Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
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
2.
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
3.
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
4.
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.

5.
Front Endocrinol (Lausanne) ; 13: 974788, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36387878

RESUMEN

Observations of women and clinicians indicated that the prevalence of menstrual cycle problems has escalated during the COVID-19 pandemic. However, it was not clear whether the observed menstrual cycle changes were related to vaccination, the disease itself or the COVID-19 pandemic-induced psychological alterations. To systematically analyze this question, we conducted a human online survey in women aged between 18 and 65 in Hungary. The menstrual cycle of 1563 individuals were analyzed in our study in relation to the COVID-19 vaccination, the COVID-19 infection, the pandemic itself and the mental health. We found no association between the COVID-19 vaccination, the vaccine types or the COVID-19 infection and the menstrual cycle changes. We also evaluated the menstrual cycle alterations focusing on three parameters of the menstrual cycle including the cycle length, the menses length and the cycle regularity in three pandemic phases: the pre-peak, the peak and the post-peak period in Hungary. Our finding was that the length of the menstrual cycle did not change in any of the periods. However, the menses length increased, while the regularity of the menstrual cycle decreased significantly during the peak of the COVID-19 pandemic when comparing to the pre- and post-peak periods. In addition, we exhibited that the length and the regularity of the menstrual cycle both correlated with the severity of depression during the post-peak period, therefore we concluded that the reported menstrual cycle abnormalities during the peak of COVID-19 in Hungary might be the result of elevated depressive symptoms.


Asunto(s)
COVID-19 , Humanos , Femenino , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , Anciano , Estudios Retrospectivos , COVID-19/epidemiología , COVID-19/prevención & control , Pandemias , Hungría/epidemiología , Vacunas contra la COVID-19 , Ciclo Menstrual/psicología , Vacunación
6.
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
7.
J Neurosci ; 30(42): 13992-4007, 2010 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-20962221

RESUMEN

Endocannabinoids, particularly 2-arachidonoyl glycerol (2-AG), impact the directional turning and motility of a developing axon by activating CB(1) cannabinoid receptors (CB(1)Rs) in its growth cone. Recent findings posit that sn-1-diacylglycerol lipases (DAGLα/ß) synthesize 2-AG in the motile axon segment of developing pyramidal cells. Coincident axonal targeting of CB(1)Rs and DAGLs prompts the hypothesis that autocrine 2-AG signaling facilitates axonal outgrowth. However, DAGLs alone are insufficient to account for the spatial specificity and dynamics of 2-AG signaling. Therefore, we hypothesized that local 2-AG degradation by monoacylglycerol lipase (MGL) must play a role. We determined how subcellular recruitment of MGL is temporally and spatially restricted to establish the signaling competence of 2-AG during axonal growth. MGL is expressed in central and peripheral axons of the fetal nervous system by embryonic day 12.5. MGL coexists with DAGLα and CB(1)Rs in corticofugal axons of pyramidal cells. Here, MGL and DAGLα undergo differential axonal targeting with MGL being excluded from the motile neurite tip. Thus, spatially confined MGL activity generates a 2-AG-sensing microdomain and configures 2-AG signaling to promote axonal growth. Once synaptogenesis commences, MGL disperses in stationary growth cones. The axonal polarity of MGL is maintained by differential proteasomal degradation because inhibiting the ubiquitin proteasome system also induces axonal MGL redistribution. Because MGL inactivation drives a CB(1)R-dependent axonal growth response, we conclude that 2-AG may act as a focal protrusive signal for developing neurons and whose regulated metabolism is critical for attaining correct axonal complexity.


Asunto(s)
Ácidos Araquidónicos/fisiología , Axones/enzimología , Moduladores de Receptores de Cannabinoides/fisiología , Glicéridos/fisiología , Monoacilglicerol Lipasas/metabolismo , Transducción de Señal/fisiología , Fracciones Subcelulares/enzimología , Animales , Axones/ultraestructura , Western Blotting , Células Cultivadas , Cromatografía Líquida de Alta Presión , Endocannabinoides , Glutamato Descarboxilasa/genética , Inmunohistoquímica , Lipoproteína Lipasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Monoacilglicerol Lipasas/genética , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Neuronas/enzimología , Neuronas/ultraestructura , Células Piramidales/enzimología , Células Piramidales/metabolismo , Receptor Cannabinoide CB1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fracciones Subcelulares/ultraestructura , Espectrometría de Masas en Tándem
8.
Cell Mol Life Sci ; 67(10): 1661-74, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20140748

RESUMEN

Estrogen plays a critical regulatory role in the development and maintenance of immunity. Its role in the regulation of antibody synthesis in vivo is still not completely clear. Here, we have compared the effect of estrogen on T cell-dependent (TD) and T cell-independent type 2 (TI-2) antibody responses. The results provide the first evidence that estrogen enhances the TD but not the TI-2 response. Ovariectomy significantly decreased, while estrogen re-administration increased the number of hapten-specific IgM- and IgG-producing cells in response to TD antigen. In vitro experiments also show that estrogen may have a direct impact on B and T cells by inducing rapid signaling events, such as Erk and AKT phosphorylation, cell-specific Ca(2+) signal, and NFkappaB activation. These non-transcriptional effects are mediated by classical estrogen receptors and partly by an as yet unidentified plasma membrane estrogen receptor. Such receptor- mediated rapid signals may modulate the in vivo T cell-dependent immune response.


Asunto(s)
Estradiol/farmacología , Inmunidad/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Animales , Formación de Anticuerpos/efectos de los fármacos , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/enzimología , Linfocitos B/inmunología , Señalización del Calcio/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Ovariectomía , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Estrógenos/metabolismo , Linfocitos T/citología , Linfocitos T/enzimología , Transcripción Genética/efectos de los fármacos
9.
Proc Natl Acad Sci U S A ; 105(25): 8760-5, 2008 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-18562289

RESUMEN

Endocannabinoids (eCBs) have recently been identified as axon guidance cues shaping the connectivity of local GABAergic interneurons in the developing cerebrum. However, eCB functions during pyramidal cell specification and establishment of long-range axonal connections are unknown. Here, we show that eCB signaling is operational in subcortical proliferative zones from embryonic day 12 in the mouse telencephalon and controls the proliferation of pyramidal cell progenitors and radial migration of immature pyramidal cells. When layer patterning is accomplished, developing pyramidal cells rely on eCB signaling to initiate the elongation and fasciculation of their long-range axons. Accordingly, CB(1) cannabinoid receptor (CB(1)R) null and pyramidal cell-specific conditional mutant (CB(1)R(f/f,NEX-Cre)) mice develop deficits in neuronal progenitor proliferation and axon fasciculation. Likewise, axonal pathfinding becomes impaired after in utero pharmacological blockade of CB(1)Rs. Overall, eCBs are fundamental developmental cues controlling pyramidal cell development during corticogenesis.


Asunto(s)
Axones/metabolismo , Moduladores de Receptores de Cannabinoides/metabolismo , Endocannabinoides , Células Piramidales/metabolismo , Transducción de Señal , Animales , Antagonistas de Receptores de Cannabinoides , Diferenciación Celular , Femenino , Humanos , Ratones , Ratones Transgénicos , Embarazo , Células Piramidales/citología , Receptores de Cannabinoides/genética , Receptores de Cannabinoides/metabolismo
10.
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.

11.
Mol Cell Endocrinol ; 286(1-2 Suppl 1): S84-90, 2008 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-18394789

RESUMEN

Endocannabinoids (eCBs) regulate a broad range of physiological functions in the postnatal brain and are implicated in the neuropathogenesis of psychiatric and metabolic diseases. Accumulating evidence indicates that eCB signaling also serves key functions during neurodevelopment; and is inherently involved in the control of neurogenesis, neural progenitor proliferation, lineage segregation, and the migration and phenotypic specification of immature neurons. Recent advances in developmental biology define fundamental eCB-driven cellular mechanisms that also contribute to our understanding of the molecular substrates of prenatal drug, in particular cannabis, actions. Here, we summarize known organizing principles of eCB-signaling systems in the developing telencephalon, and outline the sequence of decision points and underlying signaling pathways upon CB1 cannabinoid receptor activation that contribute to neuronal diversification in the developing brain. Finally, we discuss how these novel principles affect the formation of complex neuronal networks.


Asunto(s)
Tipificación del Cuerpo , Encéfalo/embriología , Encéfalo/metabolismo , Moduladores de Receptores de Cannabinoides/metabolismo , Endocannabinoides , Neuronas/citología , Neuronas/metabolismo , Animales , Encéfalo/citología , Humanos , Receptor Cannabinoide CB1/metabolismo , Transducción de Señal
13.
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
14.
J Neurosci ; 26(15): 4104-10, 2006 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-16611827

RESUMEN

In addition to classical genomic mechanisms, estrogen also exerts nonclassical effects via a signal transduction system on neurons. To study whether estrogen has a nonclassical effect on basal forebrain cholinergic system, we measured the intensity of cAMP response element-binding protein (CREB) phosphorylation (pCREB) in cholinergic neurons after administration of 17beta-estradiol to ovariectomized (OVX) mice. A significant time-dependent increase in the number of pCREB-positive cholinergic cells was detected after estrogen administration in the medial septum-diagonal band (MS-DB) and the substantia innominata (SI). The increase was first observed 15 min after estrogen administration. The role of classical estrogen receptors (ERs) was evaluated using ER knock-out mice in vivo. The estrogen-induced CREB phosphorylation in cholinergic neurons was present in ERbeta knock-out mice but completely absent in ERalpha knock-out mice in MS-DB and SI. A series of in vitro studies demonstrated that estrogen acted directly on cholinergic neurons. Selective blockade of the mitogen activated protein kinase (MAPK) pathway in vivo completely prevented estrogen-induced CREB phosphorylation in cholinergic neurons in MS-DB and SI. In contrast, blockade of protein kinase A (PKA) was effective only in SI. Finally, studies in intact female mice revealed levels of CREB phosphorylation within cholinergic neurons that were similar to those of estrogen-treated OVX mice. These observations demonstrate an ERalpha-mediated nonclassical effect of estrogen on the cholinergic neurons and that these actions are present under physiological conditions. They also reveal the role of MAPK and PKA-MAPK pathway activation in nonclassical estrogen signaling in the basal forebrain cholinergic neurons in vivo.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/fisiología , Neuronas/fisiología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/efectos de los fármacos , Estrógenos/sangre , Femenino , Isoquinolinas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovariectomía , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación , Sulfonamidas/farmacología
15.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA