Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Neuroendocrinology ; 111(12): 1187-1200, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33291119

RESUMEN

INTRODUCTION: Repeated traumatic events result in long-lasting neuropsychiatric ailments, including neuroendocrine imbalances. Neuropeptide Y (NPY) in the arcuate nucleus (Arc) is an important orexigenic peptide. However, the molecular underpinnings of its dysregulation owing to traumatic brain injury remain unknown. METHODS: Rats were subjected to repeated mild traumatic brain injury (rMTBI) using the closed head weight-drop model. Feeding behavior and the regulatory epigenetic parameters of NPY expression were measured at 48 h and 30 days post-rMTBI. Further, sodium butyrate (SB), a pan-histone deacetylase (HDAC) inhibitor, was administered to examine whether histone deacetylation is involved in NPY expression post-rMTBI. RESULTS: The rMTBI attenuated food intake, which was coincident with a decrease in NPY mRNA and protein levels in the Arc post-rMTBI. Further, rMTBI also reduced the mRNA levels of the cAMP response element-binding protein (CREB) and CREB-binding protein (CBP) and altered the mRNA levels of the various isoforms of the HDACs. Concurrently, the acetylated histone 3-lysine 9 (H3-K9) levels and the binding of CBP at the NPY promoter in the Arc of the rMTBI-exposed rats were reduced. However, the treatment with SB corrected the rMTBI-induced deficits in the H3-K9 acetylation levels and CBP occupancy at the NPY promoter, restoring both NPY expression and food intake. CONCLUSIONS: These findings suggest that histone deacetylation at the NPY promoter persistently controls NPY function in the Arc after rMTBI. This study also demonstrates the efficacy of HDAC inhibitors in mitigating trauma-induced neuroendocrine maladaptations in the hypothalamus.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Conducta Animal/fisiología , Conmoción Encefálica/metabolismo , Ácido Butírico/farmacología , Conducta Alimentaria/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Neuropéptido Y/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Conmoción Encefálica/tratamiento farmacológico , Modelos Animales de Enfermedad , Conducta Alimentaria/efectos de los fármacos , Neuropéptido Y/efectos de los fármacos , Ratas
2.
Am J Physiol Regul Integr Comp Physiol ; 318(3): R634-R648, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31967846

RESUMEN

In males, obesity increases sympathetic nerve activity (SNA), but the mechanisms are unclear. Here, we investigate insulin, via an action in the arcuate nucleus (ArcN), and downstream neuropathways, including melanocortin receptor 3/4 (MC3/4R) in the hypothalamic paraventricular nucleus (PVN) and dorsal medial hypothalamus (DMH). We studied conscious and α-chloralose-anesthetized Sprague-Dawley rats fed a high-fat diet, which causes obesity prone (OP) rats to accrue excess fat and obesity-resistant (OR) rats to maintain fat content, similar to rats fed a standard control (CON) diet. Nonspecific blockade of the ArcN with muscimol and specific blockade of ArcN insulin receptors (InsR) decreased lumbar SNA (LSNA), heart rate (HR), and mean arterial pressure (MAP) in OP, but not OR or CON, rats, indicating that insulin supports LSNA in obese males. In conscious rats, intracerebroventricular infusion of insulin increased MAP only in OP rats and also improved HR baroreflex function from subnormal to supranormal. The brain sensitization to insulin may elucidate how insulin can drive central SNA pathways when transport of insulin across the blood-brain barrier may be impaired. Blockade of PVN, but not DMH, MC3/4R with SHU9119 decreased LSNA, HR, and, MAP in OP, but not OR or CON, rats. Interestingly, nanoinjection of the MC3/4R agonist melanotan II (MTII) into the PVN increased LSNA only in OP rats, similar to PVN MTII-induced increases in LSNA in CON rats after blockade of sympathoinhibitory neuropeptide Y Y1 receptors. ArcN InsR expression was not increased in OP rats. Collectively, these data indicate that obesity increases SNA, in part via increased InsR signaling and downstream PVN MC3/4R.


Asunto(s)
Encéfalo/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Receptores de Neuropéptido Y/metabolismo , Animales , Encéfalo/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Neuropéptido Y/efectos de los fármacos , Neuropéptido Y/metabolismo , Obesidad/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptor de Melanocortina Tipo 4/metabolismo , Sistema Nervioso Simpático/fisiopatología
3.
Curr Osteoporos Rep ; 18(3): 325-335, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32249381

RESUMEN

PURPOSE OF REVIEW: Opioids have been shown to be associated with an increased risk of fracture. The purpose of this paper is to review recent research into the effects of opioids on bone formation and bone healing in animal models and in human studies. RECENT FINDINGS: Most opioids, such as morphine and fentanyl, negatively affected bone remodeling and bone healing in animal models. Conversely, remifentanil has been recently shown to promote in vitro osteoblast differentiation and to inhibit differentiation and maturation of osteoclasts, therefore reducing bone resorption. According to the possible negative role of opioids in bone healing, opioid antagonists have been shown to enhance bone mineralization, suggesting a possible therapeutic role in the future for osteoporosis. Other neuropeptides, such as the vasoactive intestinal peptide (VIP) and the neuropeptide Y (NPY), have been proved to promote osteogenesis. The increased risk of fractures among opioid users may be related to their central nervous system side effects or to the reduced bone density, partly due to their endocrine effects, and partly to their direct activity on bone cells. Clinical data strongly suggested a potential negative effect of opioids in bone healing. The risk of nonunion fracture is significantly increased in opioid users, and bone mass density was reduced in patients under long-term opioid treatment. The direct effects of opioids on bone remodeling appears evident from these reports. Not all opioids have the same potential for negatively impacting bone healing. Opioid antagonists may increase bone density and could represent a possible future treatment for low bone mass density pathologies. However, further trials are warranted to clarify the clinical relevance of these emerging findings from animal studies.


Asunto(s)
Analgésicos Opioides/farmacología , Remodelación Ósea/efectos de los fármacos , Curación de Fractura/efectos de los fármacos , Fracturas Óseas/epidemiología , Osteogénesis/efectos de los fármacos , Analgésicos Opioides/uso terapéutico , Animales , Calcificación Fisiológica/efectos de los fármacos , Fracturas no Consolidadas/epidemiología , Humanos , Antagonistas de Narcóticos/farmacología , Neuropéptido Y/efectos de los fármacos , Neuropéptido Y/metabolismo , Péptido Intestinal Vasoactivo/efectos de los fármacos , Péptido Intestinal Vasoactivo/metabolismo
4.
Neurol Sci ; 41(2): 295-303, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31494821

RESUMEN

BACKGROUND: Guillain-Barré syndrome (GBS) is a common acute immune-mediated inflammatory disorder affecting the peripheral nervous system (PNS) of humans. Studies in humans and in animal models revealed that neuropeptide Y (NPY) levels are altered in some neurodegenerative and neuroimmune disorders. Herein, we investigated the levels of NPY and cytokines in the serum of GBS patients and explored the roles of NPY in the disease severity and its short-term prognosis. METHODS: Twenty patients with GBS (case group) and twenty healthy individuals (control group) were enrolled in this study. NPY levels were analyzed by enzyme-linked immunosorbent assay (ELISA). The levels of pro- and anti-inflammatory cytokines (including interferon-γ (IFN-γ), interleukin (IL)-4, IL-10, IL-12p70, IL-17A, and tumor necrosis factor-α (TNF-α)) were analyzed using cytometric beads array (CBA). The clinical characteristics, disease severity, and short-term prognosis were compared between the two groups. RESULTS: Compared with the control group, the levels of NPY and cytokines were significantly increased in the serum of patients with GBS. NPY levels in the serum of GBS patients were correlated with the disease severity. CONCLUSION: Our results suggest that NPY and cytokines are involved in the pathogenesis of GBS. The levels of NPY can help to predict the severity of the disease.


Asunto(s)
Citocinas/sangre , Citocinas/efectos de los fármacos , Síndrome de Guillain-Barré/sangre , Síndrome de Guillain-Barré/tratamiento farmacológico , Inmunoglobulinas Intravenosas/farmacología , Neuropéptido Y/sangre , Neuropéptido Y/efectos de los fármacos , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Síndrome de Guillain-Barré/fisiopatología , Humanos , Inmunoglobulinas Intravenosas/administración & dosificación , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
5.
Int J Neurosci ; 130(2): 193-203, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31518546

RESUMEN

Purpose: Neuropeptides and neurotrophic factors are thought to be involved in epileptogenesis. This study aims to investigate the potential effects of anticonvulsant drugs on neuropeptides (galanin and neuropeptide Y) and neurotrophic factors (BDNF and NGF) in pentylenetetrazol (PTZ)-kindled seizures in the rat.Methods: Forty-eight adult male Sprague-Dawley rats were included in the study. The animals were divided into 8 groups of six rats. Group 1 was defined as naïve control, and received no medication. Group 2 (PTZ + saline) was treated with sub-convulsive doses of PTZ (35 mg/kg) and saline i.p. for 14 days. For anticonvulsant treatments, Groups 3-8 were treated with 200 mg/kg levetiracetam (PTZ + LEV), 1 mg/kg midazolam (PTZ + MDZ), 80 mg/kg phenytoin (PTZ + PHT), 80 mg/kg topiramate (PTZ + TPR), 40 mg/kg lamotrigine (PTZ + LMT) and 50 mg/kg sodium valproate (PTZ + SV), respectively. All anticonvulsant drugs were injected 30 min prior to PTZ injection throughout 14 days. Following treatment period, behavioral, biochemical and immunohistochemical studies were performed.Results: PTZ + saline group revealed significantly decreased galanin, NPY, BDNF and NGF levels compared to control. PTZ + MDZ group had significantly increased galanin, BDNF and NGF levels compared to saline group. Also, PTZ + LEV group showed increased BDNF levels. PTZ + saline group revealed significantly lower neuron count and higher GFAP (+) cells in hippocampal CA1-CA3 regions. All anticonvulsants significantly reduced hippocampal astrogliosis whereas only midazolam, levetiracetam, sodium valproate and lamotrigine prevented neuronal loss.Conclusion: Our results suggested that anticonvulsant drugs may reduce the severity of seizures, and exert neuroprotective effects by altering the expression of neuropeptides and neurotrophins in the epileptogenic hippocampus.


Asunto(s)
Anticonvulsivantes/farmacología , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Epilepsia/tratamiento farmacológico , Guanosina Monofosfato , Hipocampo/efectos de los fármacos , Inosina Monofosfato , Factor de Crecimiento Nervioso/efectos de los fármacos , Neuropéptido Y/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Convulsiones/tratamiento farmacológico , Animales , Anticonvulsivantes/administración & dosificación , Convulsivantes/farmacología , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Masculino , Fármacos Neuroprotectores/administración & dosificación , Pentilenotetrazol/farmacología , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente
6.
Int J Neuropsychopharmacol ; 22(4): 292-302, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30590608

RESUMEN

BACKGROUND: Tolerance to ethanol-induced anxiolysis promotes alcohol intake, thus contributing to alcohol use disorder development. Recent studies implicate histone deacetylase-mediated histone H3K9 deacetylation in regulating neuropeptide Y expression during rapid ethanol tolerance to the anxiolytic effects of ethanol. Furthermore, the histone methyltransferase, G9a, and G9a-mediated H3K9 dimethylation (H3K9me2) have recently emerged as regulators of addiction and anxiety; however, their role in rapid ethanol tolerance is unknown. Therefore, we investigated the role of G9a-mediated H3K9me2 in neuropeptide Y expression during rapid ethanol tolerance. METHODS: Adult male rats were administered one injection of n-saline followed by single acute ethanol injection (1 g/kg) 24 hours later (ethanol group) or 2 injections (24 hours apart) of either n-saline (saline group) or ethanol (tolerance group). Anxiety-like behaviors and global and Npy-specific G9a and H3K9me2 levels in the amygdala were measured. Effects of G9a inhibitor (UNC0642) treatment on behavioral and epigenetic measures were also examined. RESULTS: Acute ethanol produced anxiolysis and decreased global H3K9me2 and G9a protein levels in the central and medial nucleus of the amygdala as well as decreased occupancy levels of H3K9me2 and G9a near a putative binding site for cAMP-response element binding protein on the Npy gene. Two identical doses of ethanol produced no behavioral or epigenetic changes relative to controls, indicating development of rapid ethanol tolerance. Interestingly, treatment with UNC0642, before the second ethanol dose reversed rapid ethanol tolerance, decreased global H3K9me2 and increased neuropeptide Y levels in the central and medial nucleus of the amygdala. CONCLUSIONS: These results implicate amygdaloid G9a-mediated H3K9me2 mechanisms in regulating rapid tolerance to the anxiolytic effects of ethanol via neuropeptide Y expression regulation.


Asunto(s)
Amígdala del Cerebelo , Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Tolerancia a Medicamentos , Etanol/farmacología , N-Metiltransferasa de Histona-Lisina , Neuropéptido Y , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Ansiolíticos/administración & dosificación , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Etanol/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/metabolismo , Masculino , Neuropéptido Y/efectos de los fármacos , Neuropéptido Y/metabolismo , Quinazolinas/farmacología , Ratas , Ratas Sprague-Dawley
7.
Addict Biol ; 21(4): 755-65, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-25904345

RESUMEN

Neuropeptide Y (NPY), which is widely expressed in the central nervous system is involved in several neuropathologies including addiction. Here we comprehensively and systematically review alterations on the central NPY system induced by several drugs. We report on the effects of psychostimulants [cocaine, amphetamine, methamphetamine, 3,4-methylenedioxymethamphetamine (MDMA) and nicotine], ethanol, and opioids on NPY protein levels and expression of different NPY receptors. Overall, expression and function of NPY and its receptors are changed under conditions of drug exposure, thus affecting several physiologic behaviors, such as feeding, stress and anxiety. Drugs of abuse differentially affect the components of the NPY system. For example methamphetamine and nicotine lead to a consistent increase in NPY mRNA and protein levels in different brain sites whereas ethanol and opioids decrease NPY mRNA and protein expression. Drug-induced alterations on the different NPY receptors show more complex regulation pattern. Manipulation of the NPY system can have opposing effects on reinforcing and addictive properties of drugs of abuse. NPY can produce pro-addictive effects (nicotine and heroin), but can also exert inhibitory effects on addictive behavior (AMPH, ethanol). Furthermore, NPY can act as a neuroprotective agent in chronically methamphetamine and MDMA-treated rodents. In conclusion, manipulation of the NPY system seems to be a potential target to counteract neural alterations, addiction-related behaviors and cognitive deficits induced by these drugs.


Asunto(s)
Anfetaminas/farmacología , Cocaína/farmacología , Etanol/farmacología , Neuropéptido Y/efectos de los fármacos , Nicotina/farmacología , Alcaloides Opiáceos/farmacología , Animales , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones , Ratas , Receptores de Neuropéptido Y/efectos de los fármacos
8.
Genet Mol Res ; 15(1): 15017362, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26985919

RESUMEN

The hypothalamus is an important component of the nervous system, and neuropeptide Y (NPY), proopiomelanocortin (POMC), and neuromedin U (NMU) are key players in physiological regulation. Puerarin is important for nerve regulation. We investigated the effect of puerarin on the expression of NMU, NPY, and POMC genes in the hypothalamus. The results showed that the puerarin low-dose group and the other groups were significantly different (P < 0.05). However, there was no significant difference in NMU, POMC, and NPY among the groups.


Asunto(s)
Hipotálamo/metabolismo , Isoflavonas/farmacología , Neuropéptido Y/genética , Neuropéptidos/genética , Proopiomelanocortina/genética , Animales , Regulación de la Expresión Génica , Hipotálamo/efectos de los fármacos , Neuropéptido Y/efectos de los fármacos , Neuropéptidos/efectos de los fármacos , Proopiomelanocortina/efectos de los fármacos , Ratas
9.
BMC Complement Altern Med ; 15: 340, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26419631

RESUMEN

BACKGROUND: To examine whether electro-acupuncture (EA) could decrease 5-hydroxytryptamine (5-HT) and calcitonin gene-related peptide (CGRP), and increase neuro-peptide Y (NPY) in the brain-gut axis (BGA) in D-IBS using rat models. METHODS: Rats were randomly exposed to unpredictable chronic stress for 3 weeks followed by 1-hour acute restraint stress (CAS) after 7 days of rest, or daily gavage of Senna decoction (6 g/kg) plus chronic restraint stress (for a duration of 2 h, starting from 1 h prior to the gavage) for 2 weeks (ISC). The content of 5-HT, CGRP and NPY in the distal colon, spinal cord, hypothalamus was examined at the end of the treatment. RESULTS: 1. The two rat models exhibited similar characteristics, e.g., increased number of fecal pellets expelled in 1 h, decreased sacchar-intake, decreased CRD, elevated 5-HT, CGRP content and decreased NPY in the distal colon, spinal cord, hypothalamus (P < 0.05 vs. that in healthy control rats). 2. A series of equations was developed based on correlation regression analysis. The analysis results demonstrated that 5-HT mediates the changes in hypothalamus, spinal cord and colon. 5-HT and CGRP in spinal cord was closely correlated with general behavior evaluation and other transmitters in BGA. CONCLUSION: 1. In comparison to 5-HT, CGRP and NPY (particularly in the spinal cord) had closer relationship with the D-IBS symptoms induced by either stress factors or Senna decotion. 2. EA treatment could restore the brain-gut axis to balanced levels.


Asunto(s)
Terapia por Acupuntura , Síndrome del Colon Irritable/enzimología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Péptido Relacionado con Gen de Calcitonina/efectos de los fármacos , Péptido Relacionado con Gen de Calcitonina/metabolismo , Colon/metabolismo , Diarrea , Modelos Animales de Enfermedad , Hipotálamo , Masculino , Neuropéptido Y/efectos de los fármacos , Neuropéptido Y/metabolismo , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Médula Espinal/metabolismo
10.
FASEB J ; 24(8): 2670-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20335227

RESUMEN

The orexigenic effect of ghrelin is mediated by neuropeptide Y (NPY) and agouti-related protein (AgRP) in the hypothalamic arcuate nucleus (ARC). Recent evidence also indicates that ghrelin promotes feeding through a mechanism involving activation of hypothalamic AMP-activated protein kinase (AMPK) and inactivation of acetyl-CoA carboxylase and fatty acid synthase (FAS). This results in decreased hypothalamic levels of malonyl-CoA, increased carnitine palmitoyltransferase 1 (CPT1) activity, and mitochondrial production of reactive oxygen species. We evaluated whether these molecular events are part of a unique signaling cascade or whether they represent alternative pathways mediating the orexigenic effect of ghrelin. Moreover, we examined the gender dependency of these mechanisms, because recent evidence has proposed that ghrelin orexigenic effect is reduced in female rats. We studied in both genders the effect of ghrelin on the expression of AgRP and NPY, as well as their transcription factors: cAMP response-element binding protein (CREB and its phosphorylated form, pCREB), forkhead box O1 (FoxO1 and its phosphorylated form, pFoxO1), and brain-specific homeobox transcription factor (BSX). In addition, to establish a mechanistic link between ghrelin, fatty acid metabolism, and neuropeptides, we evaluated the effect of ghrelin after blockage of hypothalamic fatty acid beta oxidation, by using the CPT1 inhibitor etomoxir. Ghrelin-induced changes in the AMPK-CPT1 pathway are associated with increased levels of AgRP and NPY mRNA expression through modulation of BSX, pCREB, and FoxO1, as well as decreased expression of endoplasmic reticulum (ER) stress markers in a gender-independent manner. In addition, blockage of hypothalamic fatty acid beta oxidation prevents the ghrelin-promoting action on AgRP and NPY mRNA expression, also in a gender-independent manner. Notably, this effect is associated with decreased BSX expression and reduced food intake. Overall, our data suggest that BSX integrates changes in neuronal metabolic status with ARC-derived neuropeptides in a gender-independent manner.


Asunto(s)
Ácidos Grasos/metabolismo , Ghrelina/farmacología , Proteínas de Homeodominio/genética , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/efectos de los fármacos , Factores de Transcripción/metabolismo , Proteína Relacionada con Agouti/efectos de los fármacos , Proteína Relacionada con Agouti/genética , Animales , Ingestión de Alimentos , Femenino , Regulación de la Expresión Génica , Masculino , Neuropéptido Y/efectos de los fármacos , Neuropéptido Y/genética , Neuropéptidos/genética , Ratas , Factores Sexuales , Factores de Transcripción/genética
11.
Diabetes Obes Metab ; 13(5): 408-17, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21226821

RESUMEN

AIMS: Peptide YY (PYY) is an endogenous anorectic gut-secreted peptide that has been shown to suppress appetite in animals and humans, when given by injection. This study tested if needle-free pulmonary delivery of PYY enables food intake suppression and reduced body weight gain in rats. The PYY pharmacokinetics and effects on brain neuropeptide levels were also examined. METHODS: Rats received single or once-daily 7-day pulmonary administration of saline or PYYs. Food intake and body weight gain were monitored to study the effects of different doses (0.08-0.90 mg/kg) of PYY3-36, PYY1-36 and PYY13-36. Plasma PYY pharmacokinetics were determined via enzyme-linked immunosorbent assay. Changes in orexigenic neuropeptide Y (NPY) and c-Fos protein levels in the hypothalamus arcuate nucleus (ARC) were measured by immunofluorescence microscopy. RESULTS: PYY3-36 caused dose-dependent and 4- to 6-h food intake suppression following pulmonary delivery. At 0.80 mg/kg, the effect was significant with 35.1 ± 5.7 and 19.7 ± 4.2% suppression at 4 and 6 h, respectively. Repeated administration for 7 days reduced cumulative body weight gain by 39.4 ± 11.0%. PYY1-36, but not PYY13-36, was equipotent to PYY3-36 in food intake suppression. The plasma PYY concentration reached its peak at 10 min following pulmonary delivery with 12-14% of bioavailability. Increased c-Fos and reduced NPY expressions were observed in the hypothalamus ARC, consistent with the magnitude of food intake suppression by each of the PYYs. CONCLUSIONS: Pulmonary delivery of PYY enabled significant 4- to 6-h food intake suppression via 12-14% of lung absorption and hypothalamic ARC interaction, leading to reduced body weight gain in rats.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Neuropéptido Y/metabolismo , Péptido YY/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Aumento de Peso/efectos de los fármacos , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Microscopía Fluorescente , Neuropéptido Y/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Péptido YY/administración & dosificación , Péptido YY/farmacocinética , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Ratas
12.
Prog Neurobiol ; 196: 101894, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32777329

RESUMEN

An accelerating basic science literature is providing key insights into the mechanisms by which spinal neuropeptide Y (NPY) inhibits chronic pain. A key target of pain inhibition is the Gi-coupled neuropeptide Y1 receptor (Y1). Y1 is located in key sites of pain transmission, including the peptidergic subpopulation of primary afferent neurons and a dense subpopulation of small, excitatory, glutamatergic/somatostatinergic interneurons (Y1-INs) that are densely expressed in the dorsal horn, particularly in superficial lamina I-II. Selective ablation of spinal Y1-INs with an NPY-conjugated saporin neurotoxin attenuates the development of peripheral nerve injury-induced mechanical and cold hypersensitivity. Conversely, conditional knockdown of NPY expression or intrathecal administration of Y1 antagonists reinstates hypersensitivity in models of chronic latent pain sensitization. These and other results indicate that spinal NPY release and the consequent inhibition of pain facilitatory Y1-INs represent an important mechanism of endogenous analgesia. This mechanism can be mimicked with exogenous pharmacological approaches (e.g. intrathecal administration of Y1 agonists) to inhibit mechanical and thermal hypersensitivity and spinal neuron activity in rodent models of neuropathic, inflammatory, and postoperative pain. Pharmacological activation of Y1 also inhibits mechanical- and histamine-induced itch. These immunohistochemical, pharmacological, and cell type-directed lesioning data, in combination with recent transcriptomic findings, point to Y1-INs as a promising therapeutic target for the development of spinally directed NPY-Y1 agonists to treat both chronic pain and itch.


Asunto(s)
Dolor Crónico , Interneuronas , Neuropéptido Y , Prurito , Receptores de Neuropéptido Y , Médula Espinal , Animales , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/metabolismo , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Neuropéptido Y/efectos de los fármacos , Neuropéptido Y/metabolismo , Prurito/tratamiento farmacológico , Prurito/metabolismo , Receptores de Neuropéptido Y/efectos de los fármacos , Receptores de Neuropéptido Y/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
13.
Hippocampus ; 20(7): 820-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19623606

RESUMEN

One hypothesis of depression is that it is caused by reduced neuronal plasticity including hippocampal neurogenesis. In this study, we compared the effects of three long-term antidepressant treatments: escitalopram, voluntary running, and their combination on hippocampal cell proliferation, NPY and the NPY-Y1 receptor mRNAs, targets assumed to be important for hippocampal plasticity and mood disorders. An animal model of depression, the Flinders Sensitive Line (FSL) rat, was used and female rats were chosen because the majority of the depressed population is females. We investigated if these treatments were correlated to immobility, swimming, and climbing behaviors, which are associated with an overall, serotonergic-like and noradrenergic-like antidepressant response, in the Porsolt swim test (PST). Interestingly, while escitalopram, running and their combination increased the number of hippocampal BrdU immunoreactive cells, the antidepressant-like effect was only detected in the running group and the group with access both to running wheel and escitalopram. Hippocampal NPY mRNA and the NPY-Y1 receptor mRNA were elevated by running and the combined treatment. Moreover, correlations were detected between NPY mRNA levels and climbing and cell proliferation and NPY-Y1 receptor mRNA levels and swimming. Our results suggest that increased cell proliferation is not necessarily associated with an antidepressant effect. However, treatments that were associated with an antidepressant-like effect did regulate hippocampal levels of mRNAs encoding NPY and/or the NPY-Y1 receptor and support the notion that NPY can stimulate cell proliferation and induce an antidepressant-like response.


Asunto(s)
Antidepresivos/farmacología , Citalopram/farmacología , Depresión/fisiopatología , Actividad Motora/fisiología , Neuropéptido Y/biosíntesis , Receptores de Neuropéptido Y/biosíntesis , Animales , Bromodesoxiuridina , Proliferación Celular , Depresión/metabolismo , Depresión/terapia , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Hipocampo/fisiopatología , Inmunohistoquímica , Hibridación in Situ , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Neuropéptido Y/efectos de los fármacos , ARN Mensajero/análisis , Ratas , Receptores de Neuropéptido Y/efectos de los fármacos , Carrera
14.
Diabetes Obes Metab ; 12(7): 591-603, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20590734

RESUMEN

AIMS: Energy homeostasis is regulated by a complex interaction of molecules and pathways, and new antiobesity treatments are likely to require multiple pharmacological targeting of anorexigenic or orexigenic pathways to achieve effective loss of excess body weight and adiposity. Cannabinoids, acting via the cannabinoid-1 (CB1) receptor, and neuropeptide Y (NPY) are important modulators of feeding behaviour, energy metabolism and body composition. We investigated the interaction of CB1 and NPY in the regulation of energy homeostasis, hypothesizing that dual blockade of CB1 and NPY signalling will induce greater weight and/or fat loss than that induced by single blockade of either system alone. METHODS: We studied the effects of the CB1 antagonist Rimonabant on food intake, body weight, body composition, energy metabolism and bone physiology in wild-type (WT) and NPY knockout (NPY(-/-)) mice. Rimonabant was administered orally at 10 mg/kg body weight twice per day for 3 weeks. Oral Rimonabant was delivered voluntarily to mice via a novel method enabling studies to be carried out in the absence of gavage-induced stress. RESULTS: Mice with dual blockade of CB1 and NPY signalling (Rimonabant-treated NPY(-/-) mice) exhibited greater reductions in body weight and adiposity than mice with single blockade of either system alone (Rimonabant-treated WT or vehicle-treated NPY(-/-) mice). These changes occurred without loss of lean tissue mass or bone mass. Furthermore, Rimonabant-treated NPY(-/-) mice showed a lower respiratory exchange ratio than that seen in Rimonabant-treated WT or vehicle-treated NPY(-/-) mice, suggesting that this additive effect of dual blockade of CB1 and NPY involves promotion of lipid oxidation. On the other hand, energy expenditure and physical activity were comparable amongst all treatment groups. Interestingly, Rimonabant similarly and transiently reduced spontaneous and fasting-induced food intake in WT and NPY(-/-) mice in the first hour after administration only, suggesting independent regulation of feeding by CB1 and NPY signalling. In contrast, Rimonabant increased serum corticosterone levels in WT mice, but this effect was not seen in NPY(-/-) mice, indicating that NPY signalling may be required for effects of CB1 on the hypothalamo-pituitary-adrenal axis. CONCLUSIONS: Dual blockade of CB1 and NPY signalling leads to additive reductions in body weight and adiposity without concomitant loss of lean body mass or bone mass. An additive increase in lipid oxidation in dual CB1 and NPY blockade may contribute to the effect on adiposity. These findings open new avenues for more effective treatment of obesity via dual pharmacological manipulations of the CB1 and NPY systems.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Neuropéptido Y/efectos de los fármacos , Obesidad/tratamiento farmacológico , Piperidinas/antagonistas & inhibidores , Pirazoles/antagonistas & inhibidores , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptores de Neuropéptido Y/efectos de los fármacos , Animales , Composición Corporal , Metabolismo Energético/efectos de los fármacos , Homeostasis/fisiología , Ratones , Neuropéptido Y/metabolismo , Obesidad/metabolismo , Oxidación-Reducción , Piperidinas/administración & dosificación , Pirazoles/administración & dosificación , Receptor Cannabinoide CB1/administración & dosificación , Rimonabant
15.
Neuropharmacology ; 181: 108350, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33027625

RESUMEN

Opioid addiction is a brain disease that severely harms society and personal health. Although the tremendous numbers of patients worldwide and emerged negative events, effective treatments for opioid addiction are still lacking. Neuropeptide Y (NPY) is one of the main orexigenic peptides that play vital roles in food intake and energy metabolism. However, increasing evidence indicates that NPY may have great potential in mediating reward effects and drug dependence. In the present study, we assessed the expression changes of NPY in the nucleus accumbens at different timepoints following morphine conditioned place preference (CPP) and investigated the functional importance of potential NPY changes. Our results showed that NPY expression significantly decreased in the nucleus accumbens shell (AcbSh) immediately after chronic morphine exposure. Subsequently, it increased rapidly at first and then gradually returned to normal levels. Further data indicated that these NPY changes were involved in morphine reward memory, demonstrated by a reduction in the extinction period after blocking of the Y5 receptor by L-152,804 in the AcbSh and a prolonged duration of the extinction period following the application of NPY. More importantly, the additional results revealed that L-152,804 also remarkably suppressed the reinstatement of morphine CPP. Together, our results indicate that a complicated plasticity of the NPY pathway in AcbSh occurs following morphine CPP, and this plasticity plays an important role in modulating morphine reward memory. These findings may enhance our understanding of the role of the NPY system in opioid addiction and indicate a promising target for opioid addiction treatment.


Asunto(s)
Dependencia de Morfina/psicología , Morfina/farmacología , Narcóticos/farmacología , Neuropéptido Y/efectos de los fármacos , Animales , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Masculino , Plasticidad Neuronal/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/antagonistas & inhibidores , Recompensa
16.
Behav Brain Res ; 380: 112369, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31743731

RESUMEN

It is well accepted that opioids promote feeding for reward. Some studies suggest a potential involvement in hunger-driven intake, but they suffer from the scarcity of methodologies differentiating between factors that intersect eating for pleasure versus energy. Here, we used a unique food deprivation discrimination paradigm to test a hypothesis that, since opioids appear to control feeding reward, injection of opioid agonists would not produce effects akin to 22 h of food deprivation. We trained rats to discriminate between 22 h and 2 h food deprivation in a two-lever, operant discrimination procedure. We tested whether opioid agonists at orexigenic doses produce discriminative stimulus effects similar to 22 h deprivation. We injected DAMGO, DSLET, or orphanin FQ in the paraventricular hypothalamic nucleus (PVN), a site regulating hunger/satiety, and butorphanol subcutaneously (to produce maximum consumption). We assessed the ability of the opioid antagonist, naltrexone, to reduce the discriminative stimulus effects of 22 h deprivation and of the 22 h deprivation-like discriminative stimulus effects of PVN-injected hunger mediator, neuropeptide Y (NPY). In contrast to PVN NPY, centrally or peripherally injected opioid agonists failed to induce discriminative stimuli similar to those of 22 h deprivation. In line with that, naltrexone did not reduce the hunger discriminative stimuli induced by either 22 h deprivation or NPY administration in 2 h food-restricted subjects, even though doses used therein were sufficient to decrease deprivation-induced feeding in a non-operant setting in animals familiar with consequences of 2 h and 22 h deprivation. We conclude that opioids promote feeding for reward rather than in order to replenish lacking energy.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Animal/efectos de los fármacos , Aprendizaje Discriminativo/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Privación de Alimentos , Hambre/efectos de los fármacos , Antagonistas de Narcóticos/farmacología , Neuropéptido Y/efectos de los fármacos , Saciedad/efectos de los fármacos , Percepción del Tiempo/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Animales , Condicionamiento Operante/efectos de los fármacos , Masculino , Naltrexona/farmacología , Antagonistas de Narcóticos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Recompensa
17.
Ann Anat ; 227: 151419, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31563570

RESUMEN

BACKGROUND: Neuropeptide Y (NPY) plays a crucial role in many neurobiological functions, such as cognition and memory. Cognitive and memory impairment have been described in diabetic patients. The metabolism of NPY is determined by the activity of proteases, primarily dipeptidyl-peptidase-IV (DPP-IV). Therefore, DPP-IV inhibitors, such as sitagliptin, may modulate the function of NPY. In this study, we investigated the effect of type 1 diabetes and sitagliptin treatment on the regulation of the mRNA encoding for NPY and its receptors (Y1, Y2, and Y5 receptors) in the hippocampus. METHODS: Type 1 diabetes was induced in male Wistar rats by i.p. injection of streptozotocin. Starting two weeks after diabetes onset, animals were treated orally with sitagliptin (5mg/kg, daily) for two weeks. The mRNA expression of Npy and its receptors (Npy1r, Npy2r, and Npy5r) in the hippocampus was evaluated using in situ hybridization with 33P-labeled oligonucleotides. RESULTS: The mRNA expression of Npy, Npy1r and Npy5r was higher in the dentate gyrus, whereas Npy2r highest level was observed in the CA3 subregion. The mRNA expression of Npy, Npy1r and Npy5r in dentate gyrus, CA1 and CA3 was not affected by diabetes and/or by sitagliptin treatment. Type 1 diabetes increased the mRNA expression of Npy2r in the CA3 subregion, which was prevented by sitagliptin treatment. CONCLUSIONS: Our results show that type 1 diabetes, at early stages, induces mild changes in the NPY system in the hippocampus that were counteracted by sitagliptin treatment.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/fisiopatología , Neuropéptido Y/genética , ARN Mensajero/metabolismo , Análisis de Varianza , Animales , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Masculino , Neuropéptido Y/efectos de los fármacos , Neuropéptido Y/fisiología , Sondas de Oligonucleótidos , Distribución Aleatoria , Ratas , Ratas Wistar , Fosfato de Sitagliptina/farmacología , Fosfato de Sitagliptina/uso terapéutico
18.
Psychoneuroendocrinology ; 114: 104594, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32007669

RESUMEN

Second generation antipsychotics, particularly olanzapine, induce severe obesity, which is associated with their antagonistic effect on the histamine H1 receptor (H1R). We have previously demonstrated that oral administration of olanzapine increases the concentration of neuropeptide Y (NPY) in the hypothalamus of rats, accompanied by hyperphagia and weight gain. However, it is unclear if the increased NPY after olanzapine administration is due to its direct effect on hypothalamic neurons and its H1R antagonistic property. In the present study, we showed that with an inverted U-shape dose-response curve, olanzapine increased NPY expression in the NPY-GFP hypothalamic neurons; however, this was not the case in the hypothalamic neurons of H1R knockout mice. Olanzapine inhibited the interaction of H1R and GHSR1a (ghrelin receptor) in the primary mouse hypothalamic neurons and NPY-GFP neurons examined by confocal fluorescence resonance energy transfer (FRET) technology. Furthermore, an H1R agonist, FMPH inhibited olanzapine activation of GHSR1a downstream signaling pAMPK and transcription factors of NPY (pFOXO1 and pCREB) in the hypothalamic NPY-GFP cell. However, an olanzapine analogue (E-Olan) with lower affinity to H1R presented negligible enhancement of pCREB within the nucleus of NPY neurons. These findings suggest that the H1R antagonist property of olanzapine inhibits the interaction of H1R and GHSR1a, activates GHSR1a downstream signaling pAMPK-FOXO1/pCREB and increases hypothalamic NPY: this could be one of the important molecular mechanisms of H1R antagonism of olanzapine-induced obesity in antipsychotic management of psychiatric disorders.


Asunto(s)
Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Antipsicóticos/farmacología , Hipotálamo/efectos de los fármacos , Neuropéptido Y/efectos de los fármacos , Olanzapina/farmacología , Receptores de Ghrelina/efectos de los fármacos , Receptores Histamínicos H1/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Antagonistas de los Receptores Histamínicos H1/farmacología , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
19.
Aging (Albany NY) ; 12(3): 2101-2122, 2020 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-32007953

RESUMEN

As the elderly population grows, chronic metabolic dysfunction including obesity and diabetes are becoming increasingly common comorbidities. Hypothalamic inflammation through CNS resident microglia serves as a common pathway between developing obesity and developing systemic aging pathologies. Despite understanding aging as a life-long process involving interactions between individuals and their environment, limited studies address the dynamics of environment interactions with aging or aging therapeutics. We previously demonstrated environmental enrichment (EE) is an effective model for studying improved metabolic health and overall healthspan in mice, which acts through a brain-fat axis. Here we investigated the CSF1R inhibitor PLX5622 (PLX), which depletes microglia, and its effects on metabolic decline in aging in interaction with EE. PLX in combination with EE substantially improved metabolic outcomes in middle-aged female mice over PLX or EE alone. Chronic PLX treatment depleted 75% of microglia from the hypothalamus and reduced markers of inflammation without affecting brain-derived neurotrophic factor levels induced by EE. Adipose tissue remodeling and adipose tissue macrophage modulation were observed in response to CSF1R inhibition, which may contribute to the combined benefits seen in EE with PLX. Our study suggests benefits exist from combined drug and lifestyle interventions in aged animals.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Envejecimiento/metabolismo , Vivienda para Animales , Microglía/efectos de los fármacos , Compuestos Orgánicos/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Medio Social , Tejido Adiposo/metabolismo , Animales , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Prueba de Tolerancia a la Glucosa , Hormona Liberadora de Gonadotropina/efectos de los fármacos , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inflamación/genética , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Neuropéptido Y/efectos de los fármacos , Neuropéptido Y/genética , Proopiomelanocortina/efectos de los fármacos , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transcriptoma/efectos de los fármacos , Pérdida de Peso
20.
Neuropsychopharmacology ; 44(6): 1132-1140, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30647448

RESUMEN

Neuropeptide Y (NPY) signaling via limbic NPY1 and 2 receptors (NPY1R and NPY2R, respectively) is known to modulate binge-like ethanol consumption in rodents. However, the role of NPY signaling in the medial prefrontal cortex (mPFC), which provides top-down modulation of the limbic system, is unknown. Here, we used "drinking-in-the-dark" (DID) procedures in C57BL/6J mice to address this gap in the literature. First, the impact of DID on NPY immunoreactivity (IR) was assessed in the mPFC. Next, the role of NPY1R and NPY2R signaling in the mPFC on ethanol consumption was evaluated through site-directed pharmacology. Chemogenetic inhibition of NPY1R+ neurons in the mPFC was performed to further evaluate the role of this population. To determine the potential role of NPY1R+ neurons projecting from the mPFC to the basolateral amygdala (BLA) this efferent population was selectively silenced. Three, 4-day cycles of DID reduced NPY IR in the mPFC. Intra-mPFC activation of NPY1R and antagonism of NPY2R resulted in decreased binge-like ethanol intake. Silencing of mPFC NPY1R+ neurons overall, and specifically NPY1R+ neurons projecting to the BLA, significantly reduced binge-like ethanol intake. We provide novel evidence that (1) binge-like ethanol intake reduces NPY levels in the mPFC; (2) activation of NPY1R or blockade of NPY2R reduces binge-like ethanol intake; and (3) chemogenetic inhibition of NPY1R+ neurons in the mPFC and NPY1R+ mPFC neurons projecting to the BLA blunts binge-like drinking. These observations provide the first direct evidence that NPY signaling in the mPFC modulates binge-like ethanol consumption.


Asunto(s)
Complejo Nuclear Basolateral/metabolismo , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Neuropéptido Y/metabolismo , Corteza Prefrontal/metabolismo , Receptores de Neuropéptido Y/metabolismo , Animales , Complejo Nuclear Basolateral/efectos de los fármacos , Consumo Excesivo de Bebidas Alcohólicas/prevención & control , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Neuropéptido Y/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Receptores de Neuropéptido Y/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda