Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.294
Filtrar
Más filtros

Intervalo de año de publicación
1.
Acta Med Okayama ; 78(2): 95-106, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38688827

RESUMEN

The lungs are very complex organs, and the respiratory system performs the dual roles of repairing tissue while protecting against infection from various environmental stimuli. Persistent external irritation disrupts the immune responses of tissues and cells in the respiratory system, ultimately leading to respiratory disease. Neuropeptide Y (NPY) is a 36-amino-acid polypeptide and a neurotransmitter that regulates homeostasis. The NPY receptor is a seven-transmembrane-domain G-protein-coupled receptor with six subtypes (Y1, Y2, Y3, Y4, Y5, and Y6). Of these receptors, Y1, Y2, Y4, and Y5 are functional in humans, and Y1 plays important roles in the immune responses of many organs, including the respiratory system. NPY and the Y1 receptor have critical roles in the pathogenesis of asthma, chronic obstructive pulmonary disease, and idiopathic pulmonary fibrosis. The effects of NPY on the airway immune response and pathogenesis differ among respiratory diseases. This review focuses on the involvement of NPY in the airway immune response and pathogenesis of various respiratory diseases.


Asunto(s)
Neuropéptido Y , Receptores de Neuropéptido Y , Humanos , Neuropéptido Y/fisiología , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/fisiología , Animales , Enfermedades Respiratorias/inmunología , Asma/inmunología , Sistema Respiratorio/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología
2.
Gen Comp Endocrinol ; 315: 113930, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34673032

RESUMEN

Animal research indicates the neuropeptide Y (NPY), corticotrophin and melanocortin systems have a mediatory role in reward, however, how these substances interact with phenytoin-14 (PNX-14) induced food intake in birds remains to be identified. Accordingly, in this research eight tests were carried out to investigate the potential interactions of the NPY, melanocortin, as well as corticotrophin systems with PNX-14 on food consumption in neonatal chickens. In the first experiment, chickens were intracerebroventricular (ICV) injected with phosphate-buffered saline (PBS) and PNX-14 (0.8, 0.16, and 3.2 nmol). In second experiment, PBS, the antagonist of CRF1/CRF2 receptors (astressin-B, 30 µg) and PNX-14 + astressin-B were injected. In the rest of the experiments chicken received astressin2-B (CRF2 receptor antagonist; 30 µg), SHU9119 (MCR3/MCR4 receptor antagonist, 0.5nomol), MCL0020 (MCR4 receptor agonist, 0.5 nmol), B5063 (NPY1 receptor antagonist, 1.25 µg), SF22 (NPY2 receptor antagonist, 1.25 µg) and SML0891 (NPY5 receptor antagonist, 1.25 µg) rather than astressin-B. Then, cumulative intake of food was recorded for 2 h. Based on the findings, PNX-14 (0.16 and 3.2 nmol) led to increment in food consumption compared with the control (P < 0.05). Co-administration of the PNX-14 and astressin-B promoted PNX-14-induced hyperphagia (P < 0.05). Co-injection of the PNX-14 + astressin2-B potentiated hyperphagia PNX-14 (P < 0.05). Co-injection of PNX-14 + B5063 inhibited the effects of the PNX-14 (P < 0.05). The co-administration of the PNX-14 and SML0891 potentiated hypophagic effects of the PNX-14 (P < 0.05). The results showed that PNX-14-induced hyperphagia mediates via NPY1, NPY5, and CRF1/CRF2 receptors in neonatal chickens.


Asunto(s)
Hormona Adrenocorticotrópica , Pollos , Ingestión de Alimentos , Melanocortinas , Neuropéptido Y , Hormona Adrenocorticotrópica/fisiología , Animales , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Hormonas Hipotalámicas/farmacología , Melanocortinas/uso terapéutico , Neuropéptido Y/fisiología , Hormonas Peptídicas/farmacología
3.
J Neurosci ; 40(31): 6018-6034, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32576621

RESUMEN

Aversive learning is fundamental for animals to increase chances of survival. In addition to classical neurotransmitters, neuropeptides have emerged to modulate such complex behaviors. Among them, neuropeptide Y (NPY) is well known to promote aversive memory acquisition in mammals. Here we identify an NPY/neuropeptide F (NPF)-related neuropeptide system in Caenorhabditis elegans and show that this FLP-34/NPR-11 system is required for learning negative associations, a process that is reminiscent of NPY signaling in mammals. The Caenorhabditis elegans NPY/NPF ortholog FLP-34 displays conserved structural hallmarks of bilaterian-wide NPY/NPF neuropeptides. We show that it is required for aversive olfactory learning after pairing diacetyl with the absence of food, but not for appetitive olfactory learning in response to butanone. To mediate diacetyl learning and thus integrate the aversive food context with the diacetyl odor, FLP-34 is released from serotonergic neurons and signals through its evolutionarily conserved NPY/NPF GPCR, NPR-11, in downstream AIA interneurons. NPR-11 activation in the AIA integration center results in avoidance of a previously attractive stimulus. This study opens perspectives for a deeper understanding of stress conditions in which aversive learning results in excessive avoidance.SIGNIFICANCE STATEMENT Aversive learning evolved early in evolution to promote avoidance of dangerous and stressful situations. In addition to classical neurotransmitters, neuropeptides are emerging as modulators of complex behaviors, including learning and memory. Here, we identified the evolutionary ortholog of neuropeptide Y/neuropeptide F in the nematode Caenorhabditis elegans, and we discovered that it is required for olfactory aversive learning. In addition, we elucidated the neural circuit underlying this avoidance behavior, and we discovered a novel coordinated action of Caenorhabditis elegans neuropeptide Y/neuropeptide F and serotonin that could aid in our understanding of the molecular mechanisms underlying stress disorders in which excessive avoidance results in maladaptive behaviors.


Asunto(s)
Aprendizaje por Asociación/fisiología , Neuropéptido Y/fisiología , Neuropéptidos/fisiología , Neuronas Serotoninérgicas/fisiología , Olfato/fisiología , Animales , Conducta Apetitiva , Reacción de Prevención/efectos de los fármacos , Butanonas/farmacología , Caenorhabditis elegans , Diacetil/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica , Locomoción , Masculino , Neuropéptido Y/genética , Neuropéptidos/genética
4.
Int J Neurosci ; 131(8): 780-788, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32303141

RESUMEN

AIMS: A number of studies have shown that neuropeptide Y (NPY) is considered to be one of the key regulators of hypothalamic-pituitary-gonadal (HPG) axis in the mammals. In addition, kisspeptin (encode by Kiss1 gene), neurokinin B (encode by Tac3 gene) and dynorphin (encode by Pdyn gene) (commonly known as KNDy secreting neurons) are a powerful upstream regulators of GnRH neuron in hypothalamus. MATERIALS AND METHODS: The present study aims to investigate the effects of the intracerebroventricular (icv) injection of NPY and BIBP3226 (NPY receptor antagonist (NPYRA)) on the male sexual behavioral. Additionally, in order to see whether NPY signals can be relayed through the pathway of kisspeptin/neurokinin B/dynorphin, the gene expression of these peptides along with Gnrh1 gene in the hypothalamus were measured. RESULTS: The icv injection of NPY decreased the latencies and increase the frequencies of sexual parameters of the male rats in a significant way. In this line, NPYRA antagonized the stimulative effects of NPY. Moreover, data from real-time quantitative PCR indicated that injection of NPY significantly increased the gene expression of Gnrh1, Kiss1 and Tac3 and decrease the Pdyn while treatment with NPYRA controlled the modulative effects of NPY on these gene expression. CONCLUSIONS: In conclusion based on the results of this study, NPY can exert its impacts on the sexual behavior of male rats via modulation of the KNDy secreting neurons as an interneural pathway to GnRH neurons.


Asunto(s)
Neuropéptido Y/administración & dosificación , Neuropéptido Y/fisiología , Conducta Sexual Animal/efectos de los fármacos , Conducta Sexual Animal/fisiología , Transducción de Señal/efectos de los fármacos , Animales , Dinorfinas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Kisspeptinas , Masculino , Neuroquinina B/metabolismo , Ratas Wistar
5.
J Neurosci Res ; 97(3): 362-372, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30367522

RESUMEN

The full coding sequence of neuropeptide Y (NPY), prepro-NPY, is sequentially metabolized into three peptides; an N-terminus 28-amino acid signaling peptide, the NPY peptide itself (NPY1-36), and a 30-amino acid C-terminus peptide, known as the C-terminal flanking peptide of neuropeptide-Y (CPON). While the signaling peptide directs intracellular trafficking and NPY1-36 is well characterized, the biological function of CPON is unknown. This is noteworthy because CPON is co-stored and co-released along with NPY1-36 and could thus potentially serve important functions. To assess the role of CPON, we adapted a viral genetic approach using two different vector designs encoding NPY, but where the CPON coding sequence was excluded from one of the vectors. Thus, the effect of CPON was indirectly assessed. Male rats received intrahippocampal injections of either a vector encoding NPY1-39 whose metabolism yields NPY1-36 and not CPON, or a prepro-NPY vector encoding both NPY1-36 and CPON. A third vector encoding EGFP served as control. We subsequently studied to what extent CPON might affect seizure susceptibility and memory performance, respectively, to address two important questions to evaluate the potential of NPY gene therapy in epilepsy. Both NPY vectors, as compared to EGFP control, were found to be equally effective at suppressing acute kainate-induced seizures, and both did not influence learning and memory performance in the Morris water maze. Thus CPON itself does not appear to aid actions governed by vector-mediated overexpression of NPY1-36 within the hippocampus. Whether CPON serves other important functions remains to be determined.


Asunto(s)
Neuropéptido Y/metabolismo , Neuropéptido Y/farmacología , Neuropéptido Y/fisiología , Fragmentos de Péptidos/fisiología , Precursores de Proteínas/metabolismo , Precursores de Proteínas/farmacología , Animales , Hipocampo/metabolismo , Masculino , Ratas , Ratas Wistar , Convulsiones/tratamiento farmacológico , Convulsiones/fisiopatología , Aprendizaje Espacial/fisiología , Memoria Espacial/fisiología
6.
Herz ; 44(8): 712-716, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30151710

RESUMEN

Geminin, a key regulator of DNA replication licensing in the cell cycle, plays an essential role in determining the fate of cells via suppression of cell proliferation and cellular differentiation. Neuropeptide Y (NPY) intensifies the proliferation of vascular smooth muscle cells (VSMCs) directly by binding with Y1 receptors. In vitro experiments have shown that stimulation of NPY on VSMCs via regulation of geminin is a double-edged sword. Given that the proliferation and the phenotypic transformation of VSMCs increase the risk for progression of atherosclerosis, we focus on the role of geminin interference in determining the fate of VSMCs. Furthermore, we discuss the therapeutic potential of peripheral neurotransmitter interference, thus pointing toward future research directions in the treatment of atherosclerosis.


Asunto(s)
Geminina , Músculo Liso Vascular , Neuropéptido Y , Proliferación Celular , Geminina/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso , Neuropéptido Y/fisiología
7.
J Neurosci ; 37(1): 23-37, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28053027

RESUMEN

Neuropeptide Y (NPY) has robust anxiolytic properties and is reduced in patients with anxiety disorders. However, the mechanisms by which NPY modulates circuit function to reduce anxiety behavior are not known. Anxiolytic effects of NPY are mediated in the CA1 region of hippocampus, and NPY injection into hippocampus alleviates anxiety symptoms in the predator scent stress model of stress-induced anxiety. The mechanisms that regulate NPY release, and its effects on CA1 synaptic function, are not fully understood. Here we show in acute hippocampal slices from mice that endogenous NPY, released in response to optogenetic stimulation or synaptically evoked spiking of NPY+ cells, suppresses both of the feedforward pathways to CA1. Stimulation of temporoammonic synapses with a physiologically derived spike train causes NPY release that reduces short-term facilitation, whereas the release of NPY that modulates Schaffer collateral synapses requires integration of both the Schaffer collateral and temporoammonic pathways. Pathway specificity of NPY release is conferred by three functionally distinct NPY+ cell types, with differences in intrinsic excitability and short-term plasticity of their inputs. Predator scent stress abolishes the release of endogenous NPY onto temporoammonic synapses, a stress-sensitive pathway, thereby causing enhanced short-term facilitation. Our results demonstrate how stress alters CA1 circuit function through the impairment of endogenous NPY release, potentially contributing to heightened anxiety. SIGNIFICANCE STATEMENT: Neuropeptide Y (NPY) has robust anxiolytic properties, and its levels are reduced in patients with post-traumatic stress disorder. The effects of endogenously released NPY during physiologically relevant stimulation, and the impact of stress-induced reductions in NPY on circuit function, are unknown. By demonstrating that NPY release modulates hippocampal synaptic plasticity and is impaired by predator scent stress, our results provide a novel mechanism by which stress-induced anxiety alters circuit function. These studies fill an important gap in knowledge between the molecular and behavioral effects of NPY. This article also advances the understanding of NPY+ cells and the factors that regulate their spiking, which could pave the way for new therapeutic targets to increase endogenous NPY release in patients in a spatially and temporally appropriate manner.


Asunto(s)
Ansiedad/psicología , Hipocampo/metabolismo , Hipocampo/fisiopatología , Neuropéptido Y/fisiología , Estrés Psicológico/psicología , Animales , Ansiedad/fisiopatología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiopatología , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Técnicas In Vitro , Interneuronas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Plasticidad Neuronal/fisiología , Neuropéptido Y/metabolismo , Odorantes , Optogenética , Conducta Predatoria , Estrés Psicológico/fisiopatología , Sinapsis/fisiología
8.
Int J Obes (Lond) ; 42(11): 1925-1938, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29523877

RESUMEN

OBJECTIVE: To determine whether age and neuropeptide Y (NPY) were involved in the skeletal response to extended periods of diet-induced obesity. METHODS: Male wild-type (WT) and NPY null (NPYKO) mice were fed a mild (23% fat) high-fat diet for 10 weeks from 6 or 16 weeks of age. Metabolism and bone density were assessed during feeding. Skeletal changes were assessed by microCT and histomorphometry. RESULTS: High-fat feeding in 6-week-old WT mice led to significantly increased body weight, adiposity and serum leptin levels, accompanied with markedly suppressed cortical bone accrual. NPYKO mice were less susceptible to fat accrual but, importantly, displayed a complete lack of suppression of bone accrual or cortical bone loss. In contrast, when skeletally mature (16 week old) mice underwent 10 weeks of fat feeding, the metabolic response to HFD was similar to younger mice, however bone mass was not affected in either WT or NPYKO. Thus, growing mice are particularly susceptible to the detrimental effects of HFD on bone mass, through suppression of bone accrual involving NPY signalling. CONCLUSION: This study provides new insights into the relationship between the opposing processes of a positive weight/bone relationship and the negative 'metabolic' effect of obesity on bone mass. This negative effect is particularly active in growing skeletons, which have heightened sensitivity to changes in obesity. In addition, NPY is identified as a fundamental driver of this negative 'metabolic' pathway to bone.


Asunto(s)
Remodelación Ósea/fisiología , Hueso Cortical/patología , Neuropéptido Y/deficiencia , Obesidad/patología , Aumento de Peso/fisiología , Animales , Densidad Ósea , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos , Neuropéptido Y/fisiología , Obesidad/metabolismo
9.
Calcif Tissue Int ; 102(2): 163-173, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29098360

RESUMEN

The five-year survival rate for primary bone cancers is ~ 70% while almost all cases of secondary metastatic bone cancer are terminal. Hypoxia, the deficiency of oxygen which occurs as the rate of tumour growth exceeds the supply of vascularisation, is a key promoter of tumour progression. Hypoxia-driven effects in the primary tumour are wide ranging including changes in gene expression, dysregulation of signalling pathways, resistance to chemotherapy, neovascularisation, increased tumour cell proliferation and migration. Paget's seed and soil theory states that for a metastasising tumour cell 'the seed' it requires the correct microenvironment 'soil' to colonise. Why and how metastasising tumour cells colonise the bone is a complex and intriguing problem. However, once present tumour cells are able to disrupt bone homeostasis through increasing osteoclast activity and downregulating osteoblast function. Osteoclast resorption releases growth factors from the bone matrix that subsequently contribute to the proliferation of invasive tumour cells creating the vicious cycle of bone loss and metastatic cancer progression. Recently, we have shown that hypoxia increases expression and release of lysyl oxidase (LOX) from primary mammary tumours, which in turn disrupts bone homeostasis to favour osteolytic degradation to create pre-metastatic niches in the bone microenvironment. We also demonstrated how treatment with bisphosphonates could block this cancer-induced bone remodelling and reduce secondary bone metastases. This review describes the roles of hypoxia in primary tumour progression to metastasis, with a focus on key signalling pathways and treatment options to reduce patient morbidity and increase survival.


Asunto(s)
Neoplasias Óseas/etiología , Neoplasias Óseas/secundario , Neoplasias Óseas/epidemiología , Neoplasias Óseas/terapia , Neoplasias de la Mama/patología , Hipoxia de la Célula , Dipeptidil Peptidasa 4/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Modelos Biológicos , Mieloma Múltiple/patología , Neuropéptido Y/fisiología , Proteína-Lisina 6-Oxidasa/fisiología
10.
Epilepsia ; 59(7): 1444-1454, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29923603

RESUMEN

OBJECTIVES: Neuropeptide Y (NPY) potently suppresses spike-wave discharges (SWDs) in a genetic rat model of absence epilepsy (GAERS), but the underlying neurophysiologic mechanisms are not clear. We therefore sought to determine the in vivo effects of NPY on neuronal firing in the cortico-thalamo-cortical network activity, known to play a critical role in the generation of SWDs in these rats. METHODS: NPY was administered intracerebroventricularly (ICV) or in separate experiments locally on the neurons of caudal thalamic reticular nucleus (NRT) by use of juxtacellular iontophoresis in triple-barrel electrodes in male GAERS aged 12-15 weeks, in vivo under neuroleptic anesthesia. Drug infusions and electroencephalography (EEG) monitoring were performed simultaneously with juxtacellular single neuronal recordings. Effect of NPY on electrically induced SWD induction threshold were also measured. RESULTS: NPY administration ICV led to a decrease in the total length of SWDs in EEG recordings. Both ICV administration and iontophoresis of NPY on NRT neurons led to an increase in interictal neuronal firing of NRT neurons. During ictal periods, ICV NPY administration reduced the number of thalamic action potentials per SWDs, as well as reduced waveform correlations between field potentials within the NRT and the cortical EEG. NPY administration ICV did not significantly alter the firing patterns of relay thalamic neurons interictally and cortical neurons during ictal and interictal periods. In addition, SWD induction threshold in the S2 region of the cortex was significantly increased after NPY administration. SIGNIFICANCE: Our results show alterations in cortico-thalamo-cortical local and network properties following ICV administration of NPY, suggesting mechanisms of SWD suppression in GAERS. Cellular and network alteration of NRT activity, resulting from a direct action of NPY, may be a contributor to this effect.


Asunto(s)
Epilepsia Tipo Ausencia/fisiopatología , Potenciales Evocados/fisiología , Núcleos Talámicos Intralaminares/fisiología , Red Nerviosa/fisiopatología , Neuropéptido Y/fisiología , Núcleos Talámicos/fisiología , Animales , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Electrodos Implantados , Electroencefalografía , Masculino , Neuronas/fisiología , Ratas
11.
Biochim Biophys Acta Gen Subj ; 1862(3): 403-413, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29154902

RESUMEN

BACKGROUND: During the development of obesity the expansion of white adipose tissue (WAT) leads to a dysregulation and an excessive remodeling of extracellular matrix (ECM), leading to fibrosis formation. These ECM changes have high impact on WAT physiology and may change obesity progression. Blocking WAT fibrosis may have beneficial effects on the efficacy of diet regimen or therapeutical approaches in obesity. Since dipeptidyl peptidase IV (DPP-IV) inhibitors prevent fibrosis in tissues, such as heart, liver and kidney, the objective of this study was to assess whether vildagliptin, a DPP-IV inhibitor, prevents fibrosis in WAT in a mouse model of obesity, and to investigate the mechanisms underlying this effect. METHODS: We evaluated the inhibitory effect of vildagliptin on fibrosis markers on WAT of high-fat diet (HFD)-induced obese mice and on 3T3-L1 cell line of mouse adipocytes treated with a fibrosis inducer, transforming growth factor beta 1 (TGFß1). RESULTS: Vildagliptin prevents the increase of fibrosis markers in WAT of HFD-fed mice and reduces blood glucose, serum triglycerides, total cholesterol and leptin levels. In the in vitro study, the inhibition of DPP-IV with vildagliptin, neuropeptide Y (NPY) treatment and NPY Y1 receptor activation prevents ECM deposition and fibrosis markers increase induced by TGFß1 treatment. CONCLUSIONS: Vildagliptin prevents fibrosis formation in adipose tissue in obese mice, at least partially through NPY and NPY Y1 receptor activation. GENERAL SIGNIFICANCE: This study highlights the importance of vildagliptin in the treatment of fibrosis that occur in obesity.


Asunto(s)
Adamantano/análogos & derivados , Tejido Adiposo Blanco/efectos de los fármacos , Dipeptidil Peptidasa 4/efectos de los fármacos , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Hipolipemiantes/uso terapéutico , Nitrilos/uso terapéutico , Obesidad/tratamiento farmacológico , Pirrolidinas/uso terapéutico , Células 3T3-L1 , Adamantano/farmacología , Adamantano/uso terapéutico , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo Blanco/patología , Animales , Glucemia/análisis , Colágeno/metabolismo , Dieta Alta en Grasa , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibrosis , Hipolipemiantes/farmacología , Leptina/sangre , Leptina/fisiología , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Neuropéptido Y/agonistas , Neuropéptido Y/farmacología , Neuropéptido Y/fisiología , Nitrilos/farmacología , Obesidad/patología , Pirrolidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Neuropéptido Y/agonistas , Receptores de Neuropéptido Y/fisiología , Factor de Crecimiento Transformador beta1/farmacología , Vildagliptina
12.
Adv Exp Med Biol ; 1090: 183-198, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30390291

RESUMEN

In organism, energy homeostasis is a biological process that involves the coordinated homeostatic regulation of energy intake (food intake) and energy expenditure. The human brain, particularly the hypothalamic proopiomelanocortin (POMC)- and agouti-related protein/neuropeptide Y (AgRP/NPY)-expressing neurons in the arcuate nucleus, plays an essential role in regulating energy homeostasis. The regulation process is mainly dependent upon peripheral hormones such as leptin and insulin, as well as nutrients such as glucose, amino acids, and fatty acids. Although many studies have attempted to illustrate the exact mechanisms of glucose and hormones action on these neurons, we still cannot clearly see the full picture of this regulation action. Therefore, in this review we will mainly discuss those established theories and recent progresses in this area, demonstrating the possible physiological mechanism by which glucose, leptin, and insulin affect neuronal excitability of POMC and AgRP neurons. In addition, we will also focus on some important ion channels which are expressed by POMC and AgRP neurons, such as KATP channels and TRPC channels, and explain how these channels are regulated by peripheral hormones and nutrients and thus regulate energy homeostasis.


Asunto(s)
Fenómenos Electrofisiológicos , Metabolismo Energético , Neuronas/fisiología , Nutrientes , Proteína Relacionada con Agouti/fisiología , Núcleo Arqueado del Hipotálamo/citología , Glucosa/fisiología , Homeostasis , Humanos , Insulina/fisiología , Leptina/fisiología , Neuropéptido Y/fisiología , Proopiomelanocortina/fisiología
13.
Proc Natl Acad Sci U S A ; 112(13): E1642-51, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25775546

RESUMEN

Aging is characterized by autophagy impairment that contributes to age-related disease aggravation. Moreover, it was described that the hypothalamus is a critical brain area for whole-body aging development and has impact on lifespan. Neuropeptide Y (NPY) is one of the major neuropeptides present in the hypothalamus, and it has been shown that, in aged animals, the hypothalamic NPY levels decrease. Because caloric restriction (CR) delays aging, at least in part, by stimulating autophagy, and also increases hypothalamic NPY levels, we hypothesized that NPY could have a relevant role on autophagy modulation in the hypothalamus. Therefore, the aim of this study was to investigate the role of NPY on autophagy in the hypothalamus. Using both hypothalamic neuronal in vitro models and mice overexpressing NPY in the hypothalamus, we observed that NPY stimulates autophagy in the hypothalamus. Mechanistically, in rodent hypothalamic neurons, NPY increases autophagy through the activation of NPY Y1 and Y5 receptors, and this effect is tightly associated with the concerted activation of PI3K, MEK/ERK, and PKA signaling pathways. Modulation of hypothalamic NPY levels may be considered a potential strategy to produce protective effects against hypothalamic impairments associated with age and to delay aging.


Asunto(s)
Autofagia , Hipotálamo/citología , Neuronas/citología , Neuropéptido Y/fisiología , Envejecimiento , Animales , Encéfalo/metabolismo , Restricción Calórica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Transducción de Señal
14.
Toxicol Pathol ; 45(7): 894-903, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29113558

RESUMEN

Although the brain is well established as a master regulator of homeostasis in peripheral tissues, central regulation of bone mass represents a novel and rapidly expanding field of study. This review examines the current understanding of central regulation of the skeleton, exploring several of the key pathways connecting brain to bone and their implications both in mice and the clinical setting. Our understanding of central bone regulation has largely progressed through examination of skeletal responses downstream of nutrient regulatory pathways in the hypothalamus. Mutations and modulation of these pathways, in cases such as leptin deficiency, induce marked bone phenotypes, which have provided vital insights into central bone regulation. These studies have identified several central neuropeptide pathways that stimulate well-defined changes in bone cell activity in response to changes in energy homeostasis. In addition, this work has highlighted the endocrine nature of the skeleton, revealing a complex cross talk that directly regulates other organ systems. Our laboratory has studied bone-active neuropeptide pathways and defined osteoblast-based actions that recapitulate central pathways linking bone, fat, and glucose homeostasis. Studies of neural control of bone have produced paradigm-shifting changes in our understanding of the skeleton and its relationship with the wider array of organ systems.


Asunto(s)
Remodelación Ósea , Neuronas/fisiología , Animales , Huesos/fisiología , Homeostasis , Humanos , Hipotálamo/fisiología , Leptina/fisiología , Músculo Esquelético/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuropéptido Y/fisiología , Polipéptido Pancreático/fisiología , Péptido YY/fisiología , Proopiomelanocortina/fisiología , Receptores de Cannabinoides/fisiología , Semaforinas/fisiología , Sistema Nervioso Simpático/fisiología
15.
Artículo en Inglés | MEDLINE | ID: mdl-28557151

RESUMEN

The Asian Corn Borer Ostrinia furnacalis is a major agricultural pest. In this study, a full-length neuropeptide Y (npy) gene in O. furnacalis was sequenced and cloned from cDNA library, which contains an ORF of 273 bp by encoding 90 amino acid residues. The mature OfurNPY is composed of 29 amino acids with amidation in C-terminal. The spatiotemporal expression analysis showed that npy highest expression level was in the midgut of the fifth instar larvae (the gluttony period). When the expression of npy was knocked down by feeding or injecting dsNPY, larval food consumption, body size, and body weight were significantly inhibited compared to controls. These results indicate that NPY is an important regulator in the control of feeding of O. furnacalis.


Asunto(s)
Mariposas Nocturnas/fisiología , Neuropéptido Y/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Conducta Alimentaria , Silenciador del Gen
16.
Osteoporos Int ; 27(9): 2777-2789, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27080706

RESUMEN

UNLABELLED: Increased neuropeptide Y (NPY) expression occurred in the glucocorticoid-induced osteoporotic skeleton. NPY knockout mice exhibited a minor response to the glucocorticoid-mediated exacerbation of bone accretion and fatty marrow pathogenesis. NPY deletion restored SITR1 signaling and enhanced PPARγ ubiquitination of bone tissue, an alternative strategy for ameliorating glucocorticoid-induced skeletal deterioration. INTRODUCTION: Glucocorticoid excess is observed to worsen the pathogenesis of osteoporosis and fatty marrow. This study was undertaken to investigate the contribution of neuropeptide Y (NPY) to glucocorticoid-induced bone loss and marrow adiposity. METHODS: NPY knockout and wild-type mice were administered methylprednisolone for four consecutive weeks. Bone mineral density, microarchitecture, and calcein-labeled mineral acquisition were quantified by µCT, dual energy X-ray absorptiometry, and histomorphometry. Expression of osteogenic and adipogenic markers and acetylation states of PPARγ were detected by RT-quantitative PCR, immunoprecipitation, and immunoblotting. RESULTS: High NPY levels were associated with glucocorticoid-induced trabecular bone deterioration and marrow fat accumulation. Mice lacking NPY had high bone mass concomitant with spacious trabecular and cortical bone microstructure. NPY deletion shielded skeletal tissues from the glucocorticoid-induced impediment of bone mass, trabecular morphometric characteristics, mineral accretion activity, and fatty marrow development. Ex vivo, NPY deficiency sustained osteogenic differentiation capacity and curtailed the glucocorticoid-mediated escalation of adipocyte formation reactions of primary bone-marrow mesenchymal cells. NPY deletion appeared to modulate Y1 and Y2 receptors, sirtuin 1, ERK, and p38 signaling pathways, an effect that facilitated hypoacetylation and ubiquitination of adipogenic transcription factor PPARγ in the skeletal tissues exposed to glucocorticoid stress. CONCLUSIONS: NPY mediates the glucocorticoid-induced disturbance of mineral accretion and marrow adipogenesis through post-translational modification of PPARγ. This study brings a new molecular insight into the disintegration of adipogenic and osteogenic activities within glucocorticoid-mediated osteoporotic skeletons. Control of NPY is an alternative strategy to ameliorate glucocorticoid-induced bone destruction and fatty marrow.


Asunto(s)
Adiposidad , Médula Ósea/patología , Neuropéptido Y/fisiología , Osteogénesis , Osteoporosis/fisiopatología , Animales , Glucocorticoides/efectos adversos , Masculino , Ratones , Ratones Noqueados , Neuropéptido Y/genética , Osteoporosis/inducido químicamente
17.
Artículo en Inglés | MEDLINE | ID: mdl-27387442

RESUMEN

Maintaining adaptive control of behavior and physiology is the main strategy used by animals in responding to changes of food resources. To investigate the effects of random food deprivation (FD) and refeeding on energy metabolism and behavior in Apodemus chevrieri, we acclimated adult males to FD for 4weeks, then refed them ad libitum for 4weeks (FD-Re group). During the period of FD, animals were fed ad libitum for 4 randomly assigned days each week, and deprived of food the other 3days. A control group was fed ad libitum for 8weeks. At 4 and 8weeks we measured body mass, thermogenesis, serum leptin levels, body composition, gastrointestinal tract morphology, behavior and hypothalamic neuropeptide expression. At 4weeks, food intake, gastrointestinal mass, neuropeptide Y (NPY) and agouti-related protein (AgRP) mRNA expressions increased and thermogenesis, leptin levels, pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) expressions decreased in FD compared with controls. FD also showed more resting behavior and less activity than the controls on ad libitum day. There were no differences between FD-Re and controls at 8weeks, indicating significant plasticity. These results suggested that animals can compensate for unpredictable reduction in food availability by increasing food intake and reducing energy expended through thermogenesis and activity. Leptin levels, NPY, AgRP, POMC, and CART mRNA levels may also regulate energy metabolism. Significant plasticity in energy metabolism and behavior was shown by A. chevrieri over a short timescale, allowing them to adapt to food shortages in nutritionally unpredictable environments.


Asunto(s)
Privación de Alimentos/fisiología , Hipotálamo/fisiología , Murinae/fisiología , Neuropéptidos/fisiología , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/fisiología , Animales , Metabolismo Basal/genética , Metabolismo Basal/fisiología , Conducta Animal/fisiología , Composición Corporal , Peso Corporal , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Metabolismo Energético , Leptina/sangre , Masculino , Murinae/genética , Murinae/psicología , Proteínas del Tejido Nervioso/genética , Neuropéptido Y/genética , Neuropéptido Y/fisiología , Neuropéptidos/genética , Proopiomelanocortina/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Termogénesis/genética , Termogénesis/fisiología
18.
J Neurosci ; 34(49): 16309-19, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25471570

RESUMEN

The hypothalamic NPY system plays an important role in regulating food intake and energy expenditure. Different biological actions of NPY are assigned to NPY receptor subtypes. Recent studies demonstrated a close relationship between food intake and growth hormone (GH) secretion; however, the mechanism through which endogenous NPY modulates GH release remains unknown. Moreover, conclusive evidence demonstrating a role for NPY and Y-receptors in regulating the endogenous pulsatile release of GH does not exist. We used genetically modified mice (germline Npy, Y1, and Y2 receptor knock-out mice) to assess pulsatile GH secretion under both fed and fasting conditions. Deletion of NPY did not impact fed GH release; however, it reversed the fasting-induced suppression of pulsatile GH secretion. The recovery of GH secretion was associated with a reduction in hypothalamic somatotropin release inhibiting factor (Srif; somatostatin) mRNA expression. Moreover, observations revealed a differential role for Y1 and Y2 receptors, wherein the postsynaptic Y1 receptor suppresses GH secretion in fasting. In contrast, the presynaptic Y2 receptor maintains normal GH output under long-term ad libitum-fed conditions. These data demonstrate an integrated neural circuit that modulates GH release relative to food intake, and provide essential information to address the differential roles of Y1 and Y2 receptors in regulating the release of GH under fed and fasting states.


Asunto(s)
Ayuno/fisiología , Hormona del Crecimiento/metabolismo , Neuropéptido Y/fisiología , Receptores de Neuropéptido Y/fisiología , Animales , Glucemia , Hormona Liberadora de Hormona del Crecimiento/biosíntesis , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuropéptido Y/sangre , Neuropéptido Y/genética , Péptido YY/sangre , Receptores de Neuropéptido Y/genética , Somatostatina/biosíntesis
19.
J Neurosci ; 34(27): 9096-106, 2014 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-24990930

RESUMEN

Weight loss is a prominent early feature of Alzheimer's disease (AD) that often precedes the cognitive decline and clinical diagnosis. While the exact pathogenesis of AD remains unclear, accumulation of amyloid-ß (Aß) derived from the amyloid precursor protein (APP) in the brain is thought to lead to the neuronal dysfunction and death underlying the dementia. In this study, we examined whether transgenic mice overexpressing the Swedish mutation of APP (Tg2576), recapitulating selected features of AD, have hypothalamic leptin signaling dysfunction leading to early body weight deficits. We found that 3-month-old Tg2576 mice, before amyloid plaque formation, exhibit decreased weight with markedly decreased adiposity, low plasma leptin levels, and increased energy expenditure without alterations in feeding behavior. The expression of the orexigenic neuropeptide Y (NPY) in the hypothalamus to the low leptin state was abnormal at basal and fasting conditions. In addition, arcuate NPY neurons exhibited abnormal electrophysiological responses to leptin in Tg2576 hypothalamic slices or wild-type slices treated with Aß. Finally, the metabolic deficits worsened as Tg2576 mice aged and amyloid burden increased in the brain. These results indicate that excess Aß can potentially disrupt hypothalamic arcuate NPY neurons leading to weight loss and a pathologically low leptin state early in the disease process that progressively worsens as the amyloid burden increases. Collectively, these findings suggest that weight loss is an intrinsic pathological feature of Aß accumulation and identify hypothalamic leptin signaling as a previously unrecognized pathogenic site of action for Aß.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/fisiología , Núcleo Arqueado del Hipotálamo/fisiopatología , Hipotálamo/fisiopatología , Leptina/deficiencia , Neuropéptido Y/fisiología , Pérdida de Peso/fisiología , Adiposidad , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/análisis , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/patología , Química Encefálica , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ayuno , Conducta Alimentaria , Femenino , Genes Reporteros , Humanos , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Neuropéptido Y/genética , Técnicas de Placa-Clamp , Placa Amiloide
20.
FASEB J ; 28(12): 5337-48, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25205743

RESUMEN

An orexigenic hormone, neuropeptide Y (NPY), plays a role not only in the hypothalamic regulation of appetite, but also in the peripheral regulation of lipid metabolism. However, the intracellular mechanisms triggered by NPY to regulate lipid metabolism are poorly understood. Here we report that NPY deficiency reduces white adipose tissue (WAT) mass and ameliorates the age-related imbalance of adipose tissue metabolism in mice. Gene expression involved in adipogenesis/lipogenesis was found to decrease, whereas proteins involved in lipolysis increased in gonadal WAT (gWAT) of NPY-knockout mice. These changes were associated with an activated SIRT1- and PPARγ-mediated pathway. Moreover, the age-related decrease of de novo lipogenesis in gWAT and thermogenesis in inguinal WAT was inhibited by NPY deficiency. Further analysis using 3T3-L1 cells showed that NPY inhibited lipolysis through the Y1 receptor and enhanced lipogenesis following a reduction in cAMP response element-binding protein (CREB) and SIRT1 protein expression. Therefore, NPY appears to act as a key regulator of adipose tissue metabolism via the CREB-SIRT1 signaling pathway. Taken together, NPY deficiency reduces adiposity and ameliorates the age-related imbalance of adipose tissue metabolism, suggesting that antagonism of NPY may be a promising target for drug development to prevent age-related metabolic diseases.


Asunto(s)
Tejido Adiposo/metabolismo , Adiposidad/fisiología , Factores de Edad , Neuropéptido Y/antagonistas & inhibidores , Células 3T3-L1/metabolismo , Animales , Secuencia de Bases , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Cartilla de ADN , Masculino , Ratones , Ratones Noqueados , Neuropéptido Y/genética , Neuropéptido Y/fisiología , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA