Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Zoology (Jena) ; 162: 126147, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38277721

RESUMEN

Birds living at high latitudes perceive the photoperiod through deep-brain photoreceptors (DBP) located in deep-brain neurons. During long photoperiods the information transmitted by these photoreceptors increases the activity of the hypothalamic-pituitary-gonadal (HPG) axis, leading to gonadal development. The presence of photopigments such as VA-Opsin, Opn4, Opn5 and Opn2 in brain areas implicated in reproductive behaviors has been firmly established in several avian species with seasonal breeding, whereas their existence in opportunistic breeding birds remains unconfirmed. The Eared Dove is an urban and peri-urban dove that breeds throughout the year. Males of this species do not exhibit the typical gonadal regression/recrudescence cycle, thus posing the question of what occurs upstream of the HPG axis. We addressed this issue by first studying the presence of diverse opsins located in DBP in the brains of Eared Dove males and whether these photopigments changed their expression throughout the year. We carried out an immunohistochemistry analysis on three different opsins: Opn2 (rhodopsin), Opn3 and Opn5. Our results demonstrate the discrete neuroanatomical distribution of these opsins in the brain of Eared Dove males and strongly indicate different seasonal expressions. In the anterior region of the hypothalamus, Opn2-positive cells were detected throughout the year. By contrast, Opn5 was found to be strongly and seasonally expressed during winter in the anterior and the hypothalamic region. Opn3 was also found to be significantly and seasonally expressed during winter in the hypothalamic region. We thus demonstrate for the first time that males of the Eared Dove, have three different deep-brain opsin-expressing photoreceptors with differential location/distribution in the anterior and hypothalamic region and differential seasonality. The persistence of Opn2 and the strong seasonal expression of nonvisual photopigments Opn3 and Opn5 in two areas of the avian brain, which are associated with reproduction, could be the primary distinction between seasonal and opportunistic breeders.


Asunto(s)
Columbidae , Opsinas , Masculino , Animales , Opsinas/genética , Opsinas/metabolismo , Hipotálamo/metabolismo , Encéfalo , Gónadas/metabolismo , Estaciones del Año
2.
Free Radic Biol Med ; 194: 199-208, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36470319

RESUMEN

BACKGROUND: The maturation of the hypothalamic-pituitary-gonadal (HPG) axis is crucial for the establishment of reproductive function. In female mice, neuronal nitric oxide synthase (nNOS) activity appears to be key for the first postnatal activation of the neural network promoting the release of gonadotropin-releasing hormone (GnRH), i.e. minipuberty. However, in males, the profile of minipuberty as well as the role of nNOS-expressing neurons remain unexplored. METHODS: nNOS-deficient and wild-type mice were studied during postnatal development. The expression of androgen (AR) and estrogen receptor alpha (ERα) as well as nNOS phosphorylation were evaluated by immunohistochemistry in nNOS neurons in the median preoptic nucleus (MePO), where most GnRH neuronal cell bodies reside, and the hormonal profile of nNOS-deficient male mice was assessed using previously established radioimmunoassay and ELISA methods. Gonadectomy and pharmacological manipulation of ERα were used to elucidate the mechanism of minipubertal nNOS activation and the maturation of the HPG axis. RESULTS: In male mice, minipubertal FSH release occurred at P23, preceding the LH surge at P30, when balanopreputial separation occurs. Progesterone and testosterone remained low during minipuberty, increasing around puberty, whereas estrogen levels were high throughout postnatal development. nNOS neurons showed a sharp increase in Ser1412 phosphorylation of nNOS at P23, a phenomenon that occurred even in the absence of the gonads. In male mice, nNOS neurons did not appear to express AR, but abundantly expressed ERα throughout postnatal development. Selective pharmacological blockade of ERα during the infantile period blunted Ser1412 phosphorylation of nNOS at P23. CONCLUSIONS: Our results show that the timing of minipuberty differs in male mice when compared to females, but as in the latter, nNOS activity in the preoptic region plays a role in this process. Additionally, akin to male non-human primates, the profile of minipuberty in male mice is shaped by sex-independent mechanisms, and possibly involves extragonadal estrogen sources.


Asunto(s)
Receptor alfa de Estrógeno , Piridinolcarbamato , Femenino , Ratones , Masculino , Animales , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Receptor alfa de Estrógeno/genética , Hormona Liberadora de Gonadotropina/análisis , Hormona Liberadora de Gonadotropina/metabolismo , Estrógenos/metabolismo , Gónadas/química , Gónadas/metabolismo , Neuronas/metabolismo , Hipotálamo/metabolismo
3.
Int J Mol Sci ; 23(18)2022 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-36142817

RESUMEN

As lower vertebrates, teleost species could be affected by dynamic aquatic environments and may respond to environmental changes through the hypothalamus-pituitary-gonad (HPG) axis to ensure their normal growth and sexual development. Chinese sea bass (Lateolabrax maculatus), euryhaline marine teleosts, have an extraordinary ability to deal with a wide range of salinity changes, whereas the salinity decrease during their sex-maturation season may interfere with the HPG axis and affect their steroid hormone metabolism, resulting in abnormal reproductive functioning. To this end, in this study, 40 HPG axis genes in the L. maculatus genome were systematically characterized and their copy numbers, phylogenies, gene structures, and expression patterns were investigated, revealing the conservation of the HPG axis among teleost lineages. In addition, freshwater acclimation was carried out with maturing male L. maculatus, and their serum cortisol and 11-ketotestosterone (11-KT) levels were both increased significantly after the salinity change, while their testes were found to be partially degraded. After salinity reduction, the expression of genes involved in cortisol and 11-KT synthesis (cyp17a, hsd3b1, cyp21a, cyp11c, hsd11b2, and hsd17b3) showed generally upregulated expression in the head kidneys and testes, respectively. Moreover, cyp11c and hsd11b2 were involved in the synthesis and metabolism of both cortisol and 11-KT, and after salinity change their putative interaction may contribute to steroid hormone homeostasis. Our results proved the effects of salinity change on the HPG axis and steroidogenic pathway in L. maculatus and revealed the gene interactions involved in the regulation of steroid hormone levels. The coordinated interaction of steroidogenic genes provides comprehensive insights into steroidogenic pathway regulation, as well as sexual development, in teleost species.


Asunto(s)
Lubina , Animales , Lubina/genética , Lubina/metabolismo , China , Gónadas/metabolismo , Hidrocortisona/metabolismo , Hipotálamo/metabolismo , Masculino , Complejos Multienzimáticos/metabolismo , Salinidad
4.
Food Funct ; 13(18): 9544-9558, 2022 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-35997033

RESUMEN

This study investigated the influence of carrier oils on the in vitro and in vivo bioavailability of PTE encapsulated in scallop gonad protein isolates (SGPIs)-epigallocatechin gallate (EGCG) conjugate stabilized emulsions. The SGPIs-EGCG stabilized emulsions were subjected to an in vitro simulated digestion, and the resulting corn oil and MCT micelles were used to evaluate the PTE transportation using the Caco-2 cell model. Both emulsions remarkably improved the bioaccessibility of PTE in the micelle phase. Nevertheless, corn oil emulsions increased trans-enterocyte transportation of PTE more efficiently than MCT emulsions. Furthermore, the maximum plasma concentrations of PTE and its metabolites in mice fed with PTE emulsions were prominently higher than those in mice fed with PTE solution, while the in vivo metabolic patterns of PTE in different oil-stabilized emulsions were different. Therefore, SGPIs-EGCG stabilized emulsions could enhance the bioavailability of PTE through controlled release, in which corn oil is more suitable than MCT.


Asunto(s)
Micelas , Pectinidae , Animales , Disponibilidad Biológica , Células CACO-2 , Catequina/análogos & derivados , Aceite de Maíz/metabolismo , Preparaciones de Acción Retardada/metabolismo , Emulsiones/metabolismo , Excipientes/metabolismo , Gónadas/metabolismo , Humanos , Ratones , Aceites/metabolismo , Pectinidae/metabolismo , Proteínas/metabolismo , Estilbenos
5.
Neuropeptides ; 91: 102216, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34974357

RESUMEN

Neuropeptides being regulators of the hypothalamus-pituitary-adrenal (HPA) axis activity, also affect the function of the hypothalamus-pituitary-gonadal (HPG) axis by regulating gonadotrophin-releasing hormone (GnRH) secretion from hypothalamic neurons. Here, we review the available data on how neuropeptides affect HPG axis activity directly or indirectly via their influence on the HPA axis. The putative role of neuropeptides in stress-induced infertility, such as polycystic ovary syndrome, is also described. This review discusses both well-known neuropeptides (i.e., kisspeptin, Kp; oxytocin, OT; arginine-vasopressin, AVP) and more recently discovered peptides (i.e., relaxin-3, RLN-3; nesfatin-1, NEFA; phoenixin, PNX; spexin, SPX). For the first time, we present an up-to-date review of all published data regarding interactions between the aforementioned neuropeptide systems. The reviewed literature suggest new pathophysiological mechanisms leading to fertility disturbances that are induced by stress.


Asunto(s)
Gónadas/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Infertilidad/metabolismo , Neuropéptidos/metabolismo , Animales , Femenino , Hipotálamo/metabolismo , Estrés Psicológico/metabolismo
6.
Front Endocrinol (Lausanne) ; 12: 632060, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34149609

RESUMEN

Investigation of the negative impacts of stress on reproduction has largely centered around the effects of the adrenal steroid hormone, corticosterone (CORT), and its influence on a system of tissues vital for reproduction-the hypothalamus of the brain, the pituitary gland, and the gonads (the HPG axis). Research on the action of CORT on the HPG axis has predominated the stress and reproductive biology literature, potentially overshadowing other influential mediators. To gain a more complete understanding of how elevated CORT affects transcriptomic activity of the HPG axis, we experimentally examined its role in male and female rock doves (Columba livia). We exogenously administrated CORT to mimic circulating levels during the stress response, specifically 30 min of restraint stress, an experimental paradigm known to increase circulating CORT in vertebrates. We examined all changes in transcription within each level of the HPG axis as compared to both restraint-stressed birds and vehicle-injected controls. We also investigated the differential transcriptomic response to CORT and restraint-stress in each sex. We report causal and sex-specific effects of CORT on the HPG transcriptomic stress response. Restraint stress caused 1567 genes to uniquely differentially express while elevated circulating CORT was responsible for the differential expression of 304 genes. Only 108 genes in females and 8 in males differentially expressed in subjects that underwent restraint stress and those who were given exogenous CORT. In response to elevated CORT and restraint-stress, both sexes shared the differential expression of 5 genes, KCNJ5, CISH, PTGER3, CEBPD, and ZBTB16, all located in the pituitary. The known functions of these genes suggest potential influence of elevated CORT on immune function and prolactin synthesis. Gene expression unique to each sex indicated that elevated CORT affected more gene transcription in females than males (78 genes versus 3 genes, respectively). To our knowledge, this is the first study to isolate the role of CORT in HPG genomic transcription during a stress response. We present an extensive and openly accessible view of the role corticosterone in the HPG transcriptomic stress response. Because the HPG system is well conserved across vertebrates, these data have the potential to inspire new therapeutic strategies for reproductive dysregulation in multiple vertebrate systems, including our own.


Asunto(s)
Columbidae/fisiología , Corticosterona/fisiología , Gónadas/metabolismo , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Hipotálamo/metabolismo , Hipófisis/metabolismo , Reproducción/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Animales , Corticosterona/metabolismo , Femenino , Perfilación de la Expresión Génica , Masculino , Factores Sexuales , Estrés Fisiológico
7.
Br J Nutr ; 126(2): 172-182, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-33054887

RESUMEN

The present study aimed to evaluate the inflammatory response, oxidative status and fatty acid deposition in reproductive tissues of cats supplemented with the dried microalgae Schizochytrium spp. (Thraustochytriaceae) as a DHA source. Thirty-seven cats (males, n 21; females, n 16; 11·5 (sd 0·5) months of age) were divided by sex into five groups. Treatment diets contained algae biomass at 4·0, 8·0, 12·0 or 16·0 g/kg replacing poultry fat (n-6 source). Cats were fed the respective diet for 62 d and neutered on day 58. Blood samples were collected at the beginning of the experiment (day 1), before neutering (day 58) and 4 d after surgery (day 62) for analysis of inflammation and oxidative markers. Acute-phase protein levels were altered (P < 0·01) in the postoperative period, without any treatment effect (P > 0·05). PGE2 concentrations after surgery were reduced linearly (R2 0·8706; P = 0·002) with microalgal inclusion. Blood platelet count was reduced (P = 0·001) after the surgery regardless treatment, but it was higher in the DHA group compared with control (P < 0·001). The DHA deposition (testicles, R2 0·846; ovaries, R2 0·869) and the n-6:n-3 ratio (testicles, R2 0·859; ovaries, R2 0·955) in gonads had a pattern which fitted a quadratic model. DHA from Schizochytrium spp. modifies PGE2 response after the surgery in cats. The physiological roles of the DHA in the reproduction of cats were not investigated, but its gonadal deposition after supplementation was observed.


Asunto(s)
Gatos , Dieta/veterinaria , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Grasos/metabolismo , Inflamación/veterinaria , Microalgas , Animales , Suplementos Dietéticos , Dinoprostona , Femenino , Gónadas/metabolismo , Masculino , Microalgas/química
8.
Cell Mol Life Sci ; 78(1): 1-16, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32564094

RESUMEN

Research into the physiological actions of anti-Müllerian hormone (AMH) has rapidly expanded from its classical role in male sexual differentiation to the regulation of ovarian function, routine clinical use in reproductive health and potential use as a biomarker in the diagnosis of polycystic ovary syndrome (PCOS). During the past 10 years, the notion that AMH could act exclusively at gonadal levels has undergone another paradigm shift as several exciting studies reported unforeseen AMH actions throughout the Hypothalamic-Pituitary-Gonadal (HPG) axis. In this review, we will focus on these findings reporting novel AMH actions across the HPG axis and we will discuss their potential impact and significance to better understand human reproductive disorders characterized by either developmental alterations of neuroendocrine circuits regulating fertility and/or alterations of their function in adult life. Finally, we will summarize recent preclinical studies suggesting that elevated levels of AMH may potentially be a contributing factor to the central pathophysiology of PCOS and other reproductive diseases.


Asunto(s)
Hormona Antimülleriana/metabolismo , Gónadas/metabolismo , Hipotálamo/metabolismo , Hipófisis/metabolismo , Femenino , Humanos , Sistemas Neurosecretores/crecimiento & desarrollo , Sistemas Neurosecretores/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Reproducción , Transducción de Señal
9.
Fish Physiol Biochem ; 47(1): 93-108, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33215297

RESUMEN

Gonadotropin-inhibitory hormone (GnIH) plays a critical role in regulating gonadotropin-releasing hormone (GnRH), gonadotropin hormone (GtH), and steroidogenesis. The Lpxrfa (the piscine ortholog of GnIH) system has been found to regulate fish reproduction. To gain insight into the role of Lpxrfa in the regulation of spotted scat (Scatophagus argus) reproduction, spotted scat Lpxrfa (ssLpxrfa), and its receptor (ssLpxrfa-r) were cloned and analyzed. Tissue distribution and expression patterns at the hypothalamo-pituitary-gonadal axis (HPG axis) of sslpxrfa and sslpxrfa-r mRNA were also investigated during gonadal development of spotted scat. The open reading frame (ORF) of the sslpxrfa was 606 bp encoding 201 amino acids and includes a putative signal peptide and two mature ssLpxrfa peptides with LPXRFamide motif at their C-terminus. The sslpxrfa-r ORF was 1449 bp encoding 482 amino acids and contracted a seven-hydrophobic transmembrane (TM) domain structure. The tissue distribution showe d that the sslpxrfa was highly expressed in hypothalami, gill, and the gonads. In addition, sslpxrfa-r was highly expressed in hypothalami, pituitaries, and the gonads. Quantitative real-time polymerase chain reaction (qPCR) revealed that sslpxrfa had the highest expression in the hypothalami and pituitaries, and the lowest expression in the gonads in stage V. During gonadal development, the expression of sslpxrfa-r was gradually increased in the hypothalami but reduced in the gonads. However, no obvious trend was observed in the pituitaries. The expression of sslpxrfa and sslpxrfa-r decreased significantly after injection with 17ß-estradiol (E2). However, the expression of both sslpxrfa and sslpxrfa-r was not changed after injection with 17α-methyltestosterone(17α-MT) in the hypothalami. In addition, no changes were observed in the expression of fshß and lhß in the pituitaries after injecting ssLpxrfa-1. However, ssLpxrfa-2 could downregulate the expression of sbgnrh and fshß in the hypothalami and pituitaries, respectively. Taken together, these findings suggested that ssLpxrfa may participate in E2 feedback in reproduction and regulate the reproductive axis of spotted scat.


Asunto(s)
Proteínas de Peces/genética , Peces/genética , Neuropéptidos/genética , Receptores de Neuropéptido/genética , Reproducción/genética , Secuencia de Aminoácidos , Animales , Estradiol/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Gónadas/metabolismo , Sistema Hipotálamo-Hipofisario , Hipotálamo/metabolismo , Masculino , Metiltestosterona/farmacología , Filogenia , Hipófisis/metabolismo
10.
Int J Mol Sci ; 21(23)2020 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-33276521

RESUMEN

Anthropogenic endocrine-disrupting chemicals (EDCs) can contaminate air, soil, and water. Human exposures to EDCs occur through inhalation, absorption, and ingestion. EDCs act by disrupting various pathways in the endocrine system. When the hypothalamic-pituitary-gonadal (HPG) axis is disrupted by EDCs, there can be effects on fertility in both men and women. Not only can fertility be indirectly affected by EDC disruptions of the HPG axis, but EDCs can also directly affect the menstrual cycle and sperm morphology. In this review, we will discuss the current findings on EDCs that can be inhaled. This review examines effects of exposure to prominent EDCs: brominated and organophosphate flame retardants, diesel exhaust, polycyclic aromatic hydrocarbons, cadmium and lead, TCDD, and polychlorinated biphenyls on fertility through alterations that disrupt the HPG axis and fertility through inhalation. Although the studies included herein include multiple exposure routes, all the studies indicate receptor interactions that can occur from inhalation and the associated effects of all compounds on the HPG axis and subsequent fertility.


Asunto(s)
Contaminantes Atmosféricos/efectos adversos , Disruptores Endocrinos/efectos adversos , Gónadas/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipófisis/efectos de los fármacos , Contaminantes Atmosféricos/clasificación , Animales , Disruptores Endocrinos/clasificación , Fertilidad/efectos de los fármacos , Gónadas/metabolismo , Humanos , Hipotálamo/metabolismo , Metales Pesados/química , Hipófisis/metabolismo , Hidrocarburos Policíclicos Aromáticos/efectos adversos , Factores Sexuales , Emisiones de Vehículos/toxicidad
11.
Mol Metab ; 36: 100974, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32272092

RESUMEN

OBJECTIVE: Polyunsaturated fatty acids (PUFAs), including essential fatty acids linoleic and α-linolenic acid and derived long chain and very long chain ω3-and ω6-polyunsaturated fatty acids, are vital structures in mammalian membrane systems and signaling molecules, pivotal in brain development, lipid, and energy metabolism and in female and male fertility during human evolution. Numerous nutritional studies suggest imbalance of PUFA metabolism as a critical factor in the pathogenesis of several human lifestyle diseases: dyslipoproteinemia, obesity, cardiovascular and neurodegenerative diseases, and infertility. The lack of unbiased animal models impedes molecular interpretation of the role of synthesized and dietary supplied PUFAs in these conditions. In this study, we used a Δ6 fatty acid desaturase (FADS2) deficient mouse mutant lacking key enzyme activity in the biosynthesis of ω3-and ω6-PUFAs from EFAs to address the molecular role of PUFAs in female and male fertility. Infertility is a hallmark of the pleiotropic but auxotrophic fads2-/- phenotype and is therefore helpful for stringent dietary studies on the role of individual PUFAs. METHODS: Feeding regimens: Age- and gender-matched infertile fads2-/- mice were maintained on defined diets, normal diet containing essential fatty acids, and supplemented with ω6-arachidonic acid, ω3-docosahexaenoic acid, and arachidonic/docosahexaenoic acid, starting (a) after weaning and (b) initiated in 4-month-old female and male fads2-/- mice. Phospho- and sphingolipidomes of ovarian and testicular membrane lipid bilayers in each cohort were established and the impact on the expression and topology of membrane marker proteins, membrane morphology, germ cell development, and female and male fertility in the respective cohorts was elaborated. RESULTS: PUFA synthesis deficiency caused a halt to folliculogenesis, atresia of oocytes, and infertility of fads2-/- female mice. A PUFA-deficient membrane lipid bilayer core structure led to the disassembly of the gap junction network of the follicular granulosa cells. In fads2-/- testis, the blood-testis barrier was disrupted and spermatogenesis arrested, leading to infertility. Sustained supply of combined AA and DHA remodeled the PUFA-deficient ovarian and testicular membrane lipidomes, facilitating the reassembly of the functional gap junction network for regular ovarian cycles and the reconstitution of the blood-testis barrier in Sertoli cells, reconstituting fertility not only in developing newborns, but surprisingly also in adult infertile fads2-/- mice. CONCLUSIONS: These findings demonstrate the previously unrecognized membrane structure-based molecular link between nutrient ω3-and ω6-PUFAs, gonadal membrane structures, and female and male fertility and might foster studies of the pivotal role of dietary PUFAs in human fertility.


Asunto(s)
Ácido Graso Desaturasas/metabolismo , Ácidos Grasos Insaturados/metabolismo , Fertilidad/fisiología , Animales , Dieta , Suplementos Dietéticos , Ácido Graso Desaturasas/deficiencia , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/metabolismo , Femenino , Gónadas/efectos de los fármacos , Gónadas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad
12.
Biol Trace Elem Res ; 196(2): 494-501, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31656014

RESUMEN

The purpose of this research is to investigate the absorption, distribution, excretion, and pharmacokinetics of selenite in rats after intragastric administration, and thus illustrate the efficiency of selenium (Se) supplementation. After a single gavage of sodium selenite, a concentration of Se in plasma and tissues was determined by inductively coupled plasma mass spectrometry (ICP-MS) at different time points. Through fitting the data with the metabolic kinetic model, the corresponding kinetic parameters were determined for plasma and tissues, including kidney, liver, heart, muscle, and gonad. While the metabolic kinetics of sodium selenite in plasma, liver, and kidney of rats was well reflected by a two-compartment open model, that in heart and gonad was fitted to a one-compartment open model, and that in muscle was fitted to a one-compartment open model with a lag time. The results indicate that sodium selenite was absorbed by plasma and tissues quickly and was eliminated slowly after intragastric administration. Based on the results, we propose that multi-supplementation of Se with low dosage is superior to single supplementation with high dosage, in terms of avoiding selenosis.


Asunto(s)
Gónadas/metabolismo , Corazón , Riñón/metabolismo , Hígado/metabolismo , Músculos/metabolismo , Selenito de Sodio/farmacocinética , Administración Oral , Animales , Femenino , Gónadas/química , Riñón/química , Cinética , Hígado/química , Masculino , Músculos/química , Ratas , Ratas Wistar , Selenito de Sodio/administración & dosificación , Selenito de Sodio/sangre , Distribución Tisular
13.
Gen Comp Endocrinol ; 287: 113322, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31738909

RESUMEN

Most endocrine systems in the body are influenced by the hypothalamic-pituitary axis. Within this axis, the hypothalamus delivers precise signals to the pituitary gland, which in turn releases hormones that directly affect target tissues including the liver, thyroid gland, adrenal glands and gonads. This action modulates the release of additional hormones from the sites of action, regulating key physiological processes, including growth, metabolism, stress and reproduction. Pituitary hormones are released by five distinct hormone-producing cell types: somatotropes (which produce growth hormone), thyrotropes (thyrotropin), corticotropes (adrenocorticotropin), lactotropes (prolactin) and gonadotropes (follicle stimulating hormone and luteinizing hormone), each modulated by specific hypothalamic signals. This careful and distinct organization of the hypothalamo-pituitary axis has been classically associated with the existence of many lineal axes (e.g., the hypothalamic-pituitary-gonadal axis) in charge of the control of the different physiological processes. While this traditional concept is valid, it is becoming apparent that hormones produced by the hypothalamo-pituitary axis have diverse effects. For instance, gonadotropin-releasing hormone II has been associated with a suppressive effect on food intake in fish. Likewise, growth hormone has been shown to influence appetite, swimming activity and aggressive behavior in fish. This review will focus on the hypothalamic and pituitary hormones classically involved in regulating growth and reproduction, and will attempt to provide a general overview of the current knowledge on their actions on energy balance and appetite in fish. It will also give a brief perspective of the role of some of these peptides in integrating feeding, metabolism, growth and reproduction.


Asunto(s)
Metabolismo Energético/fisiología , Peces/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Hormona del Crecimiento/metabolismo , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Hipófisis/metabolismo , Animales , Gónadas/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hormonas Hipofisarias/metabolismo , Reproducción/fisiología
14.
Sci Rep ; 9(1): 11289, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31383892

RESUMEN

There is growing evidence that obstructive sleep apnoea (OSA) influences the hypothalamic-pituitary-gonadal axis (HPG axis) in men. The aim of the study was to assess the association of nesfatin-1 with HPG axis disturbances in OSA. This is a prospective study with consecutive enrolment. It comprises 72 newly diagnosed OSA patients ((AHI: apnoea-hypopnea index) 18 subjects: 5 ≤ AHI < 15; 24: 15 ≤ AHI < 30; 30: AHI ≥ 30) and a control group composed of 19 patients (AHI < 5). All patients underwent polysomnography and fasting blood collection for nesfatin-1, testosterone, luteinising hormone (LH), high-sensitivity C-reactive protein (hsCRP), aspartate transaminase (AST), alanine aminotransferase (ALT), creatinine and glucose. Groups had similar levels of LH, nesfatin-1 and testosterone (p = 0.87; p = 0.24; p = 0.08). Nesfatin-1 was not correlated to LH (p = 0.71), testosterone (p = 0.38), AHI (p = 0.34) or the oxygen desaturation index (ODI) (p = 0.69) either in the whole group, or in sub-groups. The study did not reveal any association between the HPG axis and nesfatin-1 in OSA adult males. It is possible that nesfatin-1 is not a mediator of HPG axis disturbances in adult patients with OSA.


Asunto(s)
Gónadas/metabolismo , Hipotálamo/metabolismo , Nucleobindinas/metabolismo , Hipófisis/metabolismo , Apnea Obstructiva del Sueño/metabolismo , Anciano , Glucemia/análisis , Ayuno , Humanos , Masculino , Persona de Mediana Edad , Nucleobindinas/sangre , Polisomnografía , Estudios Prospectivos , Apnea Obstructiva del Sueño/sangre
15.
Artículo en Inglés | MEDLINE | ID: mdl-31158494

RESUMEN

Various endocrine factors that regulate energy homeostasis are also implicated in the reproductive physiology of mammals. However, the hormonal link between metabolism and reproduction in fish is poorly understood. Ghrelin is a multifunctional hormone with both metabolic and reproductive roles in vertebrates. Post-translational acylation by ghrelin-O-acyltransferase (GOAT) is critical for its biological actions. The expression of ghrelin, ghrelin or growth hormone secretagogue receptor (GHSR), and GOAT (which forms the ghrelinergic system) in fish under metabolic stress remains unclear. In this research, we used RT-qPCR and Western blot analysis to determine the expression of the ghrelinergic system in goldfish (during the reproductively active phase) hypothalamus and gonads under 7 and 28 days of fasting. We found a significant increase in preproghrelin mRNA expresson in the ovary, and GOAT mRNA expression in the testis of goldfish deprived of food for 7 days. In fish deprived of food for 28 days, preproghrelin, GHSR and GOAT mRNA expression was significantly increased in the hypothalamus of male goldfish. Such differences were not observed in the hypothalamus of female fish, and in the testis of 28 days fasted fish. Meanwhile, preproghrelin, GHSR, and GOAT expression (both mRNA and protein) was significantly increased in the ovary of female fish fasted for 28 days. Ghrelin has been shown to suppress oocyte maturation in fish. The upregulation of a system that has ovarian inbititory roles suggests a role for ghrelin in maintaining reduced reproductive capability during metabolically challenging periods.


Asunto(s)
Aciltransferasas/genética , Ghrelina/genética , Carpa Dorada/genética , Estrés Fisiológico/genética , Animales , Ayuno , Gónadas/crecimiento & desarrollo , Gónadas/metabolismo , Hipotálamo/metabolismo , ARN Mensajero/genética
16.
Gen Comp Endocrinol ; 281: 126-136, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31163181

RESUMEN

To ascertain the significance of the Neurokinin B/Tachykinin 3 receptor (NKB/NK3) system in goldfish reproduction, two cDNAs encoding tachykinin 3 receptors, namely tacr3a and tacr3b, were cloned. Subsequent studies revealed that the downstream signalling of both Tac3rs can be activated by different NKB peptides, suggesting that the cloned receptors are biologically functional in goldfish. RT-PCR analysis showed that tacr3s are widely expressed in brain regions. During the gonadal development, tacr3a and tacr3b exhibited different expression patterns in the hypothalamus and pituitary. The actions of NKB peptides on reproductive axis was further investigated in vivo. Intraperitoneal injections of NKB peptides significantly reduced the expression of kiss2 and gonadotropin releasing hormone 3 (gnrh3) in the hypothalamus, and the expression of luteinizing hormone beta subunit (lhb) and follicle stimulating hormone beta subunit (fshb) in the pituitary in sexually immature goldfish. Taken together, our findings revealed that NKB/NK3 system plays a negative role in the reproductive axis of immature goldfish.


Asunto(s)
Carpa Dorada/fisiología , Neuroquinina B/metabolismo , Receptores de Neuroquinina-3/metabolismo , Reproducción/fisiología , Maduración Sexual , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Células HEK293 , Humanos , Hipotálamo/metabolismo , Masculino , Filogenia , Hipófisis/metabolismo , ARN Mensajero/metabolismo , Receptores de Neuroquinina-3/química , Receptores de Neuroquinina-3/genética
17.
Proc Natl Acad Sci U S A ; 116(23): 11339-11344, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31085642

RESUMEN

During their once-in-a-lifetime transoceanic spawning migration, anguillid eels do not feed, instead rely on energy stores to fuel the demands of locomotion and reproduction while they reorganize their bodies by depleting body reserves and building up gonadal tissue. Here we show how the European eel (Anguilla anguilla) breaks down its skeleton to redistribute phosphorus and calcium from hard to soft tissues during its sexual development. Using multiple analytical and imaging techniques, we characterize the spatial and temporal degradation of the skeletal framework from initial to final gonadal maturation and use elemental mass ratios in bone, muscle, liver, and gonadal tissue to determine the fluxes and fates of selected minerals and metals in the eels' bodies. We find that bone loss is more pronounced in females than in males and eventually may reach a point at which the mechanical stability of the skeleton is challenged. P and Ca are released and translocated from skeletal tissues to muscle and gonads, leaving both elements in constant proportion in remaining bone structures. The depletion of internal stores from hard and soft tissues during maturation-induced body reorganization is accompanied by the recirculation, translocation, and maternal transfer of potentially toxic metals from bone and muscle to the ovaries in gravid females, which may have direct deleterious effects on health and hinder the reproductive success of individuals of this critically endangered species.


Asunto(s)
Anguilla/metabolismo , Anguilla/fisiología , Resorción Ósea/metabolismo , Huesos/metabolismo , Huesos/fisiología , Migración Animal/fisiología , Animales , Fenómenos Biológicos , Calcio/metabolismo , Especies en Peligro de Extinción , Femenino , Gónadas/metabolismo , Gónadas/fisiología , Hígado/metabolismo , Hígado/fisiología , Masculino , Músculos/metabolismo , Músculos/fisiología , Ovario/metabolismo , Ovario/fisiología , Fósforo/metabolismo , Reproducción/fisiología
18.
Gen Comp Endocrinol ; 277: 104-111, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30923007

RESUMEN

WNT4 (wingless-type MMTV integration site family, member 4) plays a key role in the ovarian differentiation and development in mammals. However, the possible roles of Wnt4 during gonadal differentiation and development need further clarification in teleosts. In this study, we cloned and characterized the full-length cDNA of Qi river crucian carp (Carassius auratus) wnt4a gene (CA-wnt4a). The cDNA of CA-wnt4a is 2337 bp, including the ORF of 1059 bp, encoding a putative protein with a transmembrane domain and a WNT family domain. Sequence and phylogenetic analyses revealed that the CA-Wnt4a identified is a genuine Wnt4a. Tissue distribution analysis showed that CA-wnt4a is expressed in all the tissues examined, including ovary. CA-wnt4a undergoes a stepwise increase in the embryonic stages, suggesting that CA-wnt4a might be involved in the early developmental stage. Ontogenic analysis demonstrated that CA-wnt4a expression is upregulated in the ovaries at 30-50 days after hatching (dah), the critical period of sex determination/differentiation in Qi river crucian carp. From 90 dah, the expression of CA-wnt4a was gradually downregulated in the developing ovaries. Immunohistochemistry demonstrated that CA-Wnt4a was expressed in the somatic and germ cells of the ovary by 30 dah, thereafter, positive signals of Wnt4a were detected in the somatic cells, oogonia and primary growth oocytes from 60 dah. In the sex-reversed testis induced by letrozole treatment, the expression level of CA-wnt4a was significantly downregulated. When CA-wnt4a expression was inhibited by injection of FH535 (an inhibitor of canonical Wnt/ß-catenin signal pathway) in the ovaries, levels of cyp19a1a, foxl2 mRNA were significantly downregulated, while sox9b and cyp11c1 were upregulated, which suggested that together with Foxl2-leading estrogen pathway, CA-wnt4a signaling pathway might be involved in ovarian differentiation and repression of the male pathway gene expression in Qi river crucian carp.


Asunto(s)
Carpas/genética , Ríos , Triploidía , Proteína Wnt4/genética , Animales , Carpas/embriología , Clonación Molecular , ADN Complementario/genética , Regulación hacia Abajo/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Gónadas/efectos de los fármacos , Gónadas/metabolismo , Letrozol/farmacología , Masculino , Filogenia , Análisis de Secuencia de ADN , Sulfonamidas/farmacología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
19.
Brain Res ; 1714: 210-217, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30851245

RESUMEN

Growth hormone (GH) and prolactin (PRL) are known as pleiotropic hormones. Accordingly, the distribution of their receptors comprises several organs and tissues, including the central nervous system. The appropriate secretion of both hormones is essential for sexual maturation and maintenance of reproductive functions, while defects in their secretion affect puberty onset and can cause infertility. Conversely, GH therapy at a prepubertal age may accelerate puberty. On the other hand, hyperprolactinemia is a frequent cause of infertility. While the action of PRL in some central components of the Hypothalamic-Pituitary-Gonadal (HPG) axis, such as the kisspeptin neurons, has been well documented, the possible effects of GH in the hypothalamus are still elusive. Thus, the present study was designed to investigate whether somatomammotropin hormones are able to modulate the activity of critical neuronal components of the HPG axis, including kisspeptin neurons and cells of the ventral premammillary nucleus (PMv). Our results revealed that GH effects in kisspeptin neurons of the anteroventral periventricular and rostral periventricular nuclei or in PMv neurons relies predominantly on the recruitment of the signal transducer and activator of transcription 5 (STAT5) rather than through acute changes in resting membrane potential. Importantly, kisspeptin neurons located at the arcuate nucleus were not directly responsive to GH. Additionally, our findings further identified PMv neurons as potential targets of PRL, since PRL induces the phosphorylation of STAT5 and depolarizes PMv neurons. Combined, our data provide evidence that GH and PRL may affect the HPG axis via specific hypothalamic neurons.


Asunto(s)
Hormona del Crecimiento/metabolismo , Prolactina/metabolismo , Maduración Sexual/fisiología , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Gónadas/metabolismo , Hormona del Crecimiento/fisiología , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Fosforilación , Sistema Hipófiso-Suprarrenal/metabolismo , Prolactina/fisiología , Factor de Transcripción STAT5/metabolismo
20.
Elife ; 82019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30694175

RESUMEN

Leptin regulates energy balance and also exhibits neurotrophic effects during critical developmental periods. However, the actual role of leptin during development is not yet fully understood. To uncover the importance of leptin in early life, the present study restored leptin signaling either at the fourth or tenth week of age in mice formerly null for the leptin receptor (LepR) gene. We found that some defects previously considered irreversible due to neonatal deficiency of leptin signaling, including the poor development of arcuate nucleus neural projections, were recovered by LepR reactivation in adulthood. However, LepR deficiency in early life led to irreversible obesity via suppression of energy expenditure. LepR reactivation in adulthood also led to persistent reduction in hypothalamic Pomc, Cartpt and Prlh mRNA expression and to defects in the reproductive system and brain growth. Our findings revealed that early defects in leptin signaling cause permanent metabolic, neuroendocrine and developmental problems.


Asunto(s)
Envejecimiento/genética , Regulación del Desarrollo de la Expresión Génica , Leptina/genética , Obesidad/genética , Receptores de Leptina/genética , Envejecimiento/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/crecimiento & desarrollo , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/patología , Metabolismo Energético/genética , Femenino , Gónadas/crecimiento & desarrollo , Gónadas/metabolismo , Gónadas/patología , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Hipotálamo/patología , Leptina/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Obesidad/metabolismo , Obesidad/patología , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Hormona Liberadora de Prolactina/genética , Hormona Liberadora de Prolactina/metabolismo , Receptores de Leptina/deficiencia , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA