Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
STAR Protoc ; 2(3): 100787, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34485946

RESUMEN

The hypothalamic magnocellular neuroendocrine cells (MNCs) project to the posterior pituitary (PPi), regulating reproduction and fluid homeostasis. It has been challenging to selectively label and manipulate MNCs, as they are intermingled with parvocellular neuroendocrine cells projecting to the median eminence. Here, we provide a step-by-step protocol for specifically targeting the MNCs by infusing retrograde viral tracers into the PPi. When combined with optogenetics, chemogenetics, and transgenic animals, this approach allows cell-type-specific manipulation of MNCs in multiple sites for functional dissection. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2021) and Tang et al. (2020).


Asunto(s)
Hipotálamo/citología , Células Neuroendocrinas , Optogenética/métodos , Neurohipófisis/citología , Animales , Animales Modificados Genéticamente , Masculino , Eminencia Media/citología , Red Nerviosa/citología , Red Nerviosa/fisiología , Células Neuroendocrinas/citología , Células Neuroendocrinas/fisiología , Ratas , Ratas Sprague-Dawley
2.
Psychoneuroendocrinology ; 113: 104549, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31884322

RESUMEN

Endogenous cannabinoids (endocannabinoids, eCB) are expressed throughout the body and contribute to regulation of the hypothalamo-pituitary-adrenal (HPA) axis and general stress reactivity. This study assessed the contributions of CB1 receptors (CB1R) in the modulation of basal and stress-induced neural and HPA axis activities. Catheterized adult male rats were placed in chambers to acclimate overnight, with their catheters connected and exteriorized from the chambers for relatively stress-free remote injections. The next morning, the CB1R antagonist AM251 (1 or 2 mg/kg) or vehicle was administered, and 30 min later, rats were exposed to loud noise stress (30 min) or no noise (basal condition). Blood, brains, pituitary and adrenal glands were collected immediately after the procedures for analysis of c-fos and CB1R mRNAs, corticosterone (CORT) and adrenocorticotropin hormone (ACTH) plasma levels. Basally, CB1R antagonism induced c-fos mRNA in the basolateral amygdala (BLA) and auditory cortex (AUD) and elevated plasma CORT, indicating disruption of eCB-mediated constitutive inhibition of activity. CB1R blockade also potentiated stress-induced hormone levels and c-fos mRNA in several regions such as the bed nucleus of the stria terminalis (BST), lateral septum (LS), and basolateral amygdala (BLA) and the paraventricular nucleus of the hypothalamus (PVN). CB1R mRNA was detected in all central tissues investigated, and the adrenal cortex, but at very low levels in the anterior pituitary gland. Interestingly, CB1R mRNA was rapidly and bidirectionally regulated in response to stress and/or antagonist treatment in some regions. eCBs therefore modulate the HPA axis by regulating both constitutive and activity-dependent inhibition at multiple levels.


Asunto(s)
Células Neuroendocrinas/fisiología , Receptor Cannabinoide CB1/fisiología , Corteza Suprarrenal/metabolismo , Glándulas Suprarrenales/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Corticosterona/sangre , Endocannabinoides/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/metabolismo , Masculino , Células Neuroendocrinas/efectos de los fármacos , Células Neuroendocrinas/metabolismo , Sistemas Neurosecretores/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Piperidinas/farmacología , Sistema Hipófiso-Suprarrenal/metabolismo , Proteínas Proto-Oncogénicas c-fos/sangre , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Restricción Física/psicología , Estrés Fisiológico/fisiología , Estrés Psicológico/fisiopatología
3.
Physiol Rep ; 7(8): e14047, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31008554

RESUMEN

Hypothalamic magnocellular neurosecretory cells (MNCs) undergo dramatic structural reorganization during lactation in female rats that is thought to contribute to the pulsatile secretion of oxytocin critical for milk ejection. MNCs from male rats generate robust bursts of GABAergic synaptic currents, a subset of which are onset-synchronized between MNC pairs, but the functional role of the IPSC bursts is not known. To determine the physiological relevance of IPSC bursts, we compared MNCs from lactating and non-lactating female rats using whole-cell recordings in brain slices. We recorded a sixfold increase in the incidence of IPSC bursts in oxytocin (OT)-MNCs from lactating rats compared to non-lactating rats, whereas there was no change in IPSC bursts in vasopressin (VP)-MNCs. Synchronized bursts of IPSCs were observed in pairs of MNCs in slices from lactating rats. Our data indicate, therefore, that IPSC bursts are upregulated specifically in OT-MNCs during lactation, and may, therefore, contribute via rebound depolarization to the spike trains in OT neurons that lead to reflex milk ejection.


Asunto(s)
Potenciales Postsinápticos Inhibidores , Lactancia/fisiología , Células Neuroendocrinas/fisiología , Oxitocina/metabolismo , Animales , Femenino , Hipotálamo/citología , Hipotálamo/metabolismo , Hipotálamo/fisiología , Lactancia/metabolismo , Células Neuroendocrinas/metabolismo , Ratas , Ratas Wistar , Vasopresinas/metabolismo
4.
Sci Rep ; 8(1): 2760, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29426889

RESUMEN

Engineered silica nanoparticles (NPs) have attracted increasing interest in several applications, and particularly in the field of nanomedicine, thanks to the high biocompatibility of this material. For their optimal and controlled use, the understanding of the mechanisms elicited by their interaction with the biological target is a prerequisite, especially when dealing with cells particularly vulnerable to environmental stimuli like neurons. Here we have combined different electrophysiological approaches (both at the single cell and at the population level) with a genomic screening in order to analyze, in GT1-7 neuroendocrine cells, the impact of SiO2 NPs (50 ± 3 nm in diameter) on electrical activity and gene expression, providing a detailed analysis of the impact of a nanoparticle on neuronal excitability. We find that 20 µg mL-1 NPs induce depolarization of the membrane potential, with a modulation of the firing of action potentials. Recordings of electrical activity with multielectrode arrays provide further evidence that the NPs evoke a temporary increase in firing frequency, without affecting the functional behavior on a time scale of hours. Finally, NPs incubation up to 24 hours does not induce any change in gene expression.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Nanopartículas , Células Neuroendocrinas/efectos de los fármacos , Neuronas/metabolismo , Dióxido de Silicio/farmacología , Animales , Línea Celular , Expresión Génica/efectos de los fármacos , Hipotálamo/citología , Ratones , Células Neuroendocrinas/fisiología , Neuronas/efectos de los fármacos
5.
Compr Physiol ; 5(1): 217-53, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25589270

RESUMEN

The endocrine hypothalamus constitutes those cells which project to the median eminence and secrete neurohormones into the hypophysial portal blood to act on cells of the anterior pituitary gland. The entire endocrine system is controlled by these peptides. In turn, the hypothalamic neuroendocrine cells are regulated by feedback signals from the endocrine glands and other circulating factors. The neuroendocrine cells are found in specific regions of the hypothalamus and are regulated by afferents from higher brain centers. Integrated function is clearly complex and the networks between and amongst the neuroendocrine cells allows fine control to achieve homeostasis. The entry of hormones and other factors into the brain, either via the cerebrospinal fluid or through fenestrated capillaries (in the basal hypothalamus) is important because it influences the extent to which feedback regulation may be imposed. Recent evidence of the passage of factors from the pars tuberalis and the median eminence casts a new layer in our understanding of neuroendocrine regulation. The function of neuroendocrine cells and the means by which pulsatile secretion is achieved is best understood for the close relationship between gonadotropin releasing hormone and luteinizing hormone, which is reviewed in detail. The secretion of other neurohormones is less rigid, so the relationship between hypothalamic secretion and the relevant pituitary hormones is more complex.


Asunto(s)
Hipotálamo/fisiología , Animales , Retroalimentación Fisiológica/fisiología , Humanos , Hormonas Hipotalámicas/metabolismo , Hormonas Hipotalámicas/fisiología , Hipotálamo/citología , Eminencia Media/fisiología , Células Neuroendocrinas/fisiología , Sistemas Neurosecretores/fisiología , Neurotransmisores/metabolismo , Adenohipófisis/fisiología
6.
J Neurosci ; 33(46): 18331-42, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24227742

RESUMEN

Cannabinoid receptors are functionally operant at both glutamate and GABA synapses on hypothalamic magnocellular neuroendocrine cells; however, retrograde endocannabinoid actions are evoked at only glutamate synapses. We tested whether the functional targeting of evoked retrograde endocannabinoid actions to glutamate, and not GABA, synapses on magnocellular neurons is the result of the spatial restriction of extracellular endocannabinoids by astrocytes. Whole-cell GABA synaptic currents were recorded in magnocellular neurons in rat hypothalamic slices following manipulations to reduce glial buffering of extracellular signals. Depolarization- and glucocorticoid-evoked retrograde endocannabinoid suppression of synaptic GABA release was not detected under normal conditions, but occurred in both oxytocin and vasopressin neurons under conditions of attenuated glial coverage and depressed glial metabolic function, suggesting an emergent endocannabinoid modulation of GABA synapses with the loss of astrocyte function. Tonic endocannabinoid suppression of GABA release was insensitive to glial manipulation. Blocking cannabinoid transport mimicked, and increasing the extracellular viscosity reversed, the effect of suppressed glial buffering on the endocannabinoid modulation of GABA release. Evoked, but not tonic, endocannabinoid modulation of GABA synapses was mediated by 2-arachidonoylglycerol. Therefore, depolarization- and glucocorticoid-evoked 2-arachidonoylglycerol release from magnocellular neurons is spatially restricted to glutamate synapses by astrocytes, but spills over onto GABA synapses under conditions of reduced astrocyte buffering; tonic endocannabinoid modulation of GABA release, in contrast, is likely mediated by anandamide and is insensitive to astrocytic buffering. Astrocytes, therefore, provide dynamic control of stimulus-evoked 2-arachidonoylglycerol, but not tonic anandamide, regulation of GABA synaptic inputs to magnocellular neuroendocrine cells under different physiological conditions.


Asunto(s)
Endocannabinoides/fisiología , Hipotálamo/fisiología , Células Neuroendocrinas/fisiología , Neuroglía/fisiología , Sinapsis/fisiología , Animales , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
7.
Cell Calcium ; 51(3-4): 240-52, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22137240

RESUMEN

Relative to mammals, the neuroendocrine control of pituitary growth hormone (GH) secretion and synthesis in teleost fish involves numerous stimulatory and inhibitory regulators, many of which are delivered to the somatotrophs via direct innervation. Among teleosts, how multifactorial regulation of somatotroph functions are mediated at the level of post-receptor signalling is best characterized in goldfish. Supplemented with recent findings, this review focuses on the known intracellular signal transduction mechanisms mediating the ligand- and function-specific actions in multifactorial control of GH release and synthesis, as well as basal GH secretion, in goldfish somatotrophs. These include membrane voltage-sensitive ion channels, Na(+)/H(+) antiport, Ca(2+) signalling, multiple pharmacologically distinct intracellular Ca(2+) stores, cAMP/PKA, PKC, nitric oxide, cGMP, MEK/ERK and PI3K. Signalling pathways mediating the major neuroendocrine regulators of mammalian somatotrophs, as well as those in other major teleost study model systems are also briefly highlighted. Interestingly, unlike mammals, spontaneous action potential firings are not observed in goldfish somatotrophs in culture. Furthermore, three goldfish brain somatostatin forms directly affect pituitary GH secretion via ligand-specific actions on membrane ion channels and intracellular Ca(2+) levels, as well as exert isoform-specific action on basal and stimulated GH mRNA expression, suggesting the importance of somatostatins other than somatostatin-14.


Asunto(s)
Señalización del Calcio , Carpa Dorada/metabolismo , Hormona del Crecimiento/metabolismo , Células Neuroendocrinas/fisiología , Somatotrofos/fisiología , Animales , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Humanos , Canales Iónicos/metabolismo , Sistema de Señalización de MAP Quinasas , Modelos Animales , Proteína Quinasa C/metabolismo
8.
J Clin Invest ; 120(10): 3668-72, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20940512

RESUMEN

Patients with Kallmann syndrome (KS) have hypogonadotropic hypogonadism caused by a deficiency of gonadotropin-releasing hormone (GnRH) and a defective sense of smell related to olfactory bulb aplasia. Based on the findings in a fetus affected by the X chromosome­linked form of the disease, it has been suggested that hypogonadism in KS results from the failed embryonic migration of neuroendocrine GnRH1 cells from the nasal epithelium to the forebrain. We asked whether this singular observation might extend to other developmental disorders that also include arrhinencephaly. We therefore studied the location of GnRH1 cells in fetuses affected by different arrhinencephalic disorders, specifically X-linked KS, CHARGE syndrome, trisomy 13, and trisomy 18, using immunohistochemistry. Few or no neuroendocrine GnRH1 cells were detected in the preoptic and hypothalamic regions of all arrhinencephalic fetuses, whereas large numbers of these cells were present in control fetuses. In all arrhinencephalic fetuses, many GnRH1 cells were present in the frontonasal region, the first part of their migratory path, as were interrupted olfactory nerve fibers that formed bilateral neuromas. Our findings define a pathological sequence whereby a lack of migration of neuroendocrine GnRH cells stems from the primary embryonic failure of peripheral olfactory structures. This can occur either alone, as in isolated KS, or as part of a pleiotropic disease, such as CHARGE syndrome, trisomy 13, and trisomy 18.


Asunto(s)
Anomalías Múltiples/patología , Cromosomas Humanos Par 13 , Cromosomas Humanos Par 18 , Hormona Liberadora de Gonadotropina/análisis , Síndrome de Kallmann/patología , Células Neuroendocrinas/fisiología , Bulbo Olfatorio/anomalías , Trisomía , Movimiento Celular , Humanos , Hipotálamo/patología , Área Preóptica/patología
9.
Biosystems ; 100(3): 198-207, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20302907

RESUMEN

In this paper a simple one compartment Hodgkin-Huxley type electrophysiological model of GnRH neurons is presented, that is able to reasonably reproduce the most important qualitative features of the firing pattern, such as baseline potential, depolarization amplitudes, sub-baseline hyperpolarization phenomenon and average firing frequency in response to excitatory current. In addition, the same model provides an acceptable numerical fit of voltage clamp (VC) measurement results. The parameters of the model have been estimated using averaged VC traces, and characteristic values of measured current clamp traces originating from GnRH neurons in hypothalamic slices. The resulting parameter values show a good agreement with literature data in most of the cases. Applying parametric changes, which lead to the increase of baseline potential and enhance cell excitability, the model becomes capable of bursting. The effects of various parameters to burst length have been analyzed by simulation.


Asunto(s)
Hormona Liberadora de Gonadotropina/fisiología , Modelos Neurológicos , Neuronas/fisiología , Potenciales de Acción , Animales , Señalización del Calcio , Fenómenos Electrofisiológicos , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipotálamo/citología , Hipotálamo/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Células Neuroendocrinas/fisiología , Técnicas de Placa-Clamp , Potasio/metabolismo , Biología de Sistemas
10.
Endocrinology ; 150(5): 2317-24, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19131574

RESUMEN

Prenatal testosterone (T) exposure defeminizes reproductive neuroendocrine function in female sheep, although the LH surge dysfunctions are initially less severe in gonadally intact females than in females subject to neonatal ovariectomy and estradiol (E) replacement. Because prepubertal ovarian production of E differs quantitatively and qualitatively from chronic E replacement, we tested the hypothesis that postnatal E exacerbates the consequences of prenatal T on the positive, but not the negative, steroid feedback controls of GnRH secretion. Our approach was to characterize prepubertal sensitivity to E negative feedback, the onset and maintenance of progestagenic cycles, and the LH surge response in ovary intact, prenatally untreated (control), and T-treated (T) sheep that were exposed postnatally to only endogenous E, or exposed to excess E by s.c. implant. Sensitivity to E negative feedback was reduced in T females, but excess postnatal E did not further increase LH pulse frequency. Excess E prevented ovarian cycles in several control females, and increased cycle irregularity in T females. However, the LH surge mechanism was functional in all control females (regardless of postnatal E exposure) and in some T females without excess E, but nonfunctional in T females with excess E. These findings suggest that postnatal E does not program increased resistance to E negative feedback, but excess postnatal E does disrupt other mechanisms required for ovarian cyclicity. We conclude that in this precocial species, prenatal steroids are sufficient to program controls of tonic LH secretion, but the LH surge mechanism is susceptible to further programming by postnatal E.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Estradiol/farmacología , Células Neuroendocrinas/efectos de los fármacos , Maduración Sexual/efectos de los fármacos , Testosterona/farmacología , Animales , Animales Recién Nacidos , Evaluación Preclínica de Medicamentos , Desarrollo Embrionario/fisiología , Ciclo Estral/efectos de los fármacos , Ciclo Estral/fisiología , Retroalimentación Fisiológica/efectos de los fármacos , Retroalimentación Fisiológica/fisiología , Femenino , Hormona Luteinizante/metabolismo , Células Neuroendocrinas/fisiología , Ovariectomía/veterinaria , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Flujo Pulsátil/efectos de los fármacos , Maduración Sexual/fisiología , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA