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

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neuroendocrinol ; 34(1): e13071, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34904297

RESUMO

Neural circuits in female rats are exposed to sequential estradiol and progesterone to regulate the release of luteinizing hormone (LH) and ultimately ovulation. Estradiol induces progesterone receptors (PGRs) in anteroventral periventricular nucleus (AVPV) kisspeptin neurons, and as estradiol reaches peak concentrations, neuroprogesterone (neuroP) synthesis is induced in hypothalamic astrocytes. This local neuroP signals to PGRs expressed in kisspeptin neurons to trigger the LH surge. We tested the hypothesis that neuroP-PGR signaling through Src family kinase (Src) underlies the LH surge. As observed in vitro, PGR and Src are co-expressed in AVPV neurons. Estradiol treatment increased the number of PGR immunopositive cells and PGR and Src colocalization. Furthermore, estradiol treatment increased the number of AVPV cells that had extranuclear PGR and Src in close proximity (< 40 nm). Infusion of the Src inhibitor (PP2) into the AVPV region of ovariectomized/adrenalectomized (ovx/adx) rats attenuated the LH surge in trunk blood collected 53 h post-estradiol (50 µg) injection that induced neuroP synthesis. Although PP2 reduced the LH surge in estradiol benzoate treated ovx/adx rats, activation of either AVPV PGR or Src in 2 µg estradiol-primed animals significantly elevated LH concentrations compared to dimethyl sulfoxide infused rats. Finally, antagonism of either AVPV PGR or Src blocked the ability of PGR or Src activation to induce an LH surge in estradiol-primed ovx/adx rats. These results indicate that neuroP, which triggers the LH surge, signals through an extranuclear PGR-Src signaling pathway.


Assuntos
Hormônio Luteinizante/metabolismo , Neurônios/metabolismo , Progesterona/metabolismo , Receptores de Progesterona/fisiologia , Quinases da Família src/fisiologia , Animais , Feminino , Hipotálamo/metabolismo , Ovulação/sangue , Ovulação/metabolismo , Ratos , Ratos Long-Evans , Receptores de Progesterona/metabolismo , Transdução de Sinais/fisiologia , Quinases da Família src/metabolismo
2.
Nat Rev Endocrinol ; 17(2): 83-96, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33288917

RESUMO

The onset of puberty and the female ovulatory cycle are important developmental milestones of the reproductive system. These processes are controlled by a tightly organized network of neurotransmitters and neuropeptides, as well as genetic, epigenetic and hormonal factors, which ultimately drive the pulsatile secretion of gonadotropin-releasing hormone. They also strongly depend on organizational processes that take place during fetal and early postnatal life. Therefore, exposure to environmental pollutants such as endocrine-disrupting chemicals (EDCs) during critical periods of development can result in altered brain development, delayed or advanced puberty and long-term reproductive consequences, such as impaired fertility. The gonads and peripheral organs are targets of EDCs, and research from the past few years suggests that the organization of the neuroendocrine control of reproduction is also sensitive to environmental cues and disruption. Among other mechanisms, EDCs interfere with the action of steroidal and non-steroidal receptors, and alter enzymatic, metabolic and epigenetic pathways during development. In this Review, we discuss the cellular and molecular consequences of perinatal exposure (mostly in rodents) to representative EDCs with a focus on the neuroendocrine control of reproduction, pubertal timing and the female ovulatory cycle.


Assuntos
Disruptores Endócrinos/farmacologia , Exposição Ambiental , Epigênese Genética/efeitos dos fármacos , Estradiol/metabolismo , Hormônio Liberador de Gonadotropina/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Movimento Celular , Metilação de DNA/efeitos dos fármacos , Retroalimentação Fisiológica/efeitos dos fármacos , Feminino , GABAérgicos/metabolismo , Células Germinativas/metabolismo , Ácido Glutâmico/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Código das Histonas/efeitos dos fármacos , Humanos , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Masculino , Neurônios/metabolismo , Ovulação/efeitos dos fármacos , Ovulação/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal
3.
Curr Med Sci ; 39(2): 285-290, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31016523

RESUMO

In our previous study, we found that Shoutai pills could improve the embryo implantation rate as well as the levels of estrogen, progesterone and estrogen receptor in rats with stimulated ovulation. However, the mechanism is not clear. This study was designed to investigate the effect of Shoutai pills on the levels of Th1 and Th2 cytokines in rats with stimulated ovulation and the mechanism. The rat model of stimulated ovulation was established by combined injection of pregnant mare serum gonadotropin (PMSG) and human chorionic gonadotropin (HCG). Then the rats were randomly divided into model group (M), Shoutai pills group (S), progesterone group (P) and normal group (N). All the pregnant rats were treated from the first day. The S and P groups were administrated with gavage of Shoutai pills and injection of progesterone respectively, and N and M groups were given the same volume of normal saline and distilled water respectively. After treatment for 7 days, the animals were executed for serum and uterine tissues. The ELISA method was adopted to detect the contents of Th1 cytokines [interferon-γ (INF-γ), interleukin-2 (IL-2)] and Th2 cytokines (IL-4, IL-6, IL-10). The expression of leukemia inhibitory factor (LIF) and leukemia inhibitory factor receptor (LIFR) was detected by Western blotting and real-time PCR. As compared with N group, the expression levels of IFN-γ and IL-2 in M group were significantly increased, and those of IL-4, IL-6, IL-10, LIF and LIFR were significantly decreased (P<0.05). As compared with M group, the levels of IL-4, IL-6, IL-10, LIF and LIFR in S group were significantly increased (P<0.05), and those of IFN-γ and IL-2 were significantly decreased (P<0.05). It was suggested that Shoutai pills can increase the levels of IL-4, IL-6, IL-10, LIF and LIFR as well as reduce the levels of INF-γ and IL-2 in rats with stimulated ovulation. The Shoutai pills may improve endometrial receptivity and promote embryo implantation by maintaining the balance of Th1/Th2 cytokines.


Assuntos
Citocinas/sangue , Citocinas/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Endométrio/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Animais , Endométrio/metabolismo , Feminino , Fator Inibidor de Leucemia/metabolismo , Masculino , Medicina Tradicional Chinesa/métodos , Ovulação/efeitos dos fármacos , Ovulação/metabolismo , Indução da Ovulação/métodos , Progesterona , Ratos , Ratos Wistar , Células Th1/metabolismo , Células Th2/metabolismo
4.
Gynecol Endocrinol ; 32(9): 690-695, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27595157

RESUMO

Inositol is an organic compound of high biological importance that is widely distributed in nature. It belongs to the sugar family and is mainly represented by its two dominant stereoisomers: myo-inositol and D-chiro-inositol that are found in the organism in the physiological serum ratio 40:1. Inositol and its derivatives are important components of the structural phospholipids of the cell membranes and are precursors of the second messengers of many metabolic pathways. A high concentration of myoinositol is found in the follicular fluid and in semen. Inositol deficiency and the impairment of the inositol-dependent pathways may play an important role in the pathogenesis of insulin resistance and hypothyroidism. The results of the research also point out the potential beneficial role of inositol supplementation in polycystic ovarian syndrome and in the context of assisted reproduction technologies and in vitro fertilization. The main aim of the article is to overview the major inositol-dependent metabolic pathways and to discuss its importance for reproduction.


Assuntos
Inositol/fisiologia , Resistência à Insulina/fisiologia , Ovulação/metabolismo , Síndrome do Ovário Policístico/metabolismo , Técnicas de Reprodução Assistida , Feminino , Humanos , Inositol/deficiência , Inositol/metabolismo , Inositol/uso terapêutico
5.
Endocrinol Metab Clin North Am ; 44(3): 485-96, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26316238

RESUMO

In women, age-related changes in ovarian function begin in the mid-30s with decreased fertility and compensatory hormonal changes in the hypothalamus-pituitary-gonadal axis that maintain follicle development and estrogen secretion in the face of a waning pool of ovarian follicles. The menopause transition is characterized by marked variability in follicle development, ovulation, bleeding patterns, and symptoms of hyper- and hypoestrogenism. The menopause, which is clinically defined by the last menstrual period, is followed by the consistent absence of ovarian secretion of estradiol.


Assuntos
Menopausa/metabolismo , Ciclo Menstrual/metabolismo , Hormônio Antimülleriano/sangue , Feminino , Humanos , Hipotálamo/metabolismo , Inibinas/sangue , Menopausa/sangue , Ciclo Menstrual/sangue , Folículo Ovariano/metabolismo , Ovulação/sangue , Ovulação/metabolismo , Hipófise/metabolismo
6.
J Neuroendocrinol ; 26(11): 776-84, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25040132

RESUMO

Neurokinin B (NKB) is essential for human reproduction and has been shown to stimulate luteinising hormone (LH) secretion in several species, including sheep. Ewes express the neurokinin-3 receptor (NK3R) in the retrochiasmatic area (RCh) and there is one report that placement of senktide, an NK3R agonist, therein stimulates LH secretion that resembles an LH surge in ewes. In the present study, we first confirmed that local administration of senktide to the RCh produced a surge-like increase in LH secretion, and then tested the effects of this agonist in two other areas implicated in the control of LH secretion and where NK3R is found in high abundance: the preoptic area (POA) and arcuate nucleus (ARC). Bilateral microimplants containing senktide induced a dramatic surge-like increase in LH when given in the POA similar to that seen with RCh treatment. By contrast, senktide treatment in the ARC resulted in a much smaller but significant increase in LH concentrations suggestive of an effect on tonic secretion. The possible role of POA and RCh NK3R activation in the LH surge was next tested by treating ewes with SB222200, an NK3R antagonist, in each area during an oestradiol-induced LH surge. SB222200 in the RCh, but not in the POA, reduced the LH surge amplitude by approximately 40% compared to controls, indicating that NK3R activation in the former region is essential for full expression of the pre-ovulatory LH surge. Based on these data, we propose that the actions of NKB in the RCh are an important component of the pre-ovulatory LH surge in ewes.


Assuntos
Hipotálamo/efeitos dos fármacos , Hormônio Luteinizante/sangue , Ovulação/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores da Neurocinina-3/agonistas , Substância P/análogos & derivados , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Hipotálamo/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ovulação/metabolismo , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Quinolinas/farmacologia , Receptores da Neurocinina-3/antagonistas & inibidores , Receptores da Neurocinina-3/metabolismo , Ovinos , Substância P/farmacologia
7.
Am J Physiol Endocrinol Metab ; 305(6): E717-26, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23880317

RESUMO

Homozygous androgen receptor (AR)-knockout (ARKO) female mice are subfertile due to both intra- and extraovarian (neuroendocrine) defects as defined by ovary transplantation. Using ARKO mice, this study set out to reveal the precise AR-regulated pathways required for optimal androgen-regulated ovulation and fertility. ARKO females exhibit deficient neuroendocrine negative feedback, with a reduced serum luteinizing hormone (LH) response to ovariectomy (OVX) (P < 0.01). Positive feedback is also altered as intact ARKO females, at late proestrus, exhibit an often mistimed endogenous ovulatory LH surge. Furthermore, at late proestrus, intact ARKO females display diminished preovulatory serum estradiol (E2; P < 0.01) and LH (P < 0.05) surge levels and reduced Kiss1 mRNA expression in the anteroventral periventricular nucleus (P < 0.01) compared with controls. However, this reduced ovulatory LH response in intact ARKO females can be rescued by OVX and E2 priming or treatment with endogenous GnRH. These findings reveal that AR regulates the negative feedback response to E2, E2-positive feedback is compromised in ARKO mice, and AR-regulated negative and positive steroidal feedback pathways impact on intrahypothalamic control of the kisspeptin/GnRH/LH cascade. In addition, intraovarian AR-regulated pathways controlling antral to preovulatory follicle dynamics are disrupted because adult ARKO ovaries collected at proestrus have small antral follicles with reduced oocyte/follicle diameter ratios (P < 0.01) and increased proportions of unhealthy large antral follicles (P < 0.05) compared with controls. As a consequence of aberrant follicular growth patterns, proestrus ARKO ovaries also exhibit fewer preovulatory follicle (P < 0.05) and corpora lutea numbers (P < 0.01). However, embryo development to the blastocyst stage is unchanged in ARKO females, and hence, the subfertility is a consequence of reduced ovulations and not altered embryo quality. These findings reveal that the AR has a functional role in neuroendocrine regulation and timing of the ovulatory LH surge as well as antral/preovulatory follicle development.


Assuntos
Hipotálamo/metabolismo , Infertilidade Feminina/metabolismo , Ovário/metabolismo , Ovulação/metabolismo , Receptores Androgênicos/metabolismo , Animais , Corpo Lúteo/metabolismo , Estradiol/sangue , Ciclo Estral/sangue , Ciclo Estral/genética , Ciclo Estral/metabolismo , Feminino , Hipotálamo/fisiopatologia , Infertilidade Feminina/genética , Infertilidade Feminina/fisiopatologia , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormônio Luteinizante/sangue , Hormônio Luteinizante/metabolismo , Camundongos , Camundongos Knockout , Folículo Ovariano/metabolismo , Ovário/fisiopatologia , Ovulação/sangue , Ovulação/genética , Receptores Androgênicos/genética
8.
Theriogenology ; 78(1): 12-27, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22464816

RESUMO

Supplementary fat positively influences reproductive performance in dairy cattle, although the mechanisms involved are not clearly defined. Our objective was to determine the effects of four different fat supplements on follicle development, plasma steroid hormone concentrations and prostaglandin (PG) synthesis in lactating dairy cattle. Forty-eight early lactation Holstein-Friesian cows (21 primiparous, 27 multiparous) were used in a completely randomized block design. Cows were fed the same basal TMR diet and received one of four fat supplements: (i) palmitic acid (18:0 fatty acid; Control), (ii) flaxseed (rich in 18:3 n-3 fatty acid; Flax), (iii) conjugated linoleic acid (a mixture of cis-9, trans-11 and trans-10, cis-12 isomers; CLA), and (iv) fish oil (rich in 20:5 and 22:6 n-3 fatty acids; FO). All lipid supplements were formulated to be isolipidic; palmitic acid was added as necessary to provide a total lipid supplement intake of 500 g/day. Cows were synchronized to be in estrus on Day 15 of dietary treatment. All antral follicles were counted, and dominant follicles, subordinate follicles and corpora lutea were measured daily via transrectal ovarian ultrasonography for one complete estrous cycle. Blood samples were collected daily, and selected samples were analyzed for progesterone, estradiol, insulin-like growth factor-1, insulin, cholesterol and non-esterified fatty acids. Estrus was synchronized a second time, and liver and endometrial biopsies were collected on Day 7 of the estrous cycle. Gene expression was evaluated for a number of genes involved in prostaglandin synthesis (endometrium) and fatty acid uptake and utilization (liver). Fat supplementation had little effect on follicle development. Cows receiving supplementary n-3 fatty acids had lesser plasma progesterone (P4) and smaller corpora lutea than cows receiving the CLA or Control supplements. Effects of fat supplementation on the endometrial expression of genes involved in PG synthesis were minor. Hepatic expression of SREBF1, ASCL1 and FABP1 was reduced by FO supplementation. Reduced plasma P4 in n-3 supplemented cows may lead to a suboptimal uterine environment for embryo development and hence reduced fertility compared to cows receiving the control or CLA supplements.


Assuntos
Bovinos , Gorduras na Dieta/farmacologia , Suplementos Nutricionais , Lactação/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Ração Animal/provisão & distribuição , Animais , Bovinos/sangue , Bovinos/genética , Bovinos/metabolismo , Indústria de Laticínios , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/provisão & distribuição , Suplementos Nutricionais/estatística & dados numéricos , Eficiência/efeitos dos fármacos , Ácidos Graxos Insaturados/administração & dosagem , Ácidos Graxos Insaturados/farmacologia , Feminino , Lactação/genética , Lactação/fisiologia , Ácidos Linoleicos Conjugados/administração & dosagem , Ácidos Linoleicos Conjugados/farmacologia , Óleo de Semente do Linho/administração & dosagem , Óleo de Semente do Linho/farmacologia , Leite/metabolismo , Ovulação/sangue , Ovulação/efeitos dos fármacos , Ovulação/genética , Ovulação/metabolismo , Reprodução/genética , Reprodução/fisiologia
9.
J Reprod Dev ; 58(1): 10-6, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22450279

RESUMO

During recent decades, milk production per cow has increased drastically due to improved management, nutrition, and genetic selection; however, the reproductive performance of high-producing dairy cows has been declining. One of the factors responsible for this low reproductive performance is negative energy balance (NEB). NEB affects the onset of first ovulation in early postpartum cows. It is generally accepted that early first ovulation positively relates to the resumption of normal ovarian function, first service, and conception rate in dairy cows. Hence, delayed first ovulation has a negative impact on subsequent fertility. The metabolic condition of cows in NEB shifts to catabolic metabolism, which in turn causes increased plasma growth hormone and non-esterified fatty acid concentrations and decreased plasma insulin-like growth factor-1, insulin, and glucose concentrations. On the other hand, plasma ß-carotene concentrations decrease throughout the dry period and reach their nadir in about the first week postpartum, and this change reflects energy balance during the peripartum period. ß-Carotene plays a role independently of vitamin A in the reproductive performance of dairy cows, and the positive relationship between supplemental ß-carotene and reproductive function has been demonstrated in many studies during the past decades. However, ß-carotene content in corn silage, which is a popular main feed in high-producing dairy cows, is very low. This review describes nutritional factors related to ovulation during the first follicular wave postpartum in dairy cows.


Assuntos
Lactação/metabolismo , Folículo Ovariano/metabolismo , Ovulação/metabolismo , Animais , Glicemia/metabolismo , Bovinos , Metabolismo Energético , Ácidos Graxos não Esterificados/sangue , Ácidos Graxos não Esterificados/metabolismo , Feminino , Fertilidade , Hormônio do Crescimento/sangue , Insulina/sangue , Fator de Crescimento Insulin-Like I/análise , Lactação/sangue , Ovulação/sangue , beta Caroteno/sangue
10.
Gynecol Endocrinol ; 28 Suppl 1: 9-13, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22283375

RESUMO

Modern methods of diagnosis have made the distinction between hypothalamic failure and ovarian failure routine. Failure of the orderly progression of hypothalamic gonadotrophin-releasing hormone (GnRH) → pituitary gonadotrophins → ovarian steroids and inhibin → hypothalamus/pituitary results in anovulation/amenorrhea. The hypothalamic connections that regulate the pattern and amplitude of GnRH pulses are plastic and respond to external/psychological conditions and internal/metabolic factors that may affect the hypothalamic substrate on which estrogen levels can act. We trace the neuroendocrine regulation of the ovarian cycle, concentrating on hypothalamic connections that underlie negative and positive feedback control of GnRH and the complementary role of the adenohypophysis. The main hormone regulating this "central axis" and the development of the endometrium is estradiol which is exported from the developing ovarian follicles and thereby closes the feedback loop with follicle development. Progesterone and inhibin are also involved. Neuroendocrine responses to internal and external factors can cause anovulation and amenorrhea. Generally, these are accompanied by abnormal negative feedback between estradiol and the gonadotrophins; coexistence of low estradiol and luteinizing hormone/follicle-stimulating hormone. There are three main causes: (1) genetic diseases that interfere with the migration of GnRH cells into the brain or result in misfolding of GnRH; (2) input from the brain that interrupts normal feedback (e.g. stress and weight loss amenorrhea); and (3) the effect of agents which alter central neurotransmission and hypothalamic function (e.g. elevated prolactin and psychotropic medications). All types of hypothalamic insufficiency result in insufficient stimulation of the ovaries. In addition to amenorrhea, this central alteration also results in other complications (downstream disease) that make hypothalamic amenorrhea of greater consequence than simply reproductive failure. Thus, there may be more at stake in the diagnosis and treatment of hypothalamic failure than brings the patient to her caregiver.


Assuntos
Sistemas Neurossecretores/fisiologia , Ovulação/fisiologia , Animais , Feminino , Gonadotropinas/sangue , Gonadotropinas/metabolismo , Humanos , Doenças Hipotalâmicas/complicações , Doenças Hipotalâmicas/fisiopatologia , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Ciclo Menstrual/sangue , Ciclo Menstrual/metabolismo , Ciclo Menstrual/fisiologia , Sistemas Neurossecretores/metabolismo , Ovulação/sangue , Ovulação/genética , Ovulação/metabolismo
11.
J Neuroendocrinol ; 24(1): 22-33, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21951227

RESUMO

Kisspeptins, encoded by the Kiss1 gene, and their canonical receptor, GPR54 (also termed Kiss1R), are unanimously recognised as essential regulators of puberty onset and gonadotrophin secretion. These key reproductive functions stem from the capacity of kisspeptins to stimulate gonadotrophin-releasing hormone (GnRH) secretion in the hypothalamus, where discrete populations of Kiss1 neurones have been identified. In rodents, two major groups of hypothalamic Kiss1 neurones exist: one present in the arcuate nucleus (ARC) and the other located in the anteroventral periventricular area (AVPV/RP3V). In recent years, numerous signals have been identified as putative modulators of the hypothalamic Kiss1 system. Among them, the prominent role of sex steroids as being important regulators of Kiss1 neurones has been documented in different species and developmental stages, such as early brain sex differentiation, puberty, adulthood and senescence. These regulatory actions are (mainly) conducted via oestrogen receptor (ER)α, which is expressed in almost all Kiss1 neurones, and likely involve both classical and nonclassical pathways. The regulatory effects of sex steroids are nucleus-specific. Thus, sex steroids inhibit the expression of Kiss1/kisspeptin at the ARC, as a mechanism to conduct their negative-feedback actions on gonadotrophin secretion. By contrast, oestrogens enhance Kiss1 expression at the AVPV/RP3V in rodents, suggesting the involvement of this population in the positive-feedback actions of oestradiol to generate the preovulatory surge of gonadotrophins. In addition, sex steroids have been shown to act post-transcriptionally, modulating GnRH/gonadotrophin responsiveness to kisspeptin. Finally, sex steroids also regulate the expression of co-transmitters of Kiss1 neurones, such as neurokinin B, whose mRNA content in the ARC fluctuates in parallel to that of Kiss1 in response to changes in the circulating levels of sex steroids, therefore suggesting the contribution of this neuropeptide in the feedback control of gonadotrophin secretion. In sum, compelling experimental evidence obtained in different mammalian (and non-mammalian) species, including primates, demonstrates that sex steroids are essential regulators of hypothalamic Kiss1 neurones, which in turn operate as conduits for their effects on GnRH neurones. The physiological relevance of such regulatory phenomena is thoroughly discussed.


Assuntos
Hormônios Esteroides Gonadais/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Ovulação/metabolismo , Puberdade/metabolismo , Animais , Hormônio Liberador de Gonadotropina/metabolismo , Humanos
12.
Biol Reprod ; 83(6): 909-18, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20686180

RESUMO

Oocyte and embryo metabolism are closely linked with their subsequent developmental capacity. Lipids are a potent source of cellular energy, yet little is known about lipid metabolism during oocyte maturation and early embryo development. Generation of ATP from lipids occurs within mitochondria via beta-oxidation of fatty acids, with the rate-limiting step catalyzed by carnitine palmitoyl transferase I (CPT1B), a process also requiring carnitine. We sought to investigate the regulation and role of beta-oxidation during oocyte maturation and preimplantation development. Expression of Cpt1b mRNA, assessed by real-time RT-PCR in murine cumulus-oocyte complexes (COCs), increased following hormonal induction of oocyte maturation and ovulation in vivo with human chorionic gonadotropin (5 IU) and in embryos reaching the blastocyst stage. Beta-oxidation, measured by the production of (3)H(2)O from [(3)H]palmitic acid, was significantly increased over that in immature COCs following induction of maturation in vitro with epidermal growth factor (3 ng/ml) and follicle-stimulating hormone (50 mIU/ml). The importance of lipid metabolism for oocyte developmental competence and early embryo development was demonstrated by assessing the rate of embryo development following inhibition or upregulation of beta-oxidation with etomoxir (an inhibitor of CPT1B) or L-carnitine, respectively. Inhibition of beta-oxidation during oocyte maturation or zygote cleavage impaired subsequent blastocyst development. In contrast, L-carnitine supplementation during oocyte maturation significantly increased beta-oxidation, improved developmental competence, and in the absence of a carbohydrate energy supply, significantly increased 2-cell cleavage. Thus, carnitine is an important cofactor for developing oocytes, and fatty acids are an important energy source for oocyte and embryo development.


Assuntos
Desenvolvimento Embrionário , Ácidos Graxos/metabolismo , Oócitos/metabolismo , Oogênese , Zigoto/metabolismo , Animais , Blastocisto/metabolismo , Carnitina/metabolismo , Carnitina O-Palmitoiltransferase/antagonistas & inibidores , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Células Cultivadas , Células do Cúmulo/efeitos dos fármacos , Células do Cúmulo/metabolismo , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Oócitos/efeitos dos fármacos , Oogênese/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Ovulação/metabolismo , RNA Mensageiro/metabolismo , Zigoto/citologia , Zigoto/efeitos dos fármacos
13.
J Mol Endocrinol ; 38(1-2): 35-50, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17242168

RESUMO

Ovarian progesterone (Prog) is an essential steroid hormone for the secretion of GnRH and reproductive behavior. It exerts primary effects through the progesterone receptor (PR). When analyzed separately in vitro, PR isoforms (PR-A, PR-B) display striking differences in transcriptional activity. The present study was undertaken to determine the in vivo impact of each isoform on hypothalamic function in female mice with ablation of a single isoform, either PR-A or PR-B. To this end, we used single-cell RNA analyses, reverse transcriptase real-time (q)PCR mRNA analyses of punched-out tissue, immunohistochemistry, and reproductive behavior. We provide evidence for the requirement of PR-A in individual ventrolateral ventromedial nucleus (vlVMN) neurons for Prog-facilitated proceptive and receptive behaviors in estrogen benzoate (EB)-primed females and the reciprocal male interactions. We clarify histological and molecular mechanisms of PR isoform activity by showing that (1) PR-A is predominant in individual vlVMN neurons controlling female lordosis circuitry, whilst (2) PR-B is predominant in those VMN subdivisions that provide for amplification of PR-A activity. We go on to demonstrate that PR-A is dominant in the anteroventral periventricular nucleus but not the arcuate nucleus that feed fibers into and around the VMN. In the medial preoptic area, high levels of GnRH RNA in EB-primed PR-A-expressing mice were seen coincident with increased plasma LH levels. Two consecutive GnRH pulses enhanced LH only in primed PR-A-expressing females. In all, the findings are consistent with the hypothesis that hypothalamic PR-A-mediated genomic activities result in reproductive behavior coordinated with ovulation.


Assuntos
Estrogênios/fisiologia , Gonadotropinas/metabolismo , Hipotálamo/fisiologia , Receptores de Progesterona/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Feminino , Regulação da Expressão Gênica/fisiologia , Masculino , Camundongos , Camundongos Knockout , Ovulação/metabolismo , Isoformas de Proteínas/genética , Receptores de Progesterona/genética
14.
J Mol Neurosci ; 20(2): 141-52, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12794308

RESUMO

The presence of pituitary adenylate cyclase-activating polypeptide (PACAP) and its mRNAin the three levels of the hypothalamo-hypophyseal-ovarian axis was previously demonstrated using immunohistochemistry, in situ hybridization, and reverse transcriptase polymerase chain reaction (RT-PCR). In the hypothalamus, PACAP is present in neuroendocrine effector cells and in the median eminence. In the anterior pituitary and ovary, PACAP is transiently present during the proestrous stage of the estrous cycle. In the pituitary, PACAP was observed in gonadotropes. In the ovary, PACAP was demonstrated in the granulosa cells of the preovulatory ovarian follicles. The effect of PACAP on luteinizing hormone (LH) secretion was demonstrated in in vivo and in vitro models. In our work we have studied the role of PACAP in gonadotropic hormone secretion at hypothalamic and pituitary levels. At the hypothalamic level, PACAP, administered intracerebroventricularly to female rats before the critical period of the proestrus stage, can inhibit LH release and ovulation. Its inhibiting effect is mediated through corticotropin-releasing factor (CRF) and endogenous opioids. PACAP administered to neonatal female rats delayed the onset of puberty by influencing the luteinizing hormone-releasing hormone (LHRH) neuronal system. In the pituitary gland, the release of PACAP depended on the stage of the estrous cycle and on the time of day the animals were sacrificed. On the day of proestrus, the number of PACAP-releasing cells showed a diurnal change with two peaks (in the morning and in the evening). The peak was much higher in the evening at the end of the LH surge than in the morning.


Assuntos
Ciclo Estral/fisiologia , Gonadotropinas/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Neuropeptídeos/metabolismo , Animais , Animais Recém-Nascidos , Hormônio Liberador da Corticotropina/farmacologia , Ciclo Estral/efeitos dos fármacos , Feminino , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Injeções Intraventriculares , Hormônio Luteinizante/metabolismo , Antagonistas de Entorpecentes/farmacologia , Neuropeptídeos/farmacologia , Ovulação/efeitos dos fármacos , Ovulação/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Proestro/efeitos dos fármacos , Proestro/fisiologia , Ratos , Ratos Sprague-Dawley , Somatostatina/farmacologia
15.
Brain Res Bull ; 60(3): 307-15, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12754092

RESUMO

The role played by the serotoninergic system in the control of puberty onset and first ovulation in rats is studied in this paper by analyzing the effects of injecting the neurotoxin 5,6-dihydroxytryptamine (5,6-DHT) into the dorsal (DRN) or medial (MRN) raphe nucleus of 30-day-old female rats. Complete lesion to the DRN resulted in the blockade of ovulation and a decrease in both the number of ovarian follicles and the serum concentration of follicle stimulating hormone (FSH). This treatment was also found to be associated with an increase in serotoninergic activity in the anterior and medial hypothalami. A lesion to the central portion of the DRN resulted in a significant decrease in the concentration of progesterone in serum and in the number of ova shed by ovulating animals. The lesion to the lateral portion of the DRN did not have an apparent effect on ovulation rate, the number of ova shed, nor in hormone serum concentration. The injection of propranolol to rats with a lesion to the DRN restored ovulation in 73% of treated animals and returned serotoninergic activity in the anterior hypothalamus to levels similar to those of sham-operated animals. In turn, in the medial hypothalamus, the increase in serotoninergic activity was not modified. The results presented herein suggest that serotoninergic inputs to the anterior hypothalamus have a direct influence on gonadotropin secretion and first ovulation, while the noradrenergic innervation exerts an indirect influence.


Assuntos
5,6-Di-Hidroxitriptamina/administração & dosagem , Núcleo Mediodorsal do Tálamo/efeitos dos fármacos , Núcleos da Rafe/efeitos dos fármacos , Serotoninérgicos/administração & dosagem , 5,6-Di-Hidroxitriptamina/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Mapeamento Encefálico , Estradiol/sangue , Estro/efeitos dos fármacos , Estro/metabolismo , Feminino , Hormônio Foliculoestimulante/sangue , Ácido Hidroxi-Indolacético/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Hormônio Luteinizante/metabolismo , Núcleo Mediodorsal do Tálamo/fisiologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Folículo Ovariano/fisiopatologia , Ovulação/efeitos dos fármacos , Ovulação/metabolismo , Progesterona/sangue , Propranolol/farmacologia , Núcleos da Rafe/anatomia & histologia , Núcleos da Rafe/fisiologia , Ratos , Ratos Endogâmicos , Serotoninérgicos/farmacologia , Vagina/efeitos dos fármacos , Vagina/metabolismo
16.
J Cell Sci ; 116(Pt 10): 1863-73, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12692188

RESUMO

TSG-6 expression is upregulated in many cell types in response to a variety of proinflammatory mediators and growth factors. This protein is detected in several inflammatory disease states (e.g. rheumatoid arthritis) and in the context of inflammation-like processes, such as ovulation, and is often associated with extracellular matrix remodelling. TSG-6 has anti-inflammatory and chondroprotective effects in various models of inflammation and arthritis, which suggest that it is a component of a negative feedback loop capable of downregulating the inflammatory response. Growing evidence also indicates that TSG-6 acts as a crucial factor in ovulation by influencing the expansion of the hyaluronan-rich cumulus extracellular matrix in the preovulatory follicle. TSG-6 is a member of the Link module superfamily and binds to hyaluronan (a vital component of extracellular matrix), as well as other glycosaminoglycans, via its Link module. In addition, TSG-6 forms both covalent and non-covalent complexes with inter-alpha-inhibitor (a serine protease inhibitor present at high levels in serum) and potentiates its anti-plasmin activity.


Assuntos
Moléculas de Adesão Celular/fisiologia , Inflamação , Animais , Artrite/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Ácido Hialurônico/química , Inflamação/metabolismo , Modelos Biológicos , Modelos Moleculares , Ovulação/metabolismo , Estrutura Terciária de Proteína
17.
Anim Reprod Sci ; 49(2-3): 207-14, 1997 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-9505113

RESUMO

A GnRH antagonist, Ac-D-p-Cl-Phe1,2, D-Trp3, D-Arg6, D-Ala10 GnRHb (Organon), was utilized to determine the effective dosage and duration to inhibit LH secretion in the pig. In a preliminary trial, barrows received either 10, 50, or 250 micrograms/kg BW of the GnRH antagonist. Secretion of LH was inhibited within 30 min for a duration of 12 h with the 100 micrograms/kg dose but persisted for greater than 48 h with the 250 micrograms/kg treatment. A second study determined effectiveness of the antagonist for inhibiting ovulation in cyclic gilts. At first detection of standing estrus, cyclic gilts were treated with either saline (control), 100, or 200 micrograms/kg BW of the GnRH antagonist (GnRH1). A second group of GnRH antagonist gilts received 200 micrograms/kg BW of the GnRH antagonist approximately 8 h prior to standing estrus (GnRH2). The GnRH1-treatment failed to inhibit or delay ovulation. Ovulation was inhibited and estrous cycles lengthened in GnRH2-treated gilts. These preliminary results suggest that ovulation in the gilt can be inhibited if the GnRH antagonist is administered prior to the LH surge.


Assuntos
Estradiol/metabolismo , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Luteinizante/metabolismo , Ovulação/sangue , Suínos/sangue , Animais , Cateteres de Demora/veterinária , Estudos de Coortes , Feminino , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Antagonistas de Hormônios/administração & dosagem , Antagonistas de Hormônios/farmacologia , Hormônio Luteinizante/efeitos dos fármacos , Masculino , Ovulação/efeitos dos fármacos , Ovulação/metabolismo , Distribuição Aleatória , Suínos/fisiologia , Fatores de Tempo
18.
J Anim Sci ; 73(10): 3022-9, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8617673

RESUMO

Two experiments were performed. The first experiment was done to study the effects of dietary energy source on plasma insulin concentration using five gilts in a Latin square design with two diets over two periods. The diets contained either 200 g/kg of cornstarch (Starch) or soybean oil (Fat) as energy sources. Results indicate that insulin response was greater in the Starch-fed than in the Fat-fed gilts. A second experiment was performed in which 18 multiparous sows were fed one of the two experimental diets from farrowing until slaughter at d 35 of subsequent pregnancy. All sows nursed nine pigs. Blood samples were taken from a permanent jugular vein catheter every 12 min during a 12-h period on d 109 +/- 1 of pregnancy, on d 7 +/- 1, 14 +/- 1, and 21 +/- 1 of lactation, and on the day of weaning ( d 22 +/- 1). From 48 h after weaning, blood samples were taken every 4 h until 24 h after ovulation. After that, blood samples were taken at 12-h intervals until d 10 after ovulation. Differences between diets in insulin response were not significant. In Starch-fed sows, LH pulsatility at d 7 of lactation was greater (P < .05), the preovulatory LH surge was greater ( P < .05), and progesterone production was greater (P < .05) from 108 h until 256 h after the LH surge than in the Fat-fed sows. Results indicate that feeding Starch-rich diets to multiparous sows compared with Fat-rich diets, on an isocaloric basis, increases LH pulsatility during early lactation, the preovulatory LH surge, and progesterone production after the LH surge.


Assuntos
Estradiol/sangue , Estro/metabolismo , Insulina/sangue , Lactação/metabolismo , Hormônio Luteinizante/sangue , Progesterona/sangue , Suínos/sangue , Animais , Peso Corporal/fisiologia , Carboidratos da Dieta/farmacologia , Gorduras na Dieta/farmacologia , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Estro/fisiologia , Feminino , Morte Fetal/veterinária , Lactação/fisiologia , Ovulação/metabolismo , Ovulação/fisiologia , Gravidez , Óleo de Soja/administração & dosagem , Óleo de Soja/farmacologia , Amido/administração & dosagem , Amido/farmacologia , Suínos/fisiologia
19.
Poult Sci ; 72(5): 839-47, 1993 May.
Artigo em Inglês | MEDLINE | ID: mdl-8502606

RESUMO

The equilibrium dissociation constant (Kd) and the maximum binding capacity (Bmax) of estrogen receptor in soluble (cytosolic) and insoluble (nuclear) fractions in a hypotonic buffer solution of hypothalamus containing preoptic (HPOA) and median eminence (HMEA) areas and of anterior pituitary (AP) of laying and nonlaying hens were examined by Scatchard analysis of specific [3H]estradiol-17 beta ([3H]-E2) binding. The Kd of receptor in all of the tissues was different neither between soluble and insoluble fractions, nor between laying and nonlaying hens. The Bmax in laying hens was greater in the insoluble fraction and lower in the soluble fraction than that in nonlaying hens, but the total binding capacity (sum of Bmax in the soluble and insoluble fractions) was not different between laying and nonlaying hens. In laying hens, the specific [3H]-E2 binding in the insoluble fraction of HPOA was found to increase at 21 h before ovulation and again at 8 to 6 h before ovulation, and of HMEA and AP at 18 to 11 h before ovulation. No change in the specific [3H]-E2 binding in the insoluble and soluble fractions was found in any of the tissues of nonlaying hens during a 24-h period. The results suggest that in laying hens, estrogen may act on the hypothalamus and pituitary at restricted hours during the ovulatory cycle.


Assuntos
Galinhas/fisiologia , Hipotálamo/metabolismo , Ovulação/fisiologia , Hipófise/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Galinhas/metabolismo , Estradiol/metabolismo , Feminino , Ovulação/metabolismo , Ensaio Radioligante
20.
J Hirnforsch ; 33(2): 195-202, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1447521

RESUMO

The distribution of gonadotropin-releasing hormone (GnRH) has been examined in the hypothalamus of adult female bats, Miniopterus schreibersii schreibersii, along annual reproductive cycle by means of light-microscopic immunocytochemistry. GnRH-immunoreactive cells are localized throughout the medial basal hypothalamus, being specially numerous in the arcuate nucleus. They are generally bipolar or monopolar neurons with smooth contour, although spiny cells were also found exclusively in the arcuate nucleus from periovulatory bats. Depletion in both number and immunocytochemical labelling of GnRH perikarya is detected in pregnant and lactating bats. GnRH-immunoreactive fibers are distributed in the internal layer of the median eminence and the infundibular stalk. Pregnant and lactating animals show lower GnRH fibers content and a less intense labelling than in periovulatory and hibernating conditions. The results suggest that changes in the secretory activity of GnRH neurons may be associated with hibernation and delayed implantation that suffer these mammalian species.


Assuntos
Quirópteros/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Hibernação/fisiologia , Hipotálamo/anatomia & histologia , Imuno-Histoquímica , Eminência Mediana/anatomia & histologia , Eminência Mediana/fisiologia , Fibras Nervosas/metabolismo , Ovulação/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA