Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.231
Filtrar
1.
Biosensors (Basel) ; 13(11)2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37998149

RESUMEN

Despite the prevalence of diabetic retinopathy, the majority of adult diabetic patients develop visually debilitating corneal complications, including impaired wound healing. Unfortunately, there is limited treatment for diabetes-induced corneal damage. The current project investigates a novel, peptide-based combination therapy, thymosin beta-4 and vasoactive intestinal peptide (Tß4/VIP), against high-glucose-induced damage to the corneal epithelium. Electric cell-substrate impedance sensing (ECIS) was used for real-time monitoring of barrier function and wound healing of human corneal epithelial cells maintained in either normal glucose (5 mM) or high glucose (25 mM) ± Tß4 (0.1%) and VIP (5 nM). Barrier integrity was assessed by resistance, impedance, and capacitance measurements. For the wound healing assay, cell migration was also monitored. Corneal epithelial tight junction proteins (ZO-1, ZO-2, occludin, and claudin-1) were assessed to confirm our findings. Barrier integrity and wound healing were significantly impaired under high-glucose conditions. However, barrier function and cell migration significantly improved with Tß4/VIP treatment. These findings were supported by high-glucose-induced downregulation of tight junction proteins that were effectively maintained similar to normal levels when treated with Tß4/VIP. These results strongly support the premise that Tß4 and VIP work synergistically to protect corneal epithelial cells against hyperglycemia-induced damage. In addition, this work highlights the potential for significant translational impact regarding the treatment of diabetic patients and associated complications of the cornea.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Humanos , Péptido Intestinal Vasoactivo/fisiología , Células Epiteliales , Glucosa , Proteínas de Uniones Estrechas
2.
HNO ; 71(5): 337-346, 2023 May.
Artículo en Alemán | MEDLINE | ID: mdl-37041304

RESUMEN

Allergic rhinitis (AR) is a very common disease with a high prevalence worldwide. It is an IgE-mediated type 2 inflammatory disease following exposure to inhalant allergens. A multitude of different neuropeptides including substance P, vasoactive intestinal peptide (VIP), calcitonin gene-related peptide (CGRP), nerve growth factor (NGF), and neuromedin U (NMU) can be released via peripheral axon or central reflexes, interact with immune cells, and thus contribute to neurogenic inflammation which causes the nasal hyperreactivity (NHR) characteristic of AR. Independent production of neuroendocrine hormones and neuropeptides by immune cells has also been demonstrated. Neuro-immune cell units arise when immune and neuronal cells colocalize, for which typical anatomic regions are, e.g., the mast cell-nerve functional unit. The focus of this review is the elucidation of neuroimmune communication mechanisms in AR.


Asunto(s)
Neuropéptidos , Rinitis Alérgica , Humanos , Neuroinmunomodulación , Neuropéptidos/análisis , Neuropéptidos/fisiología , Péptido Intestinal Vasoactivo/análisis , Péptido Intestinal Vasoactivo/fisiología , Péptido Relacionado con Gen de Calcitonina/análisis , Péptido Relacionado con Gen de Calcitonina/fisiología , Mucosa Nasal
3.
Endocrinology ; 163(2)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34978328

RESUMEN

Vasoactive intestinal peptide (Vip) regulates luteinizing hormone (LH) release through the direct regulation of gonadotropin-releasing hormone (GnRH) neurons at the level of the brain in female rodents. However, little is known regarding the roles of Vip in teleost reproduction. Although GnRH is critical for fertility through the regulation of LH secretion in vertebrates, the exact role of the hypophysiotropic GnRH (GnRH3) in zebrafish is unclear since GnRH3 null fish are reproductively fertile. This phenomenon raises the possibility of a redundant regulatory pathway(s) for LH secretion in zebrafish. Here, we demonstrate that VipA (homologues of mammalian Vip) both inhibits and induces LH secretion in zebrafish. Despite the observation that VipA axons may reach the pituitary proximal pars distalis including LH cells, pituitary incubation with VipA in vitro, and intraperitoneal injection of VipA, did not induce LH secretion and lhß mRNA expression in sexually mature females, respectively. On the other hand, intracerebroventricular administration of VipA augmented plasma LH levels in both wild-type and gnrh3-/- females at 1 hour posttreatment, with no observed changes in pituitary GnRH2 and GnRH3 contents and gnrh3 mRNA levels in the brains. While VipA's manner of inhibition of LH secretion has yet to be explored, the stimulation seems to occur via a different pathway than GnRH3, dopamine, and 17ß-estradiol in regulating LH secretion. The results indicate that VipA induces LH release possibly by acting with or through a non-GnRH factor(s), providing proof for the existence of functional redundancy of LH release in sexually mature female zebrafish.


Asunto(s)
Hormona Liberadora de Gonadotropina/fisiología , Hormona Luteinizante/metabolismo , Hipófisis/metabolismo , Ácido Pirrolidona Carboxílico/análogos & derivados , Péptido Intestinal Vasoactivo/fisiología , Pez Cebra , Animales , Anticuerpos/farmacología , Química Encefálica , Femenino , Técnicas de Inactivación de Genes , Hormona Liberadora de Gonadotropina/análisis , Hormona Liberadora de Gonadotropina/genética , Hormona Luteinizante/sangre , Hormona Luteinizante de Subunidad beta/genética , Hipófisis/química , Ácido Pirrolidona Carboxílico/análisis , ARN Mensajero/análisis , Péptido Intestinal Vasoactivo/administración & dosificación , Péptido Intestinal Vasoactivo/genética
4.
Mol Psychiatry ; 27(6): 2879-2900, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-33990774

RESUMEN

The hippocampus contains a diverse array of inhibitory interneurons that gate information flow through local cortico-hippocampal circuits to regulate memory storage. Although most studies of interneurons have focused on their role in fast synaptic inhibition mediated by GABA release, different classes of interneurons express unique sets of neuropeptides, many of which have been shown to exert powerful effects on neuronal function and memory when applied pharmacologically. However, relatively little is known about whether and how release of endogenous neuropeptides from inhibitory cells contributes to their behavioral role in regulating memory formation. Here we report that vasoactive intestinal peptide (VIP)-expressing interneurons participate in social memory storage by enhancing information transfer from hippocampal CA3 pyramidal neurons to CA2 pyramidal neurons. Notably, this action depends on release of the neuropeptide enkephalin from VIP neurons, causing long-term depression of feedforward inhibition onto CA2 pyramidal cells. Moreover, VIP neuron activity in the CA2 region is increased selectively during exploration of a novel conspecific. Our findings, thus, enhance our appreciation of how GABAergic neurons can regulate synaptic plasticity and mnemonic behavior by demonstrating that such actions can be mediated by release of a specific neuropeptide, rather than through classic fast inhibitory transmission.


Asunto(s)
Interneuronas , Péptido Intestinal Vasoactivo , Encefalinas/farmacología , Neuronas GABAérgicas , Hipocampo , Interneuronas/fisiología , Plasticidad Neuronal/fisiología , Células Piramidales/fisiología , Péptido Intestinal Vasoactivo/farmacología , Péptido Intestinal Vasoactivo/fisiología
5.
Nat Neurosci ; 23(12): 1629-1636, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32807948

RESUMEN

Recent success in identifying gene-regulatory elements in the context of recombinant adeno-associated virus vectors has enabled cell-type-restricted gene expression. However, within the cerebral cortex these tools are largely limited to broad classes of neurons. To overcome this limitation, we developed a strategy that led to the identification of multiple new enhancers to target functionally distinct neuronal subtypes. By investigating the regulatory landscape of the disease gene Scn1a, we discovered enhancers selective for parvalbumin (PV) and vasoactive intestinal peptide-expressing interneurons. Demonstrating the functional utility of these elements, we show that the PV-specific enhancer allowed for the selective targeting and manipulation of these neurons across vertebrate species, including humans. Finally, we demonstrate that our selection method is generalizable and characterizes additional PV-specific enhancers with exquisite specificity within distinct brain regions. Altogether, these viral tools can be used for cell-type-specific circuit manipulation and hold considerable promise for use in therapeutic interventions.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Interneuronas/fisiología , Animales , Callithrix , Corteza Cerebral/citología , Femenino , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.1/genética , Neuronas , Parvalbúminas/fisiología , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Péptido Intestinal Vasoactivo/fisiología
6.
Diabetes ; 69(7): 1549-1561, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32345752

RESUMEN

Diabetic keratopathy, a sight-threatening corneal disease, comprises several symptomatic conditions including delayed epithelial wound healing, recurrent erosions, and sensory nerve (SN) neuropathy. We investigated the role of neuropeptides in mediating corneal wound healing, including epithelial wound closure and SN regeneration. Denervation by resiniferatoxin severely impaired corneal wound healing and markedly upregulated proinflammatory gene expression. Exogenous neuropeptides calcitonin gene-related peptide (CGRP), substance P (SP), and vasoactive intestinal peptide (VIP) partially reversed resiniferatoxin's effects, with VIP specifically inducing interleukin-10 expression. Hence, we focused on VIP and observed that wounding induced VIP and VIP type 1 receptor (VIPR1) expression in normal (NL) corneas, but not corneas from mice with diabetes mellitus (DM). Targeting VIPR1 in NL corneas attenuated corneal wound healing, dampened wound-induced expression of neurotrophic factors, and exacerbated inflammatory responses, while exogenous VIP had the opposite effects in DM corneas. Remarkably, wounding and diabetes also affected the expression of Sonic Hedgehog (Shh) in a VIP-dependent manner. Downregulating Shh expression in NL corneas decreased while exogenous Shh in DM corneas increased the rates of corneal wound healing. Furthermore, inhibition of Shh signaling dampened VIP-promoted corneal wound healing. We conclude that VIP regulates epithelial wound healing, inflammatory response, and nerve regeneration in the corneas in an Shh-dependent manner, suggesting a therapeutic potential for these molecules in treating diabetic keratopathy.


Asunto(s)
Enfermedades de la Córnea/fisiopatología , Diabetes Mellitus Experimental/complicaciones , Epitelio Corneal/fisiopatología , Proteínas Hedgehog/fisiología , Regeneración Nerviosa/fisiología , Péptido Intestinal Vasoactivo/fisiología , Cicatrización de Heridas/fisiología , Animales , Citocinas/análisis , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/fisiología , Transducción de Señal/fisiología
7.
Neuropeptides ; 81: 102031, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32143816

RESUMEN

Glucagon-like peptide-2 (GLP-2) has been reported to indirectly relax gastric smooth muscle. In the present study we investigated, through a combined mechanical and immunohistochemical approach, whether GLP-2 interferes with the electrical field stimulation (EFS)-induced vipergic relaxant responses and the mechanism through which it occurs. For functional experiments, strips from the mouse gastric fundus were mounted in organ baths for isometric recording of the mechanical activity. Vasoactive intestinal peptide (VIP) immunoreactivity in GLP-2 exposed specimens was also evaluated by immunohistochemistry. In carbachol pre-contracted strips, GLP-2 (20 nM) evoked a tetrodotoxin (TTX)-sensitive relaxation, similar in shape to the TTX-insensitive of 100 nM VIP. In the presence of GLP-2, VIP had no longer effects and no more response to GLP-2 was observed following VIP receptor saturation. EFS (4-16 Hz) induced a fast relaxant response followed, at the higher stimulation frequencies (≥ 8 Hz), by a slow one. This latter was abolished either by GLP-2 or VIP receptor saturation as well as by the VIP receptor antagonist, VIP 6-28 (10 µM). A decrease of VIP-immunoreactive nerve structures in the GLP-2 exposed specimens was observed. These results suggest that, in the mouse gastric fundus, GLP-2 influences the EFS-induced slow relaxant response by promoting neuronal VIP release.


Asunto(s)
Fundus Gástrico/fisiología , Péptido 2 Similar al Glucagón/fisiología , Neuronas/fisiología , Péptido Intestinal Vasoactivo/fisiología , Animales , Femenino , Ratones , Contracción Muscular/fisiología , Músculo Liso/fisiología
8.
Acta Neurobiol Exp (Wars) ; 79(4): 328-337, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31885390

RESUMEN

For years, interneurons expressing vasoactive intestinal peptide (VIP) interneurons and their function within the neocortex have been shrouded in mystery. Their relatively small size and minimal representation in the cortex have made investigation difficult. Due to their service role performed in co­operation with glia and blood vessels to supply energy during neuronal activation in the brain, the contribution of VIP interneurons to local neuronal circuit function was not appreciated. VIP interneurons in the neocortex account for roughly 12% of all interneurons. They have been described as a subgroup of the third largest population of 5-hydroxytryptamine 3a (5HT3a) receptor­expressing interneurons, non­overlapping with interneuron populations expressing parvalbumin (PV) or somatostatin (SST). However, it was recently shown that only half of VIP interneurons display a 5HT3a receptor response and a subset of VIP interneurons in visual cortex co­express SST. Over the last several years, due to new technical advancements, many facts have emerged relating to VIP interneuron phylogenetic origin, operational mechanisms within local circuits and functional significance. Some of these discoveries have dramatically shifted the perception of VIP interneurons. This review focuses on the function of the VIP interneurons residing in layer 2/3 of the mouse neocortex.


Asunto(s)
Interneuronas/fisiología , Neocórtex/citología , Péptido Intestinal Vasoactivo/fisiología , Potenciales de Acción , Animales , Conducta Animal/fisiología , Conectoma , Interneuronas/química , Interneuronas/clasificación , Aprendizaje/fisiología , Locomoción/fisiología , Ratones , Filogenia , Células Piramidales/fisiología , Receptores de Serotonina 5-HT3/metabolismo , Sensación/fisiología , Somatostatina/análisis , Visión Ocular/fisiología , Corteza Visual/citología
9.
Nat Neurosci ; 22(11): 1834-1843, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31636447

RESUMEN

Learning drives behavioral adaptations necessary for survival. While plasticity of excitatory projection neurons during associative learning has been extensively studied, little is known about the contributions of local interneurons. Using fear conditioning as a model for associative learning, we found that behaviorally relevant, salient stimuli cause learning by tapping into a local microcircuit consisting of precisely connected subtypes of inhibitory interneurons. By employing deep-brain calcium imaging and optogenetics, we demonstrate that vasoactive intestinal peptide (VIP)-expressing interneurons in the basolateral amygdala are activated by aversive events and provide a mandatory disinhibitory signal for associative learning. Notably, VIP interneuron responses during learning are strongly modulated by expectations. Our findings indicate that VIP interneurons are a central component of a dynamic circuit motif that mediates adaptive disinhibitory gating to specifically learn about unexpected, salient events, thereby ensuring appropriate behavioral adaptations.


Asunto(s)
Aprendizaje por Asociación/fisiología , Interneuronas/fisiología , Inhibición Neural/fisiología , Filtrado Sensorial/fisiología , Péptido Intestinal Vasoactivo/fisiología , Amígdala del Cerebelo/fisiología , Animales , Condicionamiento Psicológico/fisiología , Miedo/psicología , Femenino , Masculino , Ratones , Ratones Transgénicos , Optogenética
10.
F1000Res ; 82019.
Artículo en Inglés | MEDLINE | ID: mdl-31559013

RESUMEN

Vasoactive intestinal peptide (VIP), a gut peptide hormone originally reported as a vasodilator in 1970, has multiple physiological and pathological effects on development, growth, and the control of neuronal, epithelial, and endocrine cell functions that in turn regulate ion secretion, nutrient absorption, gut motility, glycemic control, carcinogenesis, immune responses, and circadian rhythms. Genetic ablation of this peptide and its receptors in mice also provides new insights into the contribution of VIP towards physiological signaling and the pathogenesis of related diseases. Here, we discuss the impact of VIP on gastrointestinal function and diseases based on recent findings, also providing insight into its possible therapeutic application to diabetes, autoimmune diseases and cancer.


Asunto(s)
Enfermedades Gastrointestinales , Tracto Gastrointestinal , Péptido Intestinal Vasoactivo , Animales , Enfermedades Gastrointestinales/metabolismo , Tracto Gastrointestinal/fisiología , Ratones , Receptores de Tipo II del Péptido Intestinal Vasoactivo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo , Transducción de Señal , Péptido Intestinal Vasoactivo/fisiología
11.
BMC Ophthalmol ; 19(1): 190, 2019 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-31429729

RESUMEN

BACKGROUND: Exploring the role of vasoactive intestinal peptide (VIP) in the lateral geniculate body (LGBd) in visual development and studying the therapeutic effect of VIP on amblyopic kittens. METHODS: Three-week-old domestic cats were divided into a control group (n = 10) and a monocular deprivation group (n = 20), with an eye mask covering the right eye of those in the deprived group. After pattern visual evoked potential (PVEP) recording confirmed the formation of monocular amblyopia, the left LGBd was isolated from 5 kittens in each group. The remaining control kittens continued to be raised, and the remaining deprivation group was divided into a VIP intervention group (n = 5), Sefsol (caprylic acid monoglyceride, VIP solution) intervention group (n = 5) and amblyopia non-intervention group (n = 5) after removal of the eye mask. Three weeks later, PVEPs, VIP immunohistochemistry and VIP mRNA expression in the left LGBd were compared across groups. RESULTS: At 6 weeks of age, there were significant differences in P100 wave latency and amplitude and VIP immunohistochemistry and in situ hybridization between the control group and the deprivation group (P < 0.05). After 3 weeks of the corresponding interventions, the latency and amplitude in the VIP intervention group were better than that in the Sefsol intervention group and amblyopia non-intervention group (P < 0.05). Furthermore, VIP treatment increased the number of immunohistochemical VIP-positive cells (P < 0.05) and the average optical density of positive cells (P > 0.05), as well as the number (P < 0.05) and average optical density of VIP mRNA-positive cells (P < 0.05). CONCLUSIONS: VIP plays an important role in visual development. Nasal administration of VIP can improve the function of neurons in the LGBd of kittens and has a certain therapeutic effect on amblyopia.


Asunto(s)
Ambliopía/fisiopatología , Privación Sensorial/fisiología , Péptido Intestinal Vasoactivo/fisiología , Animales , Gatos , Modelos Animales de Enfermedad , Potenciales Evocados Visuales/fisiología , Femenino , Inmunohistoquímica , Corteza Visual/fisiología
12.
Cell Biol Int ; 43(7): 770-780, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31026365

RESUMEN

Acute respiratory distress syndrome (ARDS) is an acute, severe, and refractory pulmonary inflammation with high morbidity and mortality. Excessive activation of fibroblast during the fibroproliferative phase plays a pivotal role in the prognosis of ARDS. Our previous study demonstrated that the vasoactive intestinal peptide (VIP) is mediated by lentivirus attenuates lipopolysaccharide (LPS)-induced ARDS in a murine model, and VIP inhibits the release of interleukin-17A (IL-17A) from activation macrophages. However, the effects of VIP on the activation of murine fibroblast and expression of IL-17 receptor (IL-17R) in ARDS remain unclear. Here, a mouse model of ARDS was established by an intratracheal injection of LPS. We found that the gene expression of col3a1 and hydroxyproline contents in the lungs were significantly increased 24 h after LPS injection. IL-17RC rather than IL-17RA was increased in the lungs of mice with ARDS. In vitro, LPS activated NIH3T3 cells, which was suppressed by VIP in a dose-dependent manner. In detail, VIP reduced the hydroxyproline content and col3a1 messenger RNA induced by LPS in NIH3T3 cells, as well as the expression of α-smooth muscle actin. Furthermore, we found that VIP inhibited the expression of IL-17R in the lungs of mice with ARDS and NIH3T3 cells stimulated with LPS, which was partly inhibited by antagonists of protein kinase A and protein kinase C. Taken together, our results demonstrated that VIP inhibited the activation of fibroblast via downregulation of IL-17RC, which may contribute to the protective effects of VIP against ARDS in mice.


Asunto(s)
Fibroblastos/inmunología , Receptores de Interleucina/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Transducción de Señal/efectos de los fármacos , Péptido Intestinal Vasoactivo , Actinas/metabolismo , Animales , Colágeno Tipo III/metabolismo , Modelos Animales de Enfermedad , Hidroxiprolina/metabolismo , Lipopolisacáridos/química , Masculino , Ratones , Células 3T3 NIH , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Interleucina-17/inmunología , Péptido Intestinal Vasoactivo/farmacología , Péptido Intestinal Vasoactivo/fisiología
13.
Neurosci Res ; 141: 13-22, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30110598

RESUMEN

Executive control supports our ability to behave flexibly in accordance with a given situation. In order to fully understand how cortical circuits achieve this task, we need to determine the intrinsic physiological and connection profiles of neuron types and analyze their functional roles during behavior. This article introduces current knowledge regarding neuron type classification in the cortex and reviews our understanding of how each neuron type is incorporated in the functional cortical circuit to implement executive control. Recent work using neuron-type specific imaging/recording has begun to reveal significant functional organizations of pyramidal neurons and their subtypes depending on the layers and long-range projection targets. GABAergic interneurons also make crucial contributions to executive control in a subtype-specific manner. Vasoactive intestinal peptide (VIP)-positive interneurons are preferentially recruited by top-down inputs from higher-order cortical regions and amplify the signals in pyramidal neurons by inhibiting other interneuron subtypes. Particularly in the prefrontal cortex, one of the hierarchically highest cortices, executive control signals are regulated by the VIP neuron-mediated disinhibition and robustly maintained through recurrent connections at a long time scale. The differences and commonalities in the functional organization between sensory areas and the prefrontal cortex are discussed.


Asunto(s)
Función Ejecutiva/fisiología , Neuronas/fisiología , Animales , Neuronas GABAérgicas/fisiología , Humanos , Interneuronas/fisiología , Modelos Neurológicos , Inhibición Neural , Vías Nerviosas/fisiología , Células Piramidales/fisiología , Péptido Intestinal Vasoactivo/fisiología
14.
J Neurosci ; 39(5): 788-801, 2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30523065

RESUMEN

Hippocampus-dependent learning processes are coordinated via a large diversity of GABAergic inhibitory mechanisms. The α5 subunit-containing GABAA receptor (α5-GABAAR) is abundantly expressed in the hippocampus populating primarily the extrasynaptic domain of CA1 pyramidal cells, where it mediates tonic inhibitory conductance and may cause functional deficits in synaptic plasticity and hippocampus-dependent memory. However, little is known about synaptic expression of the α5-GABAAR and, accordingly, its location site-specific function. We examined the cell- and synapse-specific distribution of the α5-GABAAR in the CA1 stratum oriens/alveus (O/A) using a combination of immunohistochemistry, whole-cell patch-clamp recordings and optogenetic stimulation in hippocampal slices obtained from mice of either sex. In addition, the input-specific role of the α5-GABAAR in spatial learning and anxiety-related behavior was studied using behavioral testing and chemogenetic manipulations. We demonstrate that α5-GABAAR is preferentially targeted to the inhibitory synapses made by the vasoactive intestinal peptide (VIP)- and calretinin-positive terminals onto dendrites of somatostatin-expressing interneurons. In contrast, synapses made by the parvalbumin-positive inhibitory inputs to O/A interneurons showed no or little α5-GABAAR. Inhibiting the α5-GABAAR in control mice in vivo improved spatial learning but also induced anxiety-like behavior. Inhibiting the α5-GABAAR in mice with inactivated CA1 VIP input could still improve spatial learning and was not associated with anxiety. Together, these data indicate that the α5-GABAAR-mediated phasic inhibition via VIP input to interneurons plays a predominant role in the regulation of anxiety while the α5-GABAAR tonic inhibition via this subunit may control spatial learning.SIGNIFICANCE STATEMENT The α5-GABAAR subunit exhibits high expression in the hippocampus, and regulates the induction of synaptic plasticity and the hippocampus-dependent mnemonic processes. In CA1 principal cells, this subunit occupies mostly extrasynaptic sites and mediates tonic inhibition. Here, we provide evidence that, in CA1 somatostatin-expressing interneurons, the α5-GABAAR subunit is targeted to synapses formed by the VIP- and calretinin-expressing inputs, and plays a specific role in the regulation of anxiety-like behavior.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Neuronas/metabolismo , Receptores de GABA-A/metabolismo , Sinapsis/metabolismo , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/efectos de los fármacos , Calbindina 2/fisiología , Femenino , Antagonistas de Receptores de GABA-A/farmacología , Interneuronas/efectos de los fármacos , Interneuronas/fisiología , Interneuronas/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/ultraestructura , Optogenética , Técnicas de Placa-Clamp , Somatostatina/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura , Péptido Intestinal Vasoactivo/fisiología
15.
PLoS Comput Biol ; 14(12): e1006607, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30532130

RESUMEN

Circadian clocks are autonomous oscillators driving daily rhythms in physiology and behavior. In mammals, a network of coupled neurons in the suprachiasmatic nucleus (SCN) is entrained to environmental light-dark cycles and orchestrates the timing of peripheral organs. In each neuron, transcriptional feedbacks generate noisy oscillations. Coupling mediated by neuropeptides such as VIP and AVP lends precision and robustness to circadian rhythms. The detailed coupling mechanisms between SCN neurons are debated. We analyze organotypic SCN slices from neonatal and adult mice in wild-type and multiple knockout conditions. Different degrees of rhythmicity are quantified by pixel-level analysis of bioluminescence data. We use empirical orthogonal functions (EOFs) to characterize spatio-temporal patterns. Simulations of coupled stochastic single cell oscillators can reproduce the diversity of observed patterns. Our combination of data analysis and modeling provides deeper insight into the enormous complexity of the data: (1) Neonatal slices are typically stronger oscillators than adult slices pointing to developmental changes of coupling. (2) Wild-type slices are completely synchronized and exhibit specific spatio-temporal patterns of phases. (3) Some slices of Cry double knockouts obey impaired synchrony that can lead to co-existing rhythms ("splitting"). (4) The loss of VIP-coupling leads to desynchronized rhythms with few residual local clusters. Additional information was extracted from co-culturing slices with rhythmic neonatal wild-type SCNs. These co-culturing experiments were simulated using external forcing terms representing VIP and AVP signaling. The rescue of rhythmicity via co-culturing lead to surprising results, since a cocktail of AVP-antagonists improved synchrony. Our modeling suggests that these counter-intuitive observations are pointing to an antagonistic action of VIP and AVP coupling. Our systematic theoretical and experimental study shows that dual coupling mechanisms can explain the astonishing complexity of spatio-temporal patterns in SCN slices.


Asunto(s)
Arginina Vasopresina/metabolismo , Ritmo Circadiano/fisiología , Péptido Intestinal Vasoactivo/metabolismo , Animales , Arginina Vasopresina/fisiología , Relojes Circadianos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Neuropéptidos/metabolismo , Proteínas Circadianas Period/metabolismo , Transducción de Señal , Núcleo Supraquiasmático/fisiología , Péptido Intestinal Vasoactivo/fisiología
16.
J Neuroendocrinol ; 30(12): e12660, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30422333

RESUMEN

A population of kisspeptin neurones located in the hypothalamic arcuate nucleus (ARN) very likely represent the gonadotrophin-releasing hormone pulse generator responsible for driving pulsatile luteinising hormone secretion in mammals. As such, it has become important to understand the neural inputs that modulate the activity of ARN kisspeptin (ARNKISS ) neurones. Using a transgenic GCaMP6 mouse model allowing the intracellular calcium levels ([Ca2+ ]i ) of individual ARNKISS neurones to be assessed simultaneously, we examined whether the circadian neuropeptides vasoactive intestinal peptide (VIP) and arginine vasopressin (AVP) modulated the activity of ARNKISS neurones directly. To validate this methodology, we initially evaluated the effects of neurokinin B (NKB) on [Ca2+ ]i in kisspeptin neurones residing within the rostral, middle and caudal ARN subregions of adult male and female mice. All experiments were undertaken in the presence of tetrodotoxin and ionotropic amino acid antagonists. NKB was found to evoke an abrupt increase in [Ca2+ ]i in 95%-100% of kisspeptin neurones throughout the ARN of both sexes. By contrast, both VIP and AVP were found to primarily activate kisspeptin neurones located in the caudal ARN of female mice. Although 58% and 59% of caudal ARN kisspeptin neurones responded to AVP and VIP, respectively, in female mice, only 0%-8% of kisspeptin neurones located in other ARN subregions responded in females and 0%-12% of cells in any subregion in males (P < 0.05). These observations demonstrate unexpected sex differences and marked heterogeneity in functional neuropeptide receptor expression amongst ARNKISS neurones organised on a rostro-caudal basis. The functional significance of this unexpected influence of VIP and AVP on ARNKISS neurones remains to be established.


Asunto(s)
Núcleo Arqueado del Hipotálamo/citología , Kisspeptinas/metabolismo , Neuronas/metabolismo , Caracteres Sexuales , Péptido Intestinal Vasoactivo/fisiología , Vasopresinas/fisiología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Calcio/metabolismo , Femenino , Masculino , Ratones , Ratones Transgénicos , Neuroquinina B/farmacología , Neuronas/efectos de los fármacos , Péptido Intestinal Vasoactivo/farmacología , Vasopresinas/farmacología
17.
Nat Neurosci ; 21(6): 851-859, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29786081

RESUMEN

How learning enhances neural representations for behaviorally relevant stimuli via activity changes of cortical cell types remains unclear. We simultaneously imaged responses of pyramidal cells (PYR) along with parvalbumin (PV), somatostatin (SOM), and vasoactive intestinal peptide (VIP) inhibitory interneurons in primary visual cortex while mice learned to discriminate visual patterns. Learning increased selectivity for task-relevant stimuli of PYR, PV and SOM subsets but not VIP cells. Strikingly, PV neurons became as selective as PYR cells, and their functional interactions reorganized, leading to the emergence of stimulus-selective PYR-PV ensembles. Conversely, SOM activity became strongly decorrelated from the network, and PYR-SOM coupling before learning predicted selectivity increases in individual PYR cells. Thus, learning differentially shapes the activity and interactions of multiple cell classes: while SOM inhibition may gate selectivity changes, PV interneurons become recruited into stimulus-specific ensembles and provide more selective inhibition as the network becomes better at discriminating behaviorally relevant stimuli.


Asunto(s)
Interneuronas/fisiología , Aprendizaje/fisiología , Corteza Visual/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Aprendizaje Discriminativo/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/citología , Red Nerviosa/fisiología , Parvalbúminas/fisiología , Técnicas de Placa-Clamp , Patrones de Reconocimiento Fisiológico/fisiología , Células Piramidales/metabolismo , Células Piramidales/fisiología , Filtrado Sensorial/fisiología , Somatostatina/fisiología , Péptido Intestinal Vasoactivo/fisiología , Corteza Visual/citología
18.
Nat Neurosci ; 21(5): 717-724, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29632359

RESUMEN

'Sundowning' in dementia and Alzheimer's disease is characterized by early-evening agitation and aggression. While such periodicity suggests a circadian origin, whether the circadian clock directly regulates aggressive behavior is unknown. We demonstrate that a daily rhythm in aggression propensity in male mice is gated by GABAergic subparaventricular zone (SPZGABA) neurons, the major postsynaptic targets of the central circadian clock, the suprachiasmatic nucleus. Optogenetic mapping revealed that SPZGABA neurons receive input from vasoactive intestinal polypeptide suprachiasmatic nucleus neurons and innervate neurons in the ventrolateral part of the ventromedial hypothalamus (VMH), which is known to regulate aggression. Additionally, VMH-projecting dorsal SPZ neurons are more active during early day than early night, and acute chemogenetic inhibition of SPZGABA transmission phase-dependently increases aggression. Finally, SPZGABA-recipient central VMH neurons directly innervate ventrolateral VMH neurons, and activation of this intra-VMH circuit drove attack behavior. Altogether, we reveal a functional polysynaptic circuit by which the suprachiasmatic nucleus clock regulates aggression.


Asunto(s)
Agresión/fisiología , Ritmo Circadiano/fisiología , Hipotálamo/fisiología , Vías Nerviosas/fisiología , Animales , Mapeo Encefálico , Corticosterona/sangre , Potenciales Postsinápticos Excitadores/fisiología , Hipotálamo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/citología , Optogenética , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/fisiología , Núcleo Supraquiasmático/fisiología , Péptido Intestinal Vasoactivo/fisiología , Núcleo Hipotalámico Ventromedial/citología , Núcleo Hipotalámico Ventromedial/fisiología , Ácido gamma-Aminobutírico/fisiología
19.
J Neurosci ; 38(6): 1326-1334, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29054877

RESUMEN

In mammals, the suprachiasmatic nucleus (SCN) of the hypothalamus coordinates daily rhythms including sleep-wake, hormone release, and gene expression. The cells of the SCN must synchronize to each other to drive these circadian rhythms in the rest of the body. The ontogeny of circadian cycling and intercellular coupling in the SCN remains poorly understood. Recent in vitro studies have recorded circadian rhythms from the whole embryonic SCN. Here, we tracked the onset and precision of rhythms in PERIOD2 (PER2), a clock protein, within the SCN isolated from embryonic and postnatal mice of undetermined sex. We found that a few SCN cells developed circadian periodicity in PER2 by 14.5 d after mating (E14.5) with no evidence for daily cycling on E13.5. On E15.5, the fraction of competent oscillators increased dramatically corresponding with stabilization of their circadian periods. The cells of the SCN harvested at E15.5 expressed sustained, synchronous daily rhythms. By postnatal day 2 (P2), SCN oscillators displayed the daily, dorsal-ventral phase wave in clock gene expression typical of the adult SCN. Strikingly, vasoactive intestinal polypeptide (VIP), a neuropeptide critical for synchrony in the adult SCN, and its receptor, VPAC2R, reached detectable levels after birth and after the onset of circadian synchrony. Antagonists of GABA or VIP signaling or action potentials did not disrupt circadian synchrony in the E15.5 SCN. We conclude that endogenous daily rhythms in the fetal SCN begin with few noisy oscillators on E14.5, followed by widespread oscillations that rapidly synchronize on E15.5 by an unknown mechanism.SIGNIFICANCE STATEMENT We recorded the onset of PER2 circadian oscillations during embryonic development in the mouse SCN. When isolated at E13.5, the anlagen of the SCN expresses high, arrhythmic PER2. In contrast, a few cells show noisy circadian rhythms in the isolated E14.5 SCN and most show reliable, self-sustained, synchronized rhythms in the E15.5 SCN. Strikingly, this synchrony at E15.5 appears before expression of VIP or its receptor and persists in the presence of blockers of VIP, GABA or neuronal firing. Finally, the dorsal-ventral phase wave of PER2 typical of the adult SCN appears ∼P2, indicating that multiple signals may mediate circadian synchrony during the ontogeny of the SCN.


Asunto(s)
Ritmo Circadiano/fisiología , Núcleo Supraquiasmático/fisiología , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Femenino , Antagonistas del GABA/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/fisiología , Embarazo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/biosíntesis , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/crecimiento & desarrollo , Péptido Intestinal Vasoactivo/antagonistas & inhibidores , Péptido Intestinal Vasoactivo/metabolismo , Péptido Intestinal Vasoactivo/fisiología
20.
J Physiol ; 596(5): 901-919, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29274075

RESUMEN

KEY POINTS: An increase in the excitability of GABAergic cells has typically been assumed to decrease network activity, potentially producing overall anti-epileptic effects. Recent data suggest that inhibitory networks may actually play a role in initiating epileptiform activity. We show that activation of GABAergic interneurons can elicit synchronous long-lasting network activity. Specific interneuron subpopulations differentially contributed to GABA network synchrony, indicating cell type-specific contributions of interneurons to cortical network activity. Interneurons may critically contribute to the generation of aberrant network activity characteristic of epilepsy, warranting further investigation into the contribution of distinct cortical interneuron subpopulations to the propagation and rhythmicity of epileptiform activity. ABSTRACT: In the presence of the A-type K+ channel blocker 4-aminopyrdine, spontaneous synchronous network activity develops in the neocortex of mice of either sex. This aberrant synchrony persists in the presence of excitatory amino acid receptor antagonists (EAA blockers) and is considered to arise from synchronous firing of cortical interneurons (INs). Although much attention has been given to the mechanisms underlying this GABAergic synchrony, the contribution of specific IN subtypes to the generation of these long-lasting discharges (LLDs) is incompletely understood. We employed genetically-encoded channelrhodopsin and archaerhodopsin opsins to investigate the sufficiency and necessity, respectively, of activation of parvalbumin (PV), somatostatin (SST) and vasointestinal peptide (VIP)-expressing INs for the generation of synchronous neocortical GABAergic discharges. We found light-induced activation of PV or SST INs to be equally sufficient for the generation of LLDs, whereas activation of VIP INs was not. By contrast, light-induced inhibition of PV INs strongly reduced LLD initiation, whereas suppression of SST or VIP IN activity only partially attenuated LLD magnitude. These results suggest neocortical INs perform cell type-specific roles in the generation of aberrant GABAergic cortical network activity.


Asunto(s)
Neuronas GABAérgicas/fisiología , Hormonas/farmacología , Interneuronas/fisiología , Neocórtex/fisiología , Optogenética , Parvalbúminas/fisiología , Somatostatina/fisiología , Péptido Intestinal Vasoactivo/fisiología , Potenciales de Acción , Animales , Animales Recién Nacidos , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Interneuronas/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Neocórtex/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...