismo , Luteinización/metabolismo , Hormona Luteinizante/metabolismo , Proestro/metabolismo , Animales , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/metabolismo , Dinorfinas/genética , Dinorfinas/metabolismo , Femenino , Células de la Granulosa/efectos de los fármacos , Kisspeptinas/genética , Luteinización/genética , Péptidos/farmacología , Proestro/efectos de los fármacos , Proestro/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1
13.
Endocrinology ; 156(8): 2999-3011, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25993523

RESUMEN

The kisspeptin (Kp) neurons in the anteroventral periventricular nucleus (AVPV) are essential for the preovulatory LH surge, which is gated by circulating estradiol (E2) and the time of day. We investigated whether AVPV Kp neurons in intact female mice may be the site in which both E2 and daily signals are integrated and whether these neurons may host a circadian oscillator involved in the timed LH surge. In the afternoon of proestrous day, Kp immunoreactivity displayed a marked and transient decrease 2 hours before the LH surge. In contrast, Kp content was stable throughout the day of diestrus, when LH levels are constantly low. AVPV Kp neurons expressed the clock protein period 1 (PER1) with a daily rhythm that is phase delayed compared with the PER1 rhythm measured in the main clock of the suprachiasmatic nuclei (SCN). PER1 rhythm in the AVPV, but not in the SCN, exhibited a significant phase delay of 2.8 hours in diestrus as compared with proestrus. Isolated Kp-expressing AVPV explants from PER2::LUCIFERASE mice displayed sustained circadian oscillations of bioluminescence with a circadian period (23.2 h) significantly shorter than that of SCN explants (24.5 h). Furthermore, in AVPV explants incubated with E2 (10 nM to 1 µM), the circadian period was lengthened by 1 hour, whereas the SCN clock remained unaltered. In conclusion, these findings indicate that AVPV Kp neurons display an E2-dependent daily rhythm, which may possibly be driven by an intrinsic circadian clock acting in combination with the SCN timing signal.


Asunto(s)
Núcleo Hipotalámico Anterior/metabolismo , Relojes Circadianos/genética , Kisspeptinas/genética , Animales , Diestro/efectos de los fármacos , Diestro/genética , Diestro/metabolismo , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proestro/efectos de los fármacos , Proestro/genética , Proestro/metabolismo , Núcleo Supraquiasmático/metabolismo
14.
Endocrine ; 49(3): 809-19, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25586874

RESUMEN

We analyzed the effects of chemically blocking type 1 muscarinic receptors (M1R) on either the left or right ovary on ovulation rate, number of ova shed and steroid hormones levels. M1R were unilaterally blocked in ovary with the M1R selective antagonist pirenzepine (PZP). PZP was delivered into the bursa ovarica of the left or right ovary of adult rats at 13:00 h on proestrus day. PZP treatment in the left but not in the right ovary blocked ovulation. PZP did not modify the number of ova shed, nor progesterone or 17ß-estradiol serum levels. The surge of luteinizing hormone levels was diminished while that of follicle-stimulating hormone did not change in animals treated with PZP in the left ovary. Interestingly, treatment with either synthetic luteinizing hormone-releasing hormone or human chorionic gonadotropin 1 h after PZP administration in the left ovary restored ovulation in both ovaries. The presence of M1R protein in the theca cells of the ovarian follicles as well as in cells of the corpus luteum was detected on proestrus day. These results suggest that M1R activation in the left ovary is required for pre-ovulatory gonadotropin-releasing hormone (GnRH) secretion and ovulation. Furthermore, these results also suggest that M1R in the left ovary might be regulating ovulation asymmetrically through a stimulatory neural signal relayed to the hypothalamus via the vagus nerve to induce the GnRH secretion which then triggers ovulation.


Asunto(s)
Ovario/metabolismo , Ovulación/fisiología , Proestro/fisiología , Receptor Muscarínico M1/metabolismo , Animales , Estradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Hormona Liberadora de Gonadotropina/farmacología , Hipotálamo/metabolismo , Hormona Luteinizante/sangre , Antagonistas Muscarínicos/farmacología , Folículo Ovárico/metabolismo , Ovulación/efectos de los fármacos , Pirenzepina/farmacología , Proestro/efectos de los fármacos , Progesterona/sangre , Ratas , Receptor Muscarínico M1/efectos de los fármacos , Células Tecales , Vagotomía
15.
Behav Brain Res ; 282: 95-102, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25591479

RESUMEN

Regularly cycling Fischer female rats were treated with either a low (5mg/kg) or high (5mg/RAT; approximately 30mg/kg) dose of the antiprogestin, RU486, before the morning of proestrus or on the morning of proestrus. The emergence of sexual behavior after treatment with RU486 was examined in a mating test with a sexually active male rat. Lordosis behavior was remarkably resistant to the effects of RU486. Only the high dose of RU486 given the evening before proestrus, approximately 22h before mating, reduced lordosis behavior. Independent of dose or time of treatment, proceptivity was reduced and resistance to the male's attempts to mount was increased by RU486 treatment. In addition, the effect of a 5min restraint stress on sexual behavior was examined. In contrast to the relative resistance of lordosis behavior of unrestrained rats to RU486 treatment, RU486 treated rats showed a significant decline in lordosis behavior after restraint. These findings allow the suggestion that the emergence of lordosis behavior is relatively resistant to the antiprogestin while the maintenance of lordosis behavior after restraint may require participation of intracellular progesterone receptors.


Asunto(s)
Antagonistas de Hormonas/farmacología , Mifepristona/farmacología , Conducta Sexual Animal/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Postura , Proestro/efectos de los fármacos , Ratas , Ratas Endogámicas , Restricción Física/psicología
16.
Physiol Behav ; 131: 7-16, 2014 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-24727338

RESUMEN

Exposure of proestrous Syrian hamsters to a new room, cage, and novel running wheel blocks the luteinizing hormone (LH) surge until the next day in ~75% of hamsters [1]. The studies described here tested the hypotheses that 1) exercise and/or 2) orexinergic neurotransmission mediate novel wheel blockade of the LH surge and circadian phase advances. Female hamsters were exposed to a 14L:10D photoperiod and activity rhythms were monitored with infra-red detectors. In Expt. 1, to test the effect of exercise, hamsters received jugular cannulae and on the next day, proestrus (Day 1), shortly before zeitgeber time 5 (ZT 5, 7h before lights-off) the hamsters were transported to the laboratory. After obtaining a blood sample at ZT 5, the hamsters were transferred to a new cage with a novel wheel that was either freely rotating (unlocked), or locked until ZT 9, and exposed to constant darkness (DD). Blood samples were collected hourly for 2days from ZT 5-11 under red light for determination of plasma LH levels by radioimmunoassay. Running rhythms were monitored continuously for the next 10-14days. The locked wheels were as effective as unlocked wheels in blocking LH surges (no Day 1 LH surge in 6/9 versus 8/8 hamsters, P>0.05) and phase advances in the activity rhythms did not differ between the groups (P=0.28), suggesting that intense exercise is not essential for novel wheel blockade and phase advance of the proestrous LH surge. Expt. 2 tested whether orexin neurotransmission is essential for these effects. Hamsters were treated the same as those in Expt. 1 except that they were injected (i.p.) at ZT 4.5 and 5 with either the orexin 1 receptor antagonist SB334867 (15mg/kg per injection) or vehicle (25% DMSO in 2-hydroxypropyl-beta-cyclodextrin (HCD)). SB-334867 inhibited novel wheel blockade of the LH surge (surges blocked in 2/6 SB334867-injected animals versus 16/18 vehicle-injected animals, P<0.02) and also inhibited wheel running and circadian phase shifts, indicating that activation of orexin 1 receptors is necessary for these effects. Expt. 3 tested the hypothesis that novel wheel exposure activates orexin neurons. Proestrous hamsters were transferred at ZT 5 to a nearby room within the animal facility and were exposed to a new cage with a locked or unlocked novel wheel or left in their home cages. At ZT 8, the hamsters were anesthetized, blood was withdrawn, they were perfused with fixative and brains were removed for immunohistochemical localization of Fos, GnRH, and orexin. Exposure to a wheel, whether locked or unlocked, suppressed circulating LH concentrations at ZT 8, decreased the proportion of Fos-activated GnRH neurons, and increased Fos-immunoreactive orexin cells. Unlocked wheels had greater effects than locked wheels on all three endpoints. Thus in a familiar environment, exercise potentiated the effect of the novel wheel on Fos expression because a locked wheel was not a sufficient stimulus to block the LH surge. In conclusion, these studies indicate that novel wheel exposure activates orexin neurons and that blockade of orexin 1 receptors prevents novel wheel blockade of the LH surge. These findings are consistent with a role for both exercise and arousal in mediating novel wheel blockade of the LH surge.


Asunto(s)
Nivel de Alerta/fisiología , Ritmo Circadiano/fisiología , Hormona Luteinizante/metabolismo , Actividad Motora/fisiología , Animales , Benzoxazoles/farmacología , Fármacos del Sistema Nervioso Central/farmacología , Ritmo Circadiano/efectos de los fármacos , Oscuridad , Estro/efectos de los fármacos , Estro/fisiología , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Vivienda para Animales , Mesocricetus , Actividad Motora/efectos de los fármacos , Naftiridinas , Neuronas/efectos de los fármacos , Neuronas/fisiología , Antagonistas de los Receptores de Orexina , Receptores de Orexina/metabolismo , Fotoperiodo , Proestro/efectos de los fármacos , Proestro/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Urea/análogos & derivados , Urea/farmacología
17.
Pharmacol Biochem Behav ; 122: 20-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24650589

RESUMEN

The capacity to form progesterone (P4)'s 5α-reduced metabolite, 5α-pregnan-3α-ol-20-one (3α,5α-THP; a.k.a. allopregnanolone), in the brain may be related to facilitation of lordosis among estrogen-primed (E2) mice. We investigated this idea further by comparing effects of endogenous and exogenous progestogens in mice that are deficient in the Type One 5α-reductase enzyme (5α-reductase knockout mice; 5α-RKO), and their wildtype counterparts for sexual behavior. Comparisons were made following administration of progestogens that are expected to increase 3α,5α-THP or not. Sexual receptivity of 5α-RKO mice and their wildtype counterparts was examined when mice were naturally-cycling (Experiment 1); ovariectomized (OVX), E2-primed (10 µg, subcutaneous; SC) and administered P4 (0, 125, 250, or 500 µg SC; Experiment 2); and OVX, E2-primed and administered P4, medroxyprogesterone acetate (MPA, 4 mg/kg, SC, which does not convert to 3α,5α-THP) or 3α,5α-THP (4 mg/kg, SC; Experiment 3). The percentage of mounts that elicited lordosis (lordosis quotient) or aggression/rejection behavior (aggression quotient), as well as the quality of lordosis (lordosis rating), was scored. Wildtype, but not 5α-RKO, mice in behavioral estrus demonstrated significantly greater lordosis quotients and lordosis ratings, but similar aggression quotients, compared to their diestrous counterparts. Among OVX and E2-primed mice, P4 facilitated lordosis of wildtype, but not 5α-RKO, mice. MPA neither facilitated lordosis of wildtype, nor 5α-RKO mice. 3α,5α-THP administered to wildtype or 5α-RKO mice increased lordosis quotients and lordosis ratings and decreased aggression quotients. 3α,5α-THP levels in the midbrain, one brain region important for sexual behavior, were increased during behavioral estrus, with P4 administered to WT, but not 5α-RKO mice, and 3α,5α-THP administered to WT and 5α-RKO mice. MPA did not increase 3α,5α-THP. Thus, deletion of Type One 5α-reductase among female mice may attenuate reproductive responding during the estrous cycle and after hormone-priming.


Asunto(s)
Colestenona 5 alfa-Reductasa/deficiencia , Proestro/metabolismo , Progesterona/farmacología , Reproducción/fisiología , Conducta Sexual Animal/fisiología , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/deficiencia , Animales , Femenino , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Noqueados , Proestro/efectos de los fármacos , Reproducción/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos
18.
Peptides ; 52: 1-10, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24296042

RESUMEN

Women are at greater risk for major depression, PTSD, and other anxiety disorders. ERß-selective agonists for the treatment of these disorders are the focus of pharmacologic development and clinical testing. Estradiol and its metabolites contribute to the neuroprotective effects of this steroid class, particularly in men, due to local conversion of testosterone to estiradiol in key brain regions which are predisposed to neurodegenerative diseases. We have used young adult female Sprague-Dawley rats to assess the role of TRH and TRH-like peptides, with the general structure pGlu-X-Pro-NH2 where "X" can be any amino acid residue, as mediators of the neurobiochemical effects of estradiol. The neuroprotective TRH and TRH-like peptides are coreleased with excitotoxic glutamate by glutamatergic neurons which contribute importantly to the regulation of the estrus cycle. The levels of TRH and TRH-like peptides during proestrus and/or estrus in the 12 brain regions analyzed were significantly decreased (due to accelerated release) 106 times but increased only 25 times when compared to the corresponding levels during diestrus days 1 and 2. These changes, listed by brain region in the order of decreasing number of significant decreases (↓) and/or increases (↑), were: striatum (20↓,1↑), medulla oblongata (16↓,2↑), amygdala (14↓,1↑), cerebellum (13↓,1↑), hypothalamus (12↓,1↑), entorhinal cortex (6↓,6↑), posterior cingulate (10↓,1↑), frontal cortex (3↓,5↑), nucleus accumbens (5↓,3↑), hippocampus (5↓,2↑), anterior cingulate (2↓,1↑), and piriform cortex (1↑). In peripheral tissues the corresponding changes were: ovaries (23↓), uterus (16↓,1↑), adrenals (11↓,3↑), and pancreas (1↓,6↑). We conclude that these peptides may be downstream mediators of some of the therapeutic effects of estrogen.


Asunto(s)
Encéfalo/metabolismo , Estro/efectos de los fármacos , Ovario/metabolismo , Péptidos/farmacología , Proestro/efectos de los fármacos , Tirotropina/farmacología , Secuencias de Aminoácidos , Animales , Femenino , Humanos , Ratas , Ratas Sprague-Dawley
19.
PLoS One ; 8(10): e78178, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24124622

RESUMEN

The orexigenic peptide, ghrelin is known to influence function of GnRH neurons, however, the direct effects of the hormone upon these neurons have not been explored, yet. The present study was undertaken to reveal expression of growth hormone secretagogue receptor (GHS-R) in GnRH neurons and elucidate the mechanisms of ghrelin actions upon them. Ca(2+)-imaging revealed a ghrelin-triggered increase of the Ca(2+)-content in GT1-7 neurons kept in a steroid-free medium, which was abolished by GHS-R-antagonist JMV2959 (10 µM) suggesting direct action of ghrelin. Estradiol (1nM) eliminated the ghrelin-evoked rise of Ca(2+)-content, indicating the estradiol dependency of the process. Expression of GHS-R mRNA was then confirmed in GnRH-GFP neurons of transgenic mice by single cell RT-PCR. Firing rate and burst frequency of GnRH-GFP neurons were lower in metestrous than proestrous mice. Ghrelin (40 nM-4 µM) administration resulted in a decreased firing rate and burst frequency of GnRH neurons in metestrous, but not in proestrous mice. Ghrelin also decreased the firing rate of GnRH neurons in males. The ghrelin-evoked alterations of the firing parameters were prevented by JMV2959, supporting the receptor-specific actions of ghrelin on GnRH neurons. In metestrous mice, ghrelin decreased the frequency of GABAergic mPSCs in GnRH neurons. Effects of ghrelin were abolished by the cannabinoid receptor type-1 (CB1) antagonist AM251 (1µM) and the intracellularly applied DAG-lipase inhibitor THL (10 µM), indicating the involvement of retrograde endocannabinoid signaling. These findings demonstrate that ghrelin exerts direct regulatory effects on GnRH neurons via GHS-R, and modulates the firing of GnRH neurons in an ovarian-cycle and endocannabinoid dependent manner.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Endocannabinoides/metabolismo , Ciclo Estral/efectos de los fármacos , Ghrelina/farmacología , Hormona Liberadora de Gonadotropina/biosíntesis , Neuronas/efectos de los fármacos , Neuronas/fisiología , Transducción de Señal/efectos de los fármacos , Animales , Encéfalo/metabolismo , Calcio/metabolismo , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/fisiología , Expresión Génica , Masculino , Metestro/efectos de los fármacos , Ratones , Proestro/efectos de los fármacos , ARN Mensajero/genética , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo , Potenciales Sinápticos/efectos de los fármacos
20.
Biol Reprod ; 89(4): 98, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24006283

RESUMEN

Vanin-2 (VNN2) is known to be involved in inflammation and leukocyte migration, but its regulation in follicles remains unknown. The objectives of this work were to study the regulation of VNN2 transcripts in bovine follicles prior to ovulation and to characterize the control of its expression in bovine granulosa cells. VNN2 expression was studied using total RNA extracted from granulosa cells of small follicles (2-4 mm in diameter), dominant follicles obtained on Day 5 of the estrous cycle, ovulatory follicles obtained 0-24 h after human chorionic gonadotropin (hCG), and corpora lutea on Day 5 of the cycle. The results from RT-PCR analyses showed that levels of VNN2 mRNA were high in ovulatory follicles 24 h post-hCG but low in the other tissues. In ovulatory follicles, levels of VNN2 mRNA were low at 0 h but significantly up-regulated 12-24 h post-hCG. To determine factors controlling VNN2 gene expression, established primary cultures of granulosa cells isolated from bovine dominant follicles were used. Treatment with forskolin elevated VNN2 mRNA expression as observed in vivo. Mutation studies identified the minimal region conferring basal and forskolin-stimulated VNN2 promoter activities, which were dependent on chicken ovalbumin upstream promoter-transcription factor (COUP-TF), GATA, and Ebox cis-elements. Electrophoretic mobility shift assays identified COUP-TF, GATA4, and upstream stimulating factor proteins as key factors interacting with these elements. Chromatin immunoprecipitation assays confirmed basal and forskolin-induced interactions between these proteins and the VNN2 promoter in bovine granulosa cell cultures. VNN2 promoter activity and mRNA expression were markedly stimulated by forskolin and overexpression of the catalytic subunit of PKA, but inhibited by PKA and ERK1/2 inhibitors. Collectively, the findings from this study describe for the first time the gonadotropin/forskolin-dependent up-regulation of VNN2 transcripts in granulosa cells of preovulatory follicles and provide insights into some of the molecular bases of VNN2 gene expression in follicular cells.


Asunto(s)
Amidohidrolasas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Folículo Ovárico/metabolismo , Proestro , Regiones Promotoras Genéticas , Transcripción Genética , Regulación hacia Arriba , Amidohidrolasas/biosíntesis , Amidohidrolasas/genética , Animales , Bovinos , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Células Cultivadas , Gonadotropina Coriónica/farmacología , Colforsina/farmacología , Cuerpo Lúteo/citología , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/metabolismo , Activadores de Enzimas/farmacología , Femenino , Fármacos para la Fertilidad Femenina/farmacología , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mutación , Folículo Ovárico/citología , Folículo Ovárico/efectos de los fármacos , Proestro/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Elementos de Respuesta/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA