Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 9(1): 4194, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305620

RESUMEN

Puberty is regulated by epigenetic mechanisms and is highly sensitive to metabolic and nutritional cues. However, the epigenetic pathways mediating the effects of nutrition and obesity on pubertal timing are unknown. Here, we identify Sirtuin 1 (SIRT1), a fuel-sensing deacetylase, as a molecule that restrains female puberty via epigenetic repression of the puberty-activating gene, Kiss1. SIRT1 is expressed in hypothalamic Kiss1 neurons and suppresses Kiss1 expression. SIRT1 interacts with the Polycomb silencing complex to decrease Kiss1 promoter activity. As puberty approaches, SIRT1 is evicted from the Kiss1 promoter facilitating a repressive-to-permissive switch in chromatin landscape. Early-onset overnutrition accelerates these changes, enhances Kiss1 expression and advances puberty. In contrast, undernutrition raises SIRT1 levels, protracts Kiss1 repression and delays puberty. This delay is mimicked by central pharmacological activation of SIRT1 or SIRT1 overexpression, achieved via transgenesis or virogenetic targeting to the ARC. Our results identify SIRT1-mediated inhibition of Kiss1 as key epigenetic mechanism by which nutritional cues and obesity influence mammalian puberty.


Asunto(s)
Epigénesis Genética , Kisspeptinas/genética , Fenómenos Fisiológicos de la Nutrición , Obesidad/metabolismo , Maduración Sexual , Sirtuina 1/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Cromatina/metabolismo , Femenino , Histonas/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Ratones Transgénicos , Modelos Biológicos , Neuronas/metabolismo , Estado Nutricional , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Factores de Tiempo
2.
J Neuroendocrinol ; 30(2)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28675783

RESUMEN

Pro-opiomelanocortin (POMC) neurones within the hypothalamic arcuate nucleus are vital anorexigenic neurones. Both the insulin receptor and leptin receptor are coupled to activation of phosphatidylinositide-3 kinase (PI3K) to regulate multiple functions that increase POMC neuronal excitability. Using whole-cell recording in several mammalian species, we have found that both insulin and leptin depolarised POMC neurones via activation of transient receptor potential (TRPC)5 channels. TRPC5 channels have been rigorously characterised as the downstream effector based on their biophysical properties, pharmacological profile, and localisation by immunocytochemistry and single-cell reverse transcriptase-polymerase chain reaction. By contrast, insulin and leptin hyperpolarise and inhibit neuropeptide Y/agouti-related peptide neurones via activation of KATP channels. As proof of principle, insulin given i.c.v. robustly inhibits food intake and increases O2 utilisation, CO2 production and metabolic heat production. Therefore, these findings indicate that the depolarisation/excitation of POMC neurones by insulin and leptin is preserved across mammalian species and the activation of TRPC5 channels is likely a major mechanism by which insulin and leptin regulate energy homeostasis in mammals.


Asunto(s)
Canales de Calcio/metabolismo , Hipotálamo/efectos de los fármacos , Insulina/farmacología , Leptina/farmacología , Neuronas/efectos de los fármacos , Proopiomelanocortina/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Neuronas/metabolismo
3.
Steroids ; 81: 31-5, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24269736

RESUMEN

Two populations of cells within the hypothalamus exert opposite actions on food intake: proopiomelanocortin (POMC) neurons decrease it, while neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons increase it. 17ß-Estradiol (E2) is a potent anorexigenic hormone that exerts both genomic and non-genomic, rapid actions on these metabolic neurons. This review focuses on the rapid membrane effects of E2 in both POMC and NPY/AgRP neurons and how these combined effects mediate the anorexigenic effects of this steroid.


Asunto(s)
Ingestión de Alimentos , Estradiol/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Regulación de la Temperatura Corporal , Metabolismo Energético , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Retroalimentación Fisiológica , Homeostasis , Humanos , Hipotálamo/citología , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Proopiomelanocortina , Transducción de Señal
4.
Am J Physiol Endocrinol Metab ; 305(5): E632-40, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23820624

RESUMEN

Besides its quintessential role in reproduction, 17ß-estradiol (E2) is a potent anorexigenic hormone. E2 and the selective Gq-coupled membrane estrogen receptor (Gq-mER) ligand STX rapidly increase membrane excitability in proopiomelanocortin (POMC) neurons by desensitizing the coupling of GABAB receptors to G protein-coupled inwardly rectifying K(+) channels (GIRKs), which upon activation elicit a hyperpolarizing outward current. However, it is unknown whether E2 and STX can modulate GABAB signaling in neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons. We used single-cell RT-PCR and whole cell patch clamping with selective pharmacological reagents to show that NPY/AgRP cells of mice express the GABAB-R1 and -R2 receptors and are hyperpolarized by the GABAB agonist baclofen in an E2-dependent manner. In males, E2 rapidly attenuated the coupling of GABAB receptors to GIRKs, which was blocked by the general PI3K inhibitors wortmannin and LY-294002 or the selective p110ß subunit inhibitor TGX-221. The ERα-selective agonist propyl pyrazole triol mimicked the effects of E2. STX, in contrast, enhanced the GABAB response in males, which was abrogated by the estrogen receptor (ER) antagonist ICI 182,780. In gonadectomized mice of both sexes, E2 enhanced or attenuated the GABAB response in different NPY/AgRP cells. Coperfusing wortmannin with E2 or simply applying STX always enhanced the GABAB response. Thus, in NPY/AgRP neurons, activation of the Gq-mER by E2 or STX enhances the GABAergic postsynaptic response, whereas activation of ERα by E2 attenuates it. These findings demonstrate a clear functional dichotomy of rapid E2 membrane-initiated signaling via ERα vs. Gq-mER in a CNS neuron vital for regulating energy homeostasis.


Asunto(s)
Acrilamidas/farmacología , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Receptores de GABA-B/metabolismo , Acrilamidas/metabolismo , Animales , Baclofeno/farmacología , Estradiol/metabolismo , Femenino , Agonistas de Receptores GABA-B/farmacología , Cobayas , Masculino , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp , Transducción de Señal
5.
Am J Physiol Endocrinol Metab ; 302(11): E1399-406, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22436698

RESUMEN

Hypothalamic proopiomelanocortin (POMC) neurons are controlled by many central signals, including serotonin. Serotonin increases POMC activity and reduces feeding behavior via serotonion [5-hydroxytryptamine (5-HT)] receptors by modulating K(+) currents. A potential K(+) current is the M-current, a noninactivating, subthreshold outward K(+) current. Previously, we found that M-current activity was highly reduced in fasted vs. fed states in neuropeptide Y neurons. Because POMC neurons also respond to energy states, we hypothesized that fasting may alter the M-current and/or its modulation by serotonergic input to POMC neurons. Using visualized-patch recording in neurons from fed male enhanced green fluorescent protein-POMC transgenic mice, we established that POMC neurons expressed a robust M-current (102.1 ± 6.7 pA) that was antagonized by the selective KCNQ channel blocker XE-991 (40 µM). However, the XE-991-sensitive current in POMC neurons did not differ between fed and fasted states. To determine if serotonin suppresses the M-current via the 5-HT(2C) receptor, we examined the effects of the 5-HT(2A)/5-HT(2C) receptor agonist 2,5-dimethoxy-4-iodoamphetamine (DOI) on the M-current. Indeed, DOI attenuated the M-current by 34.5 ± 6.9% and 42.0 ± 5.3% in POMC neurons from fed and fasted male mice, respectively. In addition, the 5-HT(1B)/5-HT(2C) receptor agonist m-chlorophenylpiperazine attenuated the M-current by 42.4 ± 5.4% in POMC neurons from fed male mice. Moreover, the selective 5-HT(2C) receptor antagonist RS-102221 abrogated the actions of DOI in suppressing the M-current. Collectively, these data suggest that although M-current expression does not differ between fed and fasted states in POMC neurons, serotonin inhibits the M-current via activation of 5-HT(2C) receptors to increase POMC neuronal excitability and, subsequently, reduce food intake.


Asunto(s)
Hipotálamo/metabolismo , Canales de Potasio KCNQ/antagonistas & inhibidores , Neuronas/metabolismo , Proopiomelanocortina/fisiología , Receptor de Serotonina 5-HT2C/metabolismo , Anfetaminas/farmacología , Animales , Antracenos/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Ayuno/fisiología , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Piperazinas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Receptor de Serotonina 5-HT2C/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Compuestos de Espiro/farmacología , Sulfonamidas/farmacología
6.
J Neuroendocrinol ; 24(1): 122-30, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21592235

RESUMEN

17ß-Oestradiol (E(2)) is essential for cyclical gonadotrophin-releasing hormone (GnRH) neuronal activity and secretion. In particular, E(2) increases the excitability of GnRH neurones during the afternoon of pro-oestrus in the rodent, which is associated with increased synthesis and secretion of GnRH. It is well established that E(2) regulates the activity of GnRH neurones through both presynaptic and postsynaptic mechanisms. E(2) significantly modulates the mRNA expression of numerous ion channels in GnRH neurones and alters the associated endogenous conductances, including potassium (K(ATP) , A-type) currents and low-voltage T-type and high-voltage L-type calcium currents. Notably, K(ATP) channels are critical for maintaining GnRH neurones in a hyperpolarised state for recruiting the T-type calcium channels, which are important for burst firing in GnRH neurones. In addition, there are other critical channels contributing to burst firing pattern, including the small conductance Ca(2+) -activated K(+) channels that may be modulated by E(2) . Despite these advances, the cellular mechanisms underlying the cyclical GnRH neuronal activity and GnRH release are largely unknown. Ultimately, the ensemble of both pre- and postsynaptic targets of the actions of E(2) will dictate the excitability and activity pattern of GnRH neurones.


Asunto(s)
Potenciales de Acción/fisiología , Canales de Calcio Tipo T/metabolismo , Estradiol/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/metabolismo , Animales
7.
Endocrinology ; 152(11): 4265-75, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21914775

RESUMEN

Kisspeptin (Kiss1) and neurokinin B (NKB) (encoded by the Kiss1 and Tac2 genes, respectively) are indispensable for reproduction. In the female of many species, Kiss1 neurons in the arcuate nucleus (ARC) coexpress dynorphin A and NKB. Such cells have been termed Kiss1/NKB/Dynorphin (KNDy) neurons, which are thought to mediate the negative feedback regulation of GnRH/LH secretion by 17ß-estradiol. However, we have less knowledge about the molecular physiology and regulation of Kiss1/Kiss1-expressing neurons in the ARC of the male. Our work focused on the adult male mouse, where we sought evidence for coexpression of these neuropeptides in cells in the ARC, assessed the role of Kiss1 neurons in negative feedback regulation of GnRH/LH secretion by testosterone (T), and investigated the action of NKB on KNDy and GnRH neurons. Results showed that 1) the mRNA encoding Kiss1, NKB, and dynorphin are coexpressed in neurons located in the ARC; 2) Kiss1 and dynorphin A mRNA are regulated by T through estrogen and androgen receptor-dependent pathways; 3) senktide, an agonist for the NKB receptor (neurokinin 3 receptor, encoded by Tacr3), stimulates gonadotropin secretion; 4) KNDy neurons express Tacr3, whereas GnRH neurons do not; and 5) senktide activates KNDy neurons but has no discernable effect on GnRH neurons. These observations corroborate the putative role for KNDy neurons in mediating the negative feedback effects of T on GnRH/LH secretion and provide evidence that NKB released from KNDy neurons is part of an auto-feedback loop that generates the pulsatile secretion of Kiss1 and GnRH in the male.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neuroquinina B/metabolismo , Neuronas/metabolismo , Animales , Dinorfinas/metabolismo , Retroalimentación Fisiológica/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Masculino , Ratones , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Transducción de Señal/fisiología
8.
Endocrinology ; 152(8): 3182-91, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21628385

RESUMEN

Previously, we reported that 1 nM 17ß-estradiol (E(2)) induces a rapid action, which is, in part, mediated through the G protein-coupled receptor GPR30 in primate GnRH neurons. Because it has been reported that the diphenylacrylamide compound, STX, causes estrogenic action in the mouse and guinea pig hypothalamus, the present study examined effects of STX in primate GnRH neurons and whether there is an action independent of GPR30. Results are summarized as follows. STX (10 nM) exposure increased 1) the oscillation frequency of intracellular calcium concentration ([Ca(2+)](i)), 2) the percentage of cells stimulated, and 3) the synchronization frequency of [Ca(2+)](i) oscillations. STX (10-100 nM) also stimulated GnRH release. The effects of STX on both [Ca(2+)](i) oscillations and GnRH release were similar to those caused by E(2) (1 nM), although with less magnitude. STX (10 nM)-induced changes in [Ca(2+)](i) oscillations were not altered by GPR30 small interfering RNA transfection, indicating that STX-sensitive receptors differ from GPR30. Finally, a higher dose of E(2) (10 nM) induced a larger change in [Ca(2+)](i) oscillations than that with a smaller dose of E(2) (1 nM), and the effects of 10 nM E(2) were reduced but not completely blocked by GPR30 small interfering RNA transfection, indicating that the effects of 10 nM E(2) in primate GnRH neurons are mediated by multiple membrane receptors, including GPR30 and STX-sensitive receptors. Collectively, the rapid action of E(2) mediated through GPR30 differs from that mediated through STX-sensitive receptors. The molecular structure of the STX-sensitive receptor remains to be identified.


Asunto(s)
Acrilamidas/farmacología , Calcio/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/efectos de los fármacos , Animales , Estradiol/análogos & derivados , Estradiol/farmacología , Fulvestrant , Macaca mulatta , Neuronas/metabolismo , Receptores de Estrógenos , Receptores Acoplados a Proteínas G/fisiología
9.
J Neuroendocrinol ; 21(2): 108-22, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19187398

RESUMEN

The excitatory tone to gonadotrophin-releasing hormone (GnRH) neurones is a critical component underlying the pubertal increase in GnRH secretion. However, the homeostatic mechanisms modulating the response of GnRH neurones to excitatory inputs remain poorly understood. A basic mechanism of neuronal homeostasis is the Na(+),K(+)-ATPase-dependent restoration of Na(+) and K(+) transmembrane gradients after neuronal excitation. This activity is reduced in a mouse model of Rett syndrome (RTT), a neurodevelopmental disorder in which expression of FXYD1, a modulator of Na(+),K(+)-ATPase activity, is increased. We now report that the initiation, but not the completion of puberty, is advanced in girls with RTT, and that, in rodents, FXYD1 may contribute to the neuroendocrine regulation of female puberty by modulating GnRH neuronal excitability. Fxyd1 mRNA abundance reaches maximal levels in the female rat hypothalamus by the fourth postnatal week of life (i.e., around the time when the mode of GnRH secretion acquires an adult pattern of release). Although Fxyd1 mRNA expression is low in the hypothalamus, approximately 50% of GnRH neurones contain Fxyd1 transcripts. Whole-cell patch recording of GnRH-EGFP neurones revealed that the neurones of Fxyd1-null female mice respond to somatic current injections with a lower number of action potentials than wild-type cells. Both the age at vaginal opening and at first oestrous were delayed in Fxyd1(-/-) mice, but adult reproductive capacity was normal. These results suggest that FXYD1 contributes to facilitating the advent of puberty by maintaining GnRH neuronal excitability to incoming transsynaptic stimulatory inputs.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/fisiología , Fosfoproteínas/metabolismo , Desarrollo Sexual/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Potenciales de Acción/fisiología , Adolescente , Animales , Niño , Preescolar , Femenino , Humanos , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Técnicas de Placa-Clamp , Fosfoproteínas/genética , Pubertad/fisiología , Ratas
10.
J Neuroendocrinol ; 21(4): 263-70, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19187465

RESUMEN

It is increasingly evident that 17beta-oestradiol (E(2)), via a distinct membrane oestrogen receptor (Gq-mER), can rapidly activate kinase pathways to have multiple downstream actions in central nervous system (CNS) neurones. We have found that E(2) can rapidly reduce the potency of the GABA(B) receptor agonist baclofen and mu-opioid receptor agonist DAMGO to activate G-protein-coupled, inwardly rectifying K(+) (GIRK) channels in hypothalamic neurones, thereby increasing the excitability (firing activity) of pro-opiomelanocortin (POMC) and dopamine neurones. These effects are mimicked by the membrane impermeant E(2)-BSA and a new ligand (STX) that is selective for the Gq-mER that does not bind to ERalpha or ERbeta. Both E(2) and STX are fully efficacious in attenuating the GABA(B) response in ERalpha, ERbeta and GPR 30 knockout mice in an ICI 182 780 reversible manner. These findings are further proof that E(2) signals through a unique plasma membrane ER. We have characterised the coupling of this Gq-mER to a Gq-mediated activation of phospholipase C leading to the up-regulation of protein kinase Cdelta and protein kinase A activity in these neurones, which ultimately alters gene transcription. Finally, as proof of principle, we have found that STX, similar to E(2), reduces food intake and body weight gain in ovariectomised females. STX, presumably via the Gq-mER, also regulates gene expression of a number of relevant targets including cation channels and signalling molecules that are critical for regulating (as a prime example) POMC neuronal excitability. Therefore, E(2) can activate multiple receptor-mediated pathways to modulate excitability and gene transcription in CNS neurones that are critical for controlling homeostasis and motivated behaviors.


Asunto(s)
Estradiol/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Hipotálamo/fisiología , Neuronas/fisiología , Receptores de Estrógenos/metabolismo , Transducción de Señal/fisiología , Animales , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Metabolismo Energético/fisiología , Estrógenos/metabolismo , Expresión Génica/fisiología , Homeostasis/fisiología , Transducción de Señal/genética
11.
J Neuroendocrinol ; 20(12): 1310-8, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19094079

RESUMEN

Tibolone is primarily used for the treatment of climacteric symptoms. Tibolone is rapidly converted into three major metabolites: 3 alpha- and 3beta-hydroxy (OH)-tibolone, which have oestrogenic effects, and the Delta 4-isomer (Delta 4-tibolone), which has progestogenic and androgenic effects. Because tibolone is effective in treating climacteric symptoms, the effects on the brain may be explained by the oestrogenic activity of tibolone. Using whole-cell patch clamp recording, we found previously that 17beta-oestradiol (E(2)) rapidly altered gamma-aminobutyric acid (GABA) neurotransmission in hypothalamic neurones through a membrane oestrogen receptor (mER). E(2) reduced the potency of the GABA(B) receptor agonist baclofen to activate G-protein-coupled, inwardly rectifying K(+) (GIRK) channels in hypothalamic neurones. Therefore, we hypothesised that tibolone may have some rapid effects through the mER and sought to elucidate the signalling pathway of tibolone's action using selective inhibitors and whole cell recording in ovariectomised female guinea pigs and mice. A sub-population of neurones was identified post hoc as pro-opiomelanocortin (POMC) neurones by immunocytochemical staining. Similar to E(2), we have found that tibolone and its active metabolite 3 beta OH-tibolone rapidly reduced the potency of the GABA(B) receptor agonist baclofen to activate GIRK channels in POMC neurones. The effects were blocked by the ER antagonist ICI 182 780. Other metabolites of tibolone (3 alpha OH-tibolone and Delta 4-tibolone) had no effect. Furthermore, tibolone (and 3 beta OH-tibolone) was fully efficacious in ER alpha knockout (KO) and ER beta KO mice to attenuate GABA(B) responses. The effects of tibolone were blocked by phospholipase C inhibitor U73122. However, in contrast to E(2), the effects of tibolone were not blocked by protein kinase C inhibitors or protein kinase A inhibitors. It appears that tibolone (and 3 beta OH-tibolone) activates phospholipase C leading to phosphatidylinositol bisphosphate metabolism and direct alteration of GIRK channel function. Therefore, tibolone may enhance synaptic efficacy through the G(q) signalling pathways of mER in brain circuits that are critical for maintaining homeostatic functions.


Asunto(s)
Moduladores de los Receptores de Estrógeno/metabolismo , Hipotálamo/citología , Neuronas/metabolismo , Norpregnenos/metabolismo , Receptores de GABA-B/metabolismo , Animales , Baclofeno/metabolismo , Estrenos/metabolismo , Moduladores de los Receptores de Estrógeno/química , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Agonistas del GABA/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Cobayas , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Neuronas/citología , Norpregnenos/química , Técnicas de Placa-Clamp , Pirrolidinonas/metabolismo , Transducción de Señal/fisiología , Fosfolipasas de Tipo C/antagonistas & inhibidores , Ácido gamma-Aminobutírico/metabolismo
12.
Endocrinology ; 149(12): 6113-24, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18755790

RESUMEN

During the reproductive cycle, fluctuations in circulating estrogens affect multiple homeostatic systems controlled by hypothalamic neurons. Two of these neuronal populations are arcuate proopiomelanocortin and neuropeptide Y neurons, which control energy homeostasis and feeding. Estradiol modulates these neurons either through the classical estrogen receptors (ERs) to control gene transcription or through a G protein-coupled receptor (mER) activating multiple signaling pathways. To differentiate between these two divergent ER-mediated mechanisms and their effects on homeostasis, female guinea pigs were ovariectomized and treated systemically with vehicle, estradiol benzoate (EB) or STX, a selective mER agonist, for 4 wk, starting 7 d after ovariectomy. Individual body weights were measured after each injection day for 28 d, at which time the animals were euthanized, and the arcuate nucleus was microdissected. As predicted, the body weight gain was significantly lower for EB-treated females after d 5 and for STX-treated females after d 12 compared with vehicle-treated females. Total arcuate RNA was extracted from all groups, but only the vehicle and STX-treated samples were prepared for gene microarray analysis using a custom guinea pig gene microarray. In the arcuate nucleus, 241 identified genes were significantly regulated by STX, several of which were confirmed by quantitative real-time PCR and compared with EB-treated groups. The lower weight gain of EB-treated and STX-treated females suggests that estradiol controls energy homeostasis through both ERalpha and mER-mediated mechanisms. Genes regulated by STX indicate that not only does it control neuronal excitability but also alters gene transcription via signal transduction cascades initiated from mER activation.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético/fisiología , Estrógenos/metabolismo , Homeostasis/fisiología , Transducción de Señal/fisiología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Estradiol/análogos & derivados , Estradiol/farmacología , Estrona/análogos & derivados , Estrona/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Cobayas , Homeostasis/efectos de los fármacos , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovariectomía , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
13.
J Neuroendocrinol ; 19(11): 847-59, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17927663

RESUMEN

Although it is well established that gonadotrophin-releasing hormone (GnRH) neurones and astrocytes maintain an intimate contact throughout development and adult life, the cell-surface molecules that may contribute to this adhesiveness remain largely unknown. In the peripheral nervous system, the glycosylphosphatidyl inositol (GPI)-anchored protein contactin is a cell-surface neuronal protein required for axonal-glial adhesiveness. A glial transmembrane protein recognised by neuronal contactin is receptor-like protein tyrosine phosphatase beta (RPTP beta), a phosphatase with structural similarities to cell adhesion molecules. In the present study, we show that contactin, and its preferred in cis partner Caspr1, are expressed in GnRH neurones. We also show that the RPTP beta mRNA predominantly expressed in hypothalamic astrocytes encodes an RPTP beta isoform (short RPTP beta) that uses its carbonic anhydrase (CAH) extracellular subdomain to interact with neuronal contactin. Immunoreactive contactin is most abundant in GnRH nerve terminals projecting to both the organum vasculosum of the lamina terminalis and median eminence, implying GnRH axons as an important site of contactin-dependent cell adhesiveness. GT1-7 immortalised GnRH neurones adhere to the CAH domain of RPTPbeta, and this adhesiveness is blocked when contactin GPI anchoring is disrupted or contactin binding capacity is immunoneutralised, suggesting that astrocytic RPTP beta interacts with neuronal contactin to mediate glial-GnRH neurone adhesiveness. Because the abundance of short RPTP beta mRNA increases in the female mouse hypothalamus (but not in the cerebral cortex) before puberty, it appears that an increased interaction between GnRH axons and astrocytes mediated by RPTP beta-contactin is a dynamic mechanism of neurone-glia communication during female sexual development.


Asunto(s)
Astrocitos/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Neuronas/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Animales , Astrocitos/citología , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Contactinas , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/citología , Hipotálamo/fisiología , Ratones , Neuronas/citología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
14.
Neuroscience ; 133(1): 343-52, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15893655

RESUMEN

Physiological doses of 17-beta Estradiol (E2) rapidly induce mitogen-activated protein kinase (MAPK) phosphorylation in a variety of cell culture and tissue explant preparations. Rapid MAPK phosphorylation has been implicated as a critical step in estrogen's effects on neuronal activity, gene transcription and neuroprotection. The present series of in vivo experiments were designed to determine whether acute administration of estrogen rapidly increased extracellular signal-regulated protein kinase (ERK) 2 phosphorylation. Brains were harvested 20 min after a single i.p. injection of 15 microg/kg of 17-beta or 17-alpha estradiol. Twelve brain structures were micro-dissected, homogenized and processed for Western blotting. E2-treated rats exhibited a statistically significant increase in ERK2 phosphorylation in the diagonal band of Broca, rostral nucleus accumbens, paraventricular nucleus, arcuate nucleus and anteromedial visual cortex. Administration of the same dose of 17-alpha estradiol did not enhance ERK phosphorylation in any of the brain regions examined. The in vivo data presented here extend previously published in vitro data indicating that E2 rapidly activates MAPK in primary neuronal cultures, explants and cell lines. These data also indicate that MAPK activation is a potential mediator of estrogens effects in some but not all estrogen receptor containing regions of the brain.


Asunto(s)
Encéfalo/enzimología , Estradiol/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Algoritmos , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Estradiol/administración & dosificación , Estrógenos/sangre , Femenino , Ovariectomía , Fosforilación , Área Preóptica/fisiología , Ratas
15.
J Pharmacol Exp Ther ; 299(1): 21-30, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11561059

RESUMEN

The present study sought to determine whether small-conductance, Ca2+-activated K+ currents underlie the afterhyperpolarization (AHP) in neurons of the preoptic area (POA), a brain region important in controlling reproduction. We used an ovariectomized, female guinea pig model to test two hypotheses: 1) the current associated with the AHP (I(AHP)) regulates the firing rate of POA neurons and 2) amine neurotransmitters modulate it in a gonadal steroid-sensitive manner. Intracellular recordings followed by combined histofluorescence/in situ hybridization for glutamic acid decarboxylase, 65-kDa isomer, revealed that POA neurons, including gamma-aminobutyric acid (GABA)ergic neurons, exhibited an AHP and spike frequency adaptation. The corresponding I(AHP) was sensitive to antagonism by CdCl2 (200 microM), apamin (0.3-1 microM), and dequalinium (3 microM). The beta-adrenergic receptor agonist isoproterenol inhibited the I(AHP) in a dose-dependent, timolol-sensitive fashion. In addition, the alpha1-adrenergic receptor agonist methoxamine dose dependently inhibited the I(AHP) in a prazosin-sensitive manner and increased neuronal firing rate. Twenty-four-hour pretreatment with estradiol benzoate (EB; 25 microg, s.c.) markedly potentiated the inhibitory effect of methoxamine on the I(AHP), whereas that for isoproterenol was unaffected. Similarly, bath application of 17beta-estradiol (100 nM; 15-20 min) mimicked the effect of EB on the methoxamine-induced inhibition of the I(AHP). Thus, POA GABAergic neurons express an apamin-sensitive channel that mediates, at least in part, the I(AHP), and tempers the excitability of these cells. Furthermore, these studies demonstrate that estrogen enhances the alpha1-adrenergic receptor-mediated inhibition of this current.


Asunto(s)
Apamina/farmacología , Estrógenos/farmacología , Hipotálamo/fisiología , Neuronas/metabolismo , Norepinefrina/fisiología , Canales de Potasio Calcio-Activados , Canales de Potasio/efectos de los fármacos , Reproducción/efectos de los fármacos , Animales , Femenino , Colorantes Fluorescentes , Glutamato Descarboxilasa/metabolismo , Cobayas , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Área Preóptica/citología , Área Preóptica/efectos de los fármacos , Área Preóptica/fisiología , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Receptores Adrenérgicos alfa 1/fisiología , Receptores Adrenérgicos beta/efectos de los fármacos , Receptores Adrenérgicos beta/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio , Ácido gamma-Aminobutírico/fisiología
16.
J Neurosci ; 21(6): 2085-93, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11245692

RESUMEN

The principal role of estrogen is its control of the female ovulatory cycle via negative and positive feedback on gonadotropin secretion. However, a detailed, cohesive picture of how the steroid specifically regulates the excitability of hypothalamic neurons involved in the central control of gonadotropin secretion is still emerging. Here, we used an ovariectomized female guinea pig model to test the hypothesis that estrogen acts on GABAergic neurons in the preoptic area (POA) to elicit a biphasic profile of luteinizing hormone (LH) secretion. Intracellular electrophysiological recordings revealed that estradiol benzoate (EB; 25 microgram, s.c.) decreased the hyperpolarizing response of GABAergic neurons to the GABA(B) receptor agonist baclofen 24 hr after treatment. This effect of GABA(B) receptor stimulation in unidentified POA neurons was still depressed 42 hr after EB administration. By the use of a ribonuclease protection assay, however, EB reduced glutamic acid decarboxylase mRNA expression 42 hr but not 24 hr after its administration. Thus, estrogen attenuated the autoinhibition of GABAergic POA neurons during the initial LH suppressive (i.e., negative feedback) phase and subsequently reduced GABAergic function during the LH surge (i.e., positive feedback). These studies demonstrate that the effects of estrogen on hypothalamic GABAergic neurons coincide with the inhibitory and stimulatory actions, respectively, of the steroid on LH secretion. Furthermore, the data provide novel insights into the mechanism by which estrogen regulates hypothalamic GABAergic neurons, which are critical for the biphasic modulation of LH release observed over the course of the female ovulatory cycle.


Asunto(s)
Estrógenos/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Relación Dosis-Respuesta a Droga , Estradiol/análogos & derivados , Estradiol/farmacología , Retroalimentación , Femenino , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Agonistas de Receptores GABA-B , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Cobayas , Inmunohistoquímica , Técnicas In Vitro , Isoenzimas/genética , Isoenzimas/metabolismo , Hormona Luteinizante/sangre , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ovariectomía , Canales de Potasio/química , Canales de Potasio/metabolismo , Área Preóptica/citología , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , ARN Mensajero/metabolismo , Tetrodotoxina/farmacología
17.
Neuroendocrinology ; 72(4): 208-17, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11070424

RESUMEN

The endogenous opioid neurotransmitter beta-endorphin (beta-END), a product of the proopiomelanocortin (POMC) gene, is strongly implicated in the control of the female reproductive cycle, stress responses, and antinociception. Using selective gene targeting, we have generated a strain of mice that do not express any beta-END. These mice exhibit both normal reproduction and normal basal and stress-induced hypothalamic-pituitary-axis activity, but exhibit a significantly attenuated opioid-mediated stress-induced analgesia. To further understand the cellular bases of these responses, we have studied mediobasal hypothalamic (MBH) neurons, including POMC neurons, using whole-cell patch recording in an in vitro slice preparation. Twenty-seven MBH cells were recorded in wild-type and 25 MBH cells were recorded in beta-END knockout mice. Neurons from both genotypes showed a significant positive correlation between DAMGO concentration (from 30 nM to 10 microM) and the induced outward K(+) current. The genotypes did not differ, however, in either the DAMGO-induced maximum outward current response or EC(50), or for the maximal response to the GABA(B) agonist baclofen. Furthermore, quantitative receptor autoradiography utilizing (3)H-DAMGO did not reveal any differences in total mu-opioid receptor binding between genotypes. Therefore, we conclude that the complete absence of beta-END throughout development did not alter either the expression of mu-opioid receptors or their coupling to K(+) channels in MBH neurons.


Asunto(s)
Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Hipotálamo Medio/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores Opioides mu/agonistas , betaendorfina/fisiología , Animales , Baclofeno/farmacología , Relación Dosis-Respuesta a Droga , Electrofisiología , Femenino , Agonistas del GABA/farmacología , Hipotálamo Medio/citología , Hipotálamo Medio/fisiología , Ratones , Ratones Noqueados/genética , Neuronas/fisiología , Canales de Potasio/efectos de los fármacos , Canales de Potasio/fisiología , Proopiomelanocortina/metabolismo , Receptores Opioides mu/metabolismo , Valores de Referencia , betaendorfina/genética
18.
Brain Res ; 879(1-2): 29-41, 2000 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-11011003

RESUMEN

Whole-cell and intracellular recordings were made in coronal hypothalamic slices prepared from ovariectomized female guinea pigs. 62% of preoptic area (POA) neurons fired action potentials in a bursting manner, and exhibited a significantly greater afterhyperpolarization (AHP) than did non-bursting POA neurons. The majority (70%) of POA neurons (n=76) displayed a time-dependent inward rectification (I(h)) that was blocked by CsCl (3 mM) or by ZD 7288 (30 microM). In addition, 51% of the cells expressed a low-threshold spike (LTS) associated with a transient inward current (I(T)) that was blocked by NiCl(2) (200 microM). A smaller percentage of POA neurons (29%) expressed a transient outward, A-type K(+) current that was antagonized by a high concentration of 4-aminopyridine (3 mM). Moreover, POA neurons responded to bath application of the mu-opioid receptor agonist DAMGO (93%) or the GABA(B) receptor agonist baclofen (83%) with a membrane hyperpolarization or an outward current. These responses were accompanied by a decrease in input resistance or an increase in conductance, respectively, and were attenuated by BaCl(2) (100 microM). In addition, the reversal potential for these responses closely approximated the Nernst equilibrium potential for K(+). These results suggest that POA neurons endogenously express to varying degrees an AHP, an I(h), an I(T) and an A-type K(+) current. The vast majority of these neurons also are inhibited upon mu-opioid or GABA(B) receptor stimulation via the activation of an inwardly-rectifying K(+) conductance. Such intrinsic and transmitter-activated conductances likely serve as important determinants of the firing patterns of POA neurons.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Neuronas/fisiología , Área Preóptica/fisiología , 4-Aminopiridina/farmacología , Potenciales de Acción/fisiología , Animales , Baclofeno/farmacología , Compuestos de Bario/farmacología , Cesio/farmacología , Cloruros/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Cobayas , Técnicas In Vitro , Potenciales de la Membrana , Neuronas/efectos de los fármacos , Níquel/farmacología , Ovariectomía , Técnicas de Placa-Clamp , Canales de Potasio/efectos de los fármacos , Canales de Potasio/fisiología , Área Preóptica/efectos de los fármacos , Pirimidinas/farmacología , Tiempo de Reacción , Tetrodotoxina/farmacología
19.
Neuroreport ; 10(12): 2681-7, 1999 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-10574392

RESUMEN

We combined histofluorescence with in situ hybridization to identify GABAergic neurons in the arcuate nucleus (ARC) following electrophysiological recording, using GAD65 as a marker. Intracellular recordings 91 were made in hypothalamic slices prepared from ovariectomized guinea pigs. Over 90% of ARC neurons tested with the GABA(B) receptor agonist baclofen responded with a membrane hyperpolarization or an outward current. The hyperpolarization was dose dependent, and the GABA(B) receptor antagonist CGP 35,348 produced a rightward shift in the agonist dose-response curve. Agonist potency was lower, and the efficacy greater, in GAD-positive neurons. The use of this novel technique for identifying GABAergic neurons thus reveals differences in the pharmacodynamics of GABA(B) receptor activation between GABAergic and non-GABAergic ARC neurons.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Inhibición Neural , Neuronas/fisiología , Receptores de GABA-B/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Núcleo Arqueado del Hipotálamo/citología , Baclofeno/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Glutamato Descarboxilasa/análisis , Cobayas , Hibridación in Situ , Técnicas In Vitro , Compuestos Organofosforados/farmacología
20.
J Neurosci ; 19(20): 8966-78, 1999 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-10516315

RESUMEN

Cocaine is a highly addictive drug that binds to the dopamine transporter (DAT), inhibits the reuptake of dopamine, and initiates multiple actions within midbrain dopaminergic systems. Using the rhesus monkey, we have investigated the consequences of in utero cocaine exposure on the expression of DAT in the fetal brain. By using the selective DAT ligand [125I]RTI-121 and tyrosine hydroxylase (TH) immunocytochemistry, we found that DAT binding sites are highly developed by day 70 of gestation and show a distribution pattern similar to TH. The rank order of specific 3beta-(4-[125I]iodophenyl)tropane-2beta-carboxylic acid isopropyl ester ([125I]RTI-121) binding densities was substantia nigra-ventral tegmental area > putamen > caudate > lateral hypothalamus > accumbens > linear/interfascicular nuclei >/= globus pallidus > prefrontal cortex. Furthermore, we observed that DAT mRNA was differentially expressed within fetal midbrain dopamine neurons with the highest levels detected in the ventral tier of the substantia nigra pars compacta, and the lowest levels in the ventral tegmental area and the linear/interfascicular nuclei. In utero cocaine exposure between days 22 and 70 significantly increased DAT mRNA expression, and the density of [125I]RTI-121 binding sites within midbrain dopamine neurons in the 70-d-old fetus. This increased DAT expression is accompanied by other presynaptic and postsynaptic neuronal changes, which collectively suggest that midbrain dopamine neurons are hypoactive after prolonged cocaine exposure, a state that may be a contributing factor in the development of attention deficit disorders observed in subjects exposed prenatally to cocaine.


Asunto(s)
Encéfalo/embriología , Proteínas Portadoras/metabolismo , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Feto/metabolismo , Glicoproteínas de Membrana , Proteínas de Transporte de Membrana , Proteínas del Tejido Nervioso , Animales , Sitios de Unión/efectos de los fármacos , Proteínas Portadoras/genética , Cocaína/análogos & derivados , Cocaína/farmacocinética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Desarrollo Embrionario y Fetal , Feto/citología , Feto/fisiología , Macaca mulatta/embriología , Mesencéfalo/citología , Mesencéfalo/metabolismo , Neuronas/metabolismo , ARN Mensajero/metabolismo , Distribución Tisular , Tirosina 3-Monooxigenasa/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...