Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Neuroinform ; 17: 1005936, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36970656

RESUMEN

The rapid effects of estradiol on membrane receptors are in the focus of the estradiol research field, however, the molecular mechanisms of these non-classical estradiol actions are poorly understood. Since the lateral diffusion of membrane receptors is an important indicator of their function, a deeper understanding of the underlying mechanisms of non-classical estradiol actions can be achieved by investigating receptor dynamics. Diffusion coefficient is a crucial and widely used parameter to characterize the movement of receptors in the cell membrane. The aim of this study was to investigate the differences between maximum likelihood-based estimation (MLE) and mean square displacement (MSD) based calculation of diffusion coefficients. In this work we applied both MSD and MLE to calculate diffusion coefficients. Single particle trajectories were extracted from simulation as well as from α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor tracking in live estradiol-treated differentiated PC12 (dPC12) cells. The comparison of the obtained diffusion coefficients revealed the superiority of MLE over the generally used MSD analysis. Our results suggest the use of the MLE of diffusion coefficients because as it has a better performance, especially for large localization errors or slow receptor movements.

2.
Front Endocrinol (Lausanne) ; 13: 993228, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36387875

RESUMEN

The hypothalamic gonadotropin-releasing hormone (GnRH)-kisspeptin neuronal network regulates fertility in all mammals. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide isolated from the hypothalamus that is involved in the regulation of several releasing hormones and trop hormones. It is well-known that PACAP influences fertility at central and peripheral levels. However, the effects of PACAP on GnRH and kisspeptin neurons are not well understood. The present study investigated the integrity of the estrous cycle in PACAP-knockout (KO) mice. The number and immunoreactivity of GnRH (GnRH-ir) neurons in wild-type (WT) and PACAP KO female mice were determined using immunohistochemistry. In addition, the number of kisspeptin neurons was measured by counting kisspeptin mRNA-positive cells in the rostral periventricular region of the third ventricle (RP3V) and arcuate nucleus (ARC) using the RNAscope technique. Finally, the mRNA and protein expression of estrogen receptor alpha (ERα) was also examined. Our data showed that the number of complete cycles decreased, and the length of each cycle was longer in PACAP KO mice. Furthermore, the PACAP KO mice experienced longer periods of diestrus and spent significantly less time in estrus. There was no difference in GnRH-ir or number of GnRH neurons. In contrast, the number of kisspeptin neurons was decreased in the ARC, but not in the R3PV, in PACAP KO mice compared to WT littermates. Furthermore, ERα mRNA and protein expression was decreased in the ARC, whereas in the R3PV region, ERα mRNA levels were elevated. Our results demonstrate that embryonic deletion of PACAP significantly changes the structure and presumably the function of the GnRH-kisspeptin neuronal network, influencing fertility.


Asunto(s)
Hormona Liberadora de Gonadotropina , Kisspeptinas , Animales , Femenino , Ratones , Receptor alfa de Estrógeno/metabolismo , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Ratones Noqueados , Neuronas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , ARN Mensajero/metabolismo
3.
Front Endocrinol (Lausanne) ; 13: 993552, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36686456

RESUMEN

The striatum is an essential component of the basal ganglia that is involved in motor control, action selection and motor learning. The pathophysiological changes of the striatum are present in several neurological and psychiatric disorder including Parkinson's and Huntington's diseases. The striatal cholinergic neurons are the main regulators of striatal microcircuitry. It has been demonstrated that estrogen exerts various effects on neuronal functions in dopaminergic and medium spiny neurons (MSN), however little is known about how the activity of cholinergic interneurons are influenced by estrogens. In this study we examined the acute effect of 17ß-estradiol on the function of striatal cholinergic neurons in adult mice in vitro. We also tested the effect of estrus cycle and sex on the spontaneous activity of cholinergic interneurons in the striatum. Our RNAscope experiments showed that ERα, ERß, and GPER1 receptor mRNAs are expressed in some striatal cholinergic neurons at a very low level. In cell-attached patch clamp experiments, we found that a high dose of 17ß-estradiol (100 nM) affected the spontaneous firing rate of these neurons only in old males. Our findings did not demonstrate any acute effect of a low concentration of 17ß-estradiol (100 pM) or show any association of estrus cycle or sex with the activity of striatal cholinergic neurons. Although estrogen did not induce changes in the intrinsic properties of neurons, indirect effects via modulation of the synaptic inputs of striatal cholinergic interneurons cannot be excluded.


Asunto(s)
Colinérgicos , Interneuronas , Masculino , Femenino , Ratones , Animales , Interneuronas/fisiología , Colinérgicos/farmacología , Neuronas Colinérgicas/fisiología , Estradiol/farmacología , Estrógenos
4.
Int J Mol Sci ; 22(24)2021 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-34948057

RESUMEN

Neurotrophin receptors such as the tropomyosin receptor kinase A receptor (TrkA) and the low-affinity binding p75 neurotrophin receptor p75NTR play a critical role in neuronal survival and their functions are altered in Alzheimer's disease (AD). Changes in the dynamics of receptors on the plasma membrane are essential to receptor function. However, whether receptor dynamics are affected in different pathophysiological conditions is unexplored. Using live-cell single-molecule imaging, we examined the surface trafficking of TrkA and p75NTR molecules on live neurons that were derived from human-induced pluripotent stem cells (hiPSCs) of presenilin 1 (PSEN1) mutant familial AD (fAD) patients and non-demented control subjects. Our results show that the surface movement of TrkA and p75NTR and the activation of TrkA- and p75NTR-related phosphoinositide-3-kinase (PI3K)/serine/threonine-protein kinase (AKT) signaling pathways are altered in neurons that are derived from patients suffering from fAD compared to controls. These results provide evidence for altered surface movement of receptors in AD and highlight the importance of investigating receptor dynamics in disease conditions. Uncovering these mechanisms might enable novel therapies for AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Presenilina-1/genética , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Adulto , Enfermedad de Alzheimer/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Neuronas/metabolismo , Células PC12 , Ratas , Transducción de Señal , Imagen Individual de Molécula
5.
Front Cell Dev Biol ; 9: 708715, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34631701

RESUMEN

Gonadal steroid 17ß-estradiol (E2) exerts rapid, non-genomic effects on neurons and strictly regulates learning and memory through altering glutamatergic neurotransmission and synaptic plasticity. However, its non-genomic effects on AMPARs are not well understood. Here, we analyzed the rapid effect of E2 on AMPARs using single-molecule tracking and super-resolution imaging techniques. We found that E2 rapidly decreased the surface movement of AMPAR via membrane G protein-coupled estrogen receptor 1 (GPER1) in neurites in a dose-dependent manner. The cortical actin network played a pivotal role in the GPER1 mediated effects of E2 on the surface mobility of AMPAR. E2 also decreased the surface movement of AMPAR both in synaptic and extrasynaptic regions on neurites and increased the synaptic dwell time of AMPARs. Our results provide evidence for understanding E2 action on neuronal plasticity and glutamatergic neurotransmission at the molecular level.

6.
Front Bioeng Biotechnol ; 9: 692075, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34660546

RESUMEN

Stimulated emission depletion (STED) nanoscopy has overcome a serious diffraction barrier on the optical resolution and facilitated new discoveries on detailed nanostructures in cell biology. Traditional fluorescence probes employed in the super-resolution imaging approach include organic dyes and fluorescent proteins. However, some limitations of these probes, such as photobleaching, short emission wavelengths, and high saturation intensity, still hamper the promotion of optical resolution and bio-applications. Recently, lanthanide luminescent probes with unique optical properties of non-photobleaching and sharp emissions have been applied in super-resolution imaging. In this mini-review, we will introduce several different mechanisms for lanthanide ions to achieve super-resolution imaging based on an STED-like setup. Then, several lanthanide ions used in super-resolution imaging will be described in detail and discussed. Last but not least, we will emphasize the future challenges and outlooks in hope of advancing the next-generation lanthanide fluorescent probes for super-resolution optical imaging.

7.
Angew Chem Int Ed Engl ; 60(44): 23545-23551, 2021 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-34487416

RESUMEN

Fluorescence lifetime imaging provides more possibility of in vivo multiplexing in second near infrared (NIR-II) window. However, it still faces the obstacle that fluorescent probes with differentiable lifetime often exhibit quite different fluorescence intensity, especially the short lifetime usually accompanies with a weak fluorescence intensity, resulting in the difficulty for simultaneously decoding multiplexed lifetime information due to the interference of background noise. To facilitate high-fidelity lifetime multiplexed imaging, we developed a series of Er3+ doped double interface fluorescent nanoprobes (Er-DINPs): α-NaYF4 @NaErF4 : Ce@NaYbF4 @NaErF4 : Ce@NaYF4 with strong fluorescence intensity and easily distinguishable fluorescence lifetime. Both in vitro and in vivo experimental results confirmed the advantage of these probes with comparable fluorescence intensity for high-fidelity multiplexed lifetime bioimaging.


Asunto(s)
Elementos de la Serie de los Lantanoides/química , Nanopartículas del Metal/química , Neoplasias/diagnóstico por imagen , Imagen Óptica , Animales , Rayos Infrarrojos , Hígado/diagnóstico por imagen , Ratones , Tamaño de la Partícula
8.
Int J Mol Sci ; 22(1)2020 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-33374952

RESUMEN

Patients surviving traumatic brain injury (TBI) face numerous neurological and neuropsychological problems significantly affecting their quality of life. Extensive studies over the past decades have investigated pharmacological treatment options in different animal models, targeting various pathological consequences of TBI. Sex and gender are known to influence the outcome of TBI in animal models and in patients, respectively. Apart from its well-known effects on reproduction, 17ß-estradiol (E2) has a neuroprotective role in brain injury. Hence, in this review, we focus on the effect of E2 in TBI in humans and animals. First, we discuss the clinical classification and pathomechanism of TBI, the research in animal models, and the neuroprotective role of E2. Based on the results of animal studies and clinical trials, we discuss possible E2 targets from early to late events in the pathomechanism of TBI, including neuroinflammation and possible disturbances of the endocrine system. Finally, the potential relevance of selective estrogenic compounds in the treatment of TBI will be discussed.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Encefálicas/tratamiento farmacológico , Estradiol/uso terapéutico , Neuroprotección/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Estradiol/farmacología , Estrógenos/farmacología , Estrógenos/uso terapéutico , Humanos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
9.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096776

RESUMEN

Somatostatin is an important mood and pain-regulating neuropeptide, which exerts analgesic, anti-inflammatory, and antidepressant effects via its Gi protein-coupled receptor subtype 4 (SST4) without endocrine actions. SST4 is suggested to be a unique novel drug target for chronic neuropathic pain, and depression, as a common comorbidity. However, its neuronal expression and cellular mechanism are poorly understood. Therefore, our goals were (i) to elucidate the expression pattern of Sstr4/SSTR4 mRNA, (ii) to characterize neurochemically, and (iii) electrophysiologically the Sstr4/SSTR4-expressing neuronal populations in the mouse and human brains. Here, we describe SST4 expression pattern in the nuclei of the mouse nociceptive and anti-nociceptive pathways as well as in human brain regions, and provide neurochemical and electrophysiological characterization of the SST4-expressing neurons. Intense or moderate SST4 expression was demonstrated predominantly in glutamatergic neurons in the major components of the pain matrix mostly also involved in mood regulation. The SST4 agonist J-2156 significantly decreased the firing rate of layer V pyramidal neurons by augmenting the depolarization-activated, non-inactivating K+ current (M-current) leading to remarkable inhibition. These are the first translational results explaining the mechanisms of action of SST4 agonists as novel analgesic and antidepressant candidates.


Asunto(s)
Analgésicos/farmacología , Encéfalo/metabolismo , Neuronas/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Afecto/fisiología , Animales , Encéfalo/citología , Butanos/farmacología , Células CHO , Cricetulus , Electrofisiología/métodos , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Terapia Molecular Dirigida , Naftalenos/farmacología , Neuronas/efectos de los fármacos , Receptores de Somatostatina/agonistas , Sulfonas/farmacología , Proteína 1 de Transporte Vesicular de Glutamato/genética
10.
Int J Mol Sci ; 21(17)2020 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-32825350

RESUMEN

Melatonin, a pineal gland secretion, is an amphiphilic neurohormone involved in the biological and physiologic regulation of bodily functions. Numerous studies have shown the effects of melatonin on the release of gonadotropins and their actions at one or several levels of the hypothalamic-pituitary-gonadal axis. However, direct melatonin action on gonadotropin-releasing hormone (GnRH) neurons and its mechanism of action remain unclear. Here, plasma melatonin levels were measured and the effect of melatonin on GnRH neurons was assessed using brain slice patch clamp techniques. The plasma melatonin levels in prepubertal mice were higher than those in the adults. Melatonin itself did not change the firing activity of GnRH neurons. Interestingly, the kainate receptor-mediated responses but not the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)- and N-methyl-D-aspartic acid (NMDA)-induced responses were suppressed by melatonin in both the voltage clamp and current clamp modes. The inhibitory effects of the kainate-induced response by melatonin tended to increase with higher melatonin concentrations and persisted in the presence of tetrodotoxin, a voltage-sensitive Na+ channel blocker, or luzindole, a non-selective melatonin receptor antagonist. However, the response was completely abolished by pretreatment with pertussis toxin. These results suggest that melatonin can regulate GnRH neuronal activities in prepubertal mice by partially suppressing the excitatory signaling mediated by kainate receptors through pertussis toxin-sensitive G-protein-coupled receptors.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Melatonina/farmacología , Neuronas/fisiología , Receptores de Ácido Kaínico/metabolismo , Animales , Encéfalo/efectos de los fármacos , Agonistas de Aminoácidos Excitadores , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ácido Kaínico/farmacología , Masculino , Melatonina/sangre , Ratones Endogámicos C57BL , Ratones Transgénicos , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Toxina del Pertussis/farmacología , Pubertad , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
11.
Int J Mol Sci ; 21(9)2020 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-32365920

RESUMEN

Gonadal hormone 17ß-estradiol (E2) and its receptors are key regulators of gene transcription by binding to estrogen responsive elements in the genome. Besides the classical genomic action, E2 regulates gene transcription via the modification of epigenetic marks on DNA and histone proteins. Depending on the reaction partner, liganded estrogen receptor (ER) promotes DNA methylation at the promoter or enhancer regions. In addition, ERs are important regulators of passive and active DNA demethylation. Furthermore, ERs cooperating with different histone modifying enzymes and chromatin remodeling complexes alter gene transcription. In this review, we survey the basic mechanisms and interactions between estrogen receptors and DNA methylation, demethylation and histone modification processes as well as chromatin remodeling complexes. The particular relevance of these mechanisms to physiological processes in memory formation, embryonic development, spermatogenesis and aging as well as in pathophysiological changes in carcinogenesis is also discussed.


Asunto(s)
Estradiol/farmacología , Animales , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Histonas/metabolismo , Humanos , Regiones Promotoras Genéticas/genética
12.
Int J Mol Sci ; 21(2)2020 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-31947687

RESUMEN

: Inflammation has a well-known suppressive effect on fertility. The function of gonadotropin-releasing hormone (GnRH) neurons, the central regulator of fertility is substantially altered during inflammation in females. In our review we discuss the latest results on how the function of GnRH neurons is modified by inflammation in females. We first address the various effects of inflammation on GnRH neurons and their functional consequences. Second, we survey the possible mechanisms underlying the inflammation-induced actions on GnRH neurons. The role of several factors will be discerned in transmitting inflammatory signals to the GnRH neurons: cytokines, kisspeptin, RFamide-related peptides, estradiol and the anti-inflammatory cholinergic pathway. Since aging and obesity are both characterized by reproductive decline our review also focuses on the mechanisms and pathophysiological consequences of the impact of inflammation on GnRH neurons in aging and obesity.


Asunto(s)
Citocinas/metabolismo , Hormona Liberadora de Gonadotropina/biosíntesis , Inflamación/metabolismo , Neuronas/metabolismo , Transducción de Señal , Envejecimiento/genética , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Astrocitos/metabolismo , Biomarcadores , Barrera Hematoencefálica/metabolismo , Estradiol/metabolismo , Retroalimentación Fisiológica , Femenino , Fertilidad/genética , Hormona Liberadora de Gonadotropina/genética , Humanos , Inflamación/etiología , Kisspeptinas/genética , Kisspeptinas/metabolismo , Lipopolisacáridos/inmunología , Microglía/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Reproducción/genética , Reproducción/inmunología
13.
J Neuroendocrinol ; 32(1): e12830, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31943420

RESUMEN

Rapid nonclassical effects of 17ß-oestradiol (E2 ) on intracellular signalling have been identified in the basal forebrain, although the extent to which these actions may be different in males and females is unknown. Previous work has shown that E2 rapidly phosphorylates cAMP responsive element binding protein (CREB) via ΕRα in female cholinergic neurones. Using this indicator, the present study examined whether nonclassical actions of E2 occur in a sexually dimorphic manner within basal forebrain cholinergic neurones in mice. In addition, we investigated the expression and intracellular distribution of oestrogen receptor (ΕR)α in cholinergic neurones in female and male mice. Animals were gonadectomised and treated 2 weeks later with E2 . The number of CREB-expressing cholinergic neurones was not altered in any of the brain regions after E2 treatment in both males and females. However, E2 treatment rapidly (< 15 minutes) increased (P < 0.05) the number of cholinergic neurones expressing phosphorylated CREB (pCREB) in the substantia innominata and medial septum but not in the striatum in female mice. By contrast, E2 did not change pCREB expression in cholinergic neurones in male mice at any time point (15 minutes, 1 hour, 4 hours), irrespective of the neuroanatomical location. We also observed that, in females, more cholinergic neurones expressed nuclear ΕRα in all regions, whereas males showed more cholinergic neurones with cytoplasmic or both nuclear and cytoplasmic expression of ΕRα. Taken together, these results demonstrate a marked sex difference in the E2 -induced nonclassical effect and intracellular distribution of ΕRα in basal forebrain cholinergic neurones in vivo.


Asunto(s)
Prosencéfalo Basal/efectos de los fármacos , Neuronas Colinérgicas/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Prosencéfalo Basal/metabolismo , Neuronas Colinérgicas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Masculino , Ratones , Ovariectomía , Fosforilación/efectos de los fármacos , Factores Sexuales
14.
eNeuro ; 5(5)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30406179

RESUMEN

Immune challenge alters neural functioning via cytokine production. Inflammation has profound impact on the central regulation of fertility, but the mechanisms involved are not clearly defined. The anti-inflammatory cytokine interleukin (IL)-10 is responsible for balancing the immune response in the brain. To examine whether IL-10 has an effect on the function of the gonadotropin-releasing hormone (GnRH) neurons, we first examined the effect of immune responses with distinct cytokine profiles, such as the T cell-dependent (TD) and T cell-independent (TI) B-cell response. We investigated the effect of the TD and TI immune responses on ERK1/2 phosphorylation in GnRH neurons by administering fluorescein isothiocyanate/keyhole limpet hemocyanin (KLH-FITC) or dextran-FITC to female mice. Although dextran-FITC had no effect, KLH-FITC induced ERK1/2 phosphorylation in GnRH neurons after 6 d. KLH-FITC treatment increased the levels of IL-10 in the hypothalamus (HYP), but this treatment did not cause lymphocyte infiltration or an increase in the levels of proinflammatory cytokines. In IL-10 knock-out (KO) mice, KLH-FITC-induced ERK1/2 phosphorylation in the GnRH neurons was absent. We also showed that in IL-10 KO mice, the estrous cycle was disrupted. Perforated patch-clamp recordings from GnRH-GFP neurons, IL-10 immunohistochemistry, and in vitro experiments on acute brain slices revealed that IL-10 can directly alter GnRH neuron firing and induce ERK1/2 phosphorylation. These observations demonstrate that IL-10 plays a role in influencing signaling of GnRH neurons in the TD immune response. These results also provide the first evidence that IL-10 can directly alter the function of GnRH neurons and may help the maintenance of the integrity of the estrous cycle.


Asunto(s)
Hormona Liberadora de Gonadotropina/inmunología , Hipotálamo/efectos de los fármacos , Interleucina-10/inmunología , Neuronas/inmunología , Animales , Citocinas/inmunología , Estradiol/inmunología , Estradiol/farmacología , Ciclo Estral/inmunología , Femenino , Hipotálamo/inmunología , Interleucina-10/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
15.
Horm Behav ; 104: 183-191, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29775570

RESUMEN

Contribution to Special Issue on Fast effects of steroids. Although rapid effects of steroid hormones on membrane receptors and intracellular signaling molecules have been extensively studied in neurons, we are only beginning to understand the molecular mechanisms behind these non-classical steroid actions. Single molecule tracking (SMT) studies on live cells demonstrated that surface trafficking of membrane receptors determines their ligand binding properties and downstream signaling events. Recent findings suggest that one of the underlying mechanisms of non-classical steroid actions is the alteration of receptor movements on the membrane surface. In order to highlight this novel aspect of steroid effects, we first address the types of receptor movements in the plasma membrane and the role of cortical actin dynamics in receptor movement. We then discuss how single molecules and the surface movements of receptors can be detected in live cells. Next, we review the fundamental processes, which determine the effect of steroids on the plasma membrane: steroid movement through the lipid bilayer and the role of steroid membrane receptors. Using glutamate and neurotrophin receptors (NTRs) as examples, we demonstrate the features of receptor dynamics in the membrane. In addition, we survey the available data of rapid steroid actions on membrane receptor trafficking: we discuss how glucocorticoids act on the surface diffusion of glutamate receptor molecules and how estradiol acts on NTRs and gamma-aminobutyric acid type A receptors (GABAARs) and their related signaling events as well as on cortical actin. Finally, we address the physiological relevance of rapid steroid action on membrane receptors dynamics.


Asunto(s)
Hormonas Esteroides Gonadales/farmacología , Neuronas/efectos de los fármacos , Receptores de Esteroides/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Estradiol/farmacología , Glucocorticoides/farmacología , Humanos , Neuronas/fisiología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
16.
Sci Rep ; 7(1): 14847, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29093525

RESUMEN

Non-classical signaling in the intracellular second messenger system plays a pivotal role in the cytoprotective effect of estradiol. Estrogen receptor is a common target of sex steroids and important in mediating estradiol-induced neuroprotection. Whereas the mechanism of genomic effects of sex steroids is fairly understood, their non-classical effects have not been elucidated completely. We use real time molecular dynamics calculations to uncover the interaction network of estradiol and activator estren. Besides steroid interactions, we also investigate the co-activation of the receptor. We show how steroid binding to the alternative binding site of the non-classical action is facilitated by the presence of a steroid in the classical binding site and the absence of the co-activator peptide. Uncovering such dynamic mechanisms behind steroid action will help the structure-based design of new drugs with non-classical responses and cytoprotective potential.


Asunto(s)
Estradiol/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Simulación de Dinámica Molecular , Animales , Sitios de Unión , Estrenos/metabolismo , Humanos , Fármacos Neuroprotectores , Receptores de Estrógenos/metabolismo , Sistemas de Mensajero Secundario
17.
Endocrinology ; 158(10): 3249-3258, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28977586

RESUMEN

Sex differences exist in chronic pain pathologies, and gonadal estradiol (E2) alters the pain sensation. The nocisensor transient receptor potential vanilloid 1 (TRPV1) receptor plays a critical role in triggering pain. Here we examined the impact of E2 on the function of TRPV1 receptor in mice sensory neurons in vitro and in vivo. Both mechano- and thermonociceptive thresholds of the plantar surface of the paw of female mice were significantly lower in proestrus compared with the estrus phase. These thresholds were higher in ovariectomized (OVX) mice and significantly lower in sham-operated mice in proestrus compared with the sham-operated mice in estrus phase. This difference was absent in TRPV1 receptor-deficient mice. Furthermore, E2 potentiated the TRPV1 receptor activation-induced mechanical hyperalgesia in OVX mice. Long pretreatment (14 hours) with E2 induced a significant increase in TRPV1 receptor messenger RNA expression and abolished the capsaicin-induced TRPV1 receptor desensitization in primary sensory neurons. The short E2 incubation (10 minutes) also prevented the desensitization, which reverted after coadministration of E2 and the tropomyosin-related kinase A (TrkA) receptor inhibitor. Our study provides in vivo and in vitro evidence for E2-induced TRPV1 receptor upregulation and sensitization mediated by TrkAR via E2-induced genomic and nongenomic mechanisms. The sensitization and upregulation of TRPV1 receptor by E2 in sensory neurons may explain the greater pain sensitivity in female mice.


Asunto(s)
Estradiol/farmacología , Dolor/fisiopatología , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/fisiología , Animales , Capsaicina/farmacología , Células Cultivadas , Tolerancia a Medicamentos , Estro/fisiología , Femenino , Expresión Génica/efectos de los fármacos , Calor , Masculino , Mecanorreceptores/efectos de los fármacos , Mecanorreceptores/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nociceptores/efectos de los fármacos , Nociceptores/fisiología , Ovariectomía , Proestro/fisiología , ARN Mensajero/análisis , Células Receptoras Sensoriales/química , Células Receptoras Sensoriales/fisiología , Caracteres Sexuales , Canales Catiónicos TRPV/genética , Regulación hacia Arriba/efectos de los fármacos
18.
Sci Rep ; 6: 21101, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26879842

RESUMEN

In Alzheimer's disease (AD), there is a loss in cholinergic innervation targets of basal forebrain which has been implicated in substantial cognitive decline. Amyloid beta peptide (Aß(1-42)) accumulates in AD that is highly toxic for basal forebrain cholinergic (BFC) neurons. Although the gonadal steroid estradiol is neuroprotective, the administration is associated with risk of off-target effects. Previous findings suggested that non-classical estradiol action on intracellular signaling pathways has ameliorative potential without estrogenic side effects. After Aß(1-42) injection into mouse basal forebrain, a single dose of 4-estren-3α, 17ß-diol (estren), the non-classical estradiol pathway activator, restored loss of cholinergic cortical projections and also attenuated the Aß(1-42)-induced learning deficits. Estren rapidly and directly phosphorylates c-AMP-response-element-binding-protein and extracellular-signal-regulated-kinase-1/2 in BFC neurons and restores the cholinergic fibers via estrogen receptor-α. These findings indicated that selective activation of non-classical intracellular estrogen signaling has a potential to treat the damage of cholinergic neurons in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Prosencéfalo Basal/metabolismo , Fibras Colinérgicas/metabolismo , Estrógenos/metabolismo , Fragmentos de Péptidos/metabolismo , Transducción de Señal , Péptidos beta-Amiloides/farmacología , Animales , Prosencéfalo Basal/efectos de los fármacos , Prosencéfalo Basal/patología , Recuento de Células , Fibras Colinérgicas/efectos de los fármacos , Fibras Colinérgicas/patología , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/patología , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Femenino , Aprendizaje/efectos de los fármacos , Ratones , Ratones Noqueados , Fragmentos de Péptidos/farmacología , Fosforilación , Transducción de Señal/efectos de los fármacos , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/metabolismo , Corteza Somatosensorial/patología
19.
Endocrinology ; 156(2): 613-26, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25415243

RESUMEN

The neuroprotective effect of estradiol (E2) on basal forebrain cholinergic neurons (BFCNs) has been suggested to occur as a result of E2 modulation of the neurotrophin system on these neurons. The present study provides a comprehensive examination of the relationship between E2 and neurotrophin signaling on BFCNs by investigating the effect of E2 deficiency on the expression levels of neurotrophin receptors (NRs), TrkA, TrkB, and p75 on BFCNs. The number of TrkA receptor-expressing choline acetyltransferase-positive neurons was significantly reduced in the medial septum (MS) in the absence of E2. A significant reduction in TrkB-expressing choline acetyltransferase-positive cells was also observed in ovariectomized mice in the MS and nucleus basalis magnocellularis (NBM). p75 receptor expression was reduced in the NBM and striatum but not in the MS. We also showed that estrogen receptor (ER)-α was expressed by a small percentage of TrkA- and TrkB-positive neurons in the MS (12%) and NBM (19%) and by a high percentage of TrkB-positive neurons in the striatum (69%). Similarly, ERα was expressed at low levels by p75 neurons in the MS (6%) and NBM (9%) but was not expressed on striatal neurons. Finally, ERα knockout using neuron-specific estrogen receptor-α knockout transgenic mice abolished all E2-mediated changes in the NR expression on BFCNs. These results indicate that E2 differentially regulates NR expression on BFCNs, with effects depending on the NR type and neuroanatomical location, and also provide some evidence that alterations in the NR expression are, at least in part, mediated via ERα.


Asunto(s)
Prosencéfalo Basal/metabolismo , Neuronas Colinérgicas/metabolismo , Estradiol/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Femenino , Ratones Noqueados , Ovariectomía
20.
Front Neuroendocrinol ; 35(1): 31-41, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23978477

RESUMEN

Gonadotropin-releasing hormone (GnRH) is produced by a heterogenous neuronal population in the hypothalamus to control pituitary gonadotropin production and reproductive function in all mammalian species. Estradiol is a critical component for the communication between the gonads and the central nervous system. Resolving the mechanisms by which estradiol modulates GnRH neurons is critical for the understanding of how fertility is regulated. Extensive studies during the past decades have provided compelling evidence that estradiol has the potential to alter the intracellular signal transduction mechanisms. The common target of many signaling pathways is the phosphorylation of a key transcription factor, the cAMP response element binding protein (CREB). This review first addresses the aspects of estradiol action on CREB phosphorylation (pCREB) in GnRH neurons. Secondly, this review considers the receptors and signaling network that regulates estradiol's action on pCREB within GnRH neurons and finally it summarizes the physiological significance of CREB to estrogen feedback.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/metabolismo , Animales , Estrógenos/metabolismo , Humanos , Fosforilación/fisiología , Fosfotransferasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...