Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Comp Neurol ; 529(4): 657-693, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32621762

RESUMEN

The parabrachial nucleus (PB) is a complex structure located at the junction of the midbrain and hindbrain. Its neurons have diverse genetic profiles and influence a variety of homeostatic functions. While its cytoarchitecture and overall efferent projections are known, we lack comprehensive information on the projection patterns of specific neuronal subtypes in the PB. In this study, we compared the projection patterns of glutamatergic neurons here with a subpopulation expressing the transcription factor Foxp2 and a further subpopulation expressing the neuropeptide Pdyn. To do this, we injected an AAV into the PB region to deliver a Cre-dependent anterograde tracer (synaptophysin-mCherry) in three different strains of Cre-driver mice. We then analyzed 147 neuroanatomical regions for labeled boutons in every brain (n = 11). Overall, glutamatergic neurons in the PB region project to a wide variety of sites in the cerebral cortex, basal forebrain, bed nucleus of the stria terminalis, amygdala, diencephalon, and brainstem. Foxp2 and Pdyn subpopulations project heavily to the hypothalamus, but not to the cortex, basal forebrain, or amygdala. Among the few differences between Foxp2 and Pdyn cases was a notable lack of Pdyn projections to the ventromedial hypothalamic nucleus. Our results indicate that genetic identity determines connectivity (and therefore, function), providing a framework for mapping all PB output projections based on the genetic identity of its neurons. Using genetic markers to systematically classify PB neurons and their efferent projections will enhance the translation of research findings from experimental animals to humans.


Asunto(s)
Encefalinas/biosíntesis , Factores de Transcripción Forkhead/biosíntesis , Núcleos Parabraquiales/metabolismo , Precursores de Proteínas/biosíntesis , Proteínas Represoras/biosíntesis , Proteína 2 de Transporte Vesicular de Glutamato/biosíntesis , Animales , Tronco Encefálico/química , Tronco Encefálico/metabolismo , Corteza Cerebral/química , Corteza Cerebral/metabolismo , Vías Eferentes/química , Vías Eferentes/metabolismo , Encefalinas/análisis , Encefalinas/genética , Femenino , Factores de Transcripción Forkhead/análisis , Factores de Transcripción Forkhead/genética , Hipotálamo/química , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Núcleos Parabraquiales/química , Precursores de Proteínas/análisis , Precursores de Proteínas/genética , Proteínas Represoras/análisis , Proteínas Represoras/genética , Tálamo/química , Tálamo/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/análisis , Proteína 2 de Transporte Vesicular de Glutamato/genética
2.
Gene ; 742: 144583, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32184167

RESUMEN

BACKGROUND: Studies showed that increased let-7b-5p microRNA during repeated electroacupuncture (EA) treatment was associated the formation of EA tolerance, which manifested as gradually decreased nociceptive threshold. Proenkephalin (PENK) is the precursor of enkephalin which is a pivot neuropeptide responsible for the decreased nociceptive threshold in EA. The aim of this study was to evaluate the relationship between let-7b-5p and PENK in EA tolerance. METHODS: The target gene of let-7b-5p microRNA was determined through the dual-luciferase reporter assay in cortical neurons. Seventy-two Sprague Dawley rats received a combination of EA and intracerebroventricular injection of microRNA (let-7b-5p agomir, antagomir or their controls). The nociceptive thresholds were assessed with radiant heat tail-flick method. PENK and let-7b-5p were measured with Western Blot and qPCR, respectively, after administration of let-7b-5p agomir, antagomir, and their controls at day 1, 4 and 7. RESULTS: Let-7b-5p targeted the 3' untranslated region of Penk1. The nociceptive thresholds in Let-7b-5p agomir + EA group were decreased (p < 0.05) compared with those in Let-7b-5p antagomir + EA group at day 1 to 7. Compared with Let-7b-5p agomir + EA group, the expression level of PENK in Let-7b-5p antagomir + EA group was increased at days 1, 4, and 7 (p < 0.05) CONCLUSION: Let-7b-5p may be a new potential target for decreasing the EA tolerance effect and facilitating the application of EA in treating chronic nociception of patients.


Asunto(s)
Electroacupuntura , Encefalinas/genética , MicroARNs/metabolismo , Dolor Nociceptivo/terapia , Precursores de Proteínas/genética , Animales , Antagomirs/administración & dosificación , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/inmunología , Humanos , Inyecciones Intraventriculares , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Nocicepción/efectos de los fármacos , Dolor Nociceptivo/diagnóstico , Dolor Nociceptivo/genética , Dolor Nociceptivo/inmunología , Umbral del Dolor/efectos de los fármacos , Ratas
3.
Physiol Rep ; 8(5): e14399, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32170819

RESUMEN

Expression of particular genes in hypothami of ewes was measured across the natural pubertal transition by in situ hybridization. The ewes were allocated to three groups (n = 4); prepubertal, postpubertal and postpubertally gonadectomized (GDX). Prepubertal sheep were euthanized at 20 weeks of age and postpubertal animals at 32 weeks. GDX sheep were also euthanized at 32 weeks, 1 week after surgery. Expression of KISS1, TAC3, PDYN in the arcuate nucleus (ARC), RFRP in the dorsomedial hypothalamus and GNRH1 in the preoptic area was quantified on a cellular basis. KISS1R expression by GNRH1 cells was quantified by double-label in situ hybridization. Across puberty, detectable KISS1 cell number increased in the caudal ARC and whilst PDYN cell numbers were low, numbers increased in the rostral ARC. TAC3 expression did not change but RFRP expression/cell was reduced across puberty. There was no change across puberty in the number of GNRH1 cells that expressed the kisspeptin receptor (KISS1R). GDX shortly after puberty did not increase expression of any of the genes of interest. We conclude that KISS1 expression in the ARC increases during puberty in ewes and this may be a causative factor in the pubertal activation of the reproductive axis. A reduction in expression of RFRP may be a factor in the onset of puberty, removing negative tone on GNRH1 cells. The lack of changes in expression of genes following GDX suggest that the effects of gonadal hormones may differ in young and mature animals.


Asunto(s)
Encefalinas/genética , Expresión Génica , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Kisspeptinas/genética , Neuroquinina B/genética , Neuropéptidos/genética , Precursores de Proteínas/genética , Animales , Femenino , Receptores de Kisspeptina-1/genética , Oveja Doméstica/genética , Oveja Doméstica/crecimiento & desarrollo
4.
Genes Brain Behav ; 18(8): e12600, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31339663

RESUMEN

Persistent alterations of proopiomelanocortin (Pomc) and mu-opioid receptor (Oprm1) activity and stress responses after alcohol are critically involved in vulnerability to alcohol dependency. Gene transcriptional regulation altered by alcohol may play important roles. Mice with genome-wide deletion of neuronal Pomc enhancer1 (nPE1-/- ), had hypothalamic-specific partial reductions of beta-endorphin and displayed lower alcohol consumption, compared to wildtype littermates (nPE1+/+ ). We used RNA-Seq to measure steady-state nuclear mRNA transcripts of opioid and stress genes in hypothalamus of nPE1+/+ and nPE1-/- mice after 1-day acute withdrawal from chronic excessive alcohol drinking or after water. nPE1-/- had lower basal Pomc and Pdyn (prodynorphin) levels compared to nPE1+/+ , coupled with increased basal Oprm1 and Oprk1 (kappa-opioid receptor) levels, and low alcohol drinking increased Pomc and Pdyn to the basal levels of nPE1+/+ in the water group, without significant effects on Oprm1 and Oprk1. In nPE1+/+ , excessive alcohol intake increased Pomc and Oprm1, with no effect on Pdyn or Oprk1. For stress genes, nPE1-/- had lowered basal Oxt (oxytocin) and Avp (arginine vasopressin) that were restored by low alcohol intake to basal levels of nPE1+/+ . In nPE1+/+ , excessive alcohol intake decreased Oxt and Avpi1 (AVP-induced protein1). Functionally examining the effect of pharmacological blockade of mu-opioid receptor, we found that naltrexone reduced excessive alcohol intake in nPE1+/+ , but not nPE1-/- . Our results provide evidence relevant to the transcriptional profiling of the critical genes in mouse hypothalamus: enhanced opioid and reduced stress gene transcripts after acute withdrawal from excessive alcohol may contribute to altered reward and stress responses.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/genética , Elementos de Facilitación Genéticos/genética , Hipotálamo/metabolismo , Proopiomelanocortina/genética , Animales , Arginina Vasopresina/metabolismo , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Encefalinas/genética , Encefalinas/metabolismo , Etanol/farmacología , Hipotálamo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Oxitocina/metabolismo , Proopiomelanocortina/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Transcriptoma
5.
Toxicology ; 366-367: 1-9, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27495953

RESUMEN

Atrazine is an herbicide used to control broadleaf grasses and a suspected endocrine disrupting chemical. Snapping turtles lay eggs between late May and early June, which could lead to atrazine exposure via field runoff. Our goal was to determine whether a single exposure to 2ppb or 40ppb atrazine during embryogenesis could induce short- and long-term changes in gene expression within the hypothalamus of snapping turtles. We treated eggs with atrazine following sex determination and measured gene expression within the hypothalamus. We selected genes a priori for their role in the hypothalamus-pituitary-gonad or the hypothalamus-pituitary-adrenal axes of the endocrine system. We did not identify any changes in gene expression 24-h after treatment. However, at hatching AR, Kiss1R, and POMC expression was upregulated in both sexes, while expression of CYP19A1 and PDYN was increased in females. Six months after hatching, CYP19A1 and PRLH expression was increased in animals treated with 2ppb atrazine. Our study shows persistent changes in hypothalamic gene expression due to low-dose embryonic exposure to the herbicide atrazine with significant effects in both the HPG and HPA axes. Effects reported here appear to be conserved among vertebrates.


Asunto(s)
Atrazina/toxicidad , Herbicidas/toxicidad , Hipotálamo/efectos de los fármacos , Organogénesis/efectos de los fármacos , Reproducción/efectos de los fármacos , Animales , Aromatasa/genética , Aromatasa/metabolismo , Relación Dosis-Respuesta a Droga , Encefalinas/genética , Encefalinas/metabolismo , Femenino , Regulación de la Expresión Génica , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/metabolismo , Masculino , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Hormona Liberadora de Prolactina/genética , Hormona Liberadora de Prolactina/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Factores Sexuales , Tortugas/embriología
6.
Oncol Rep ; 35(4): 1941-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26782786

RESUMEN

5-Aza-2'-deoxycytidine (5-Aza-CdR) is currently acknowledged as a demethylation drug, and causes a certain degree of demethylation in a variety of cancer cells, including pancreatic cancer cells. Emodin, a traditional Chinese medicine (TCM), is an effective monomer extracted from rhubarb and has been reported to exhibit antitumor activity in different manners in pancreatic cancer. In the present study, we examined whether emodin caused demethylation and increased the demethylation of three tumor-suppressor genes P16, RASSF1A and ppENK with a high degree of methylation in pancreatic cancer when combined with 5-Aza-CdR. Our research showed that emodin inhibited the growth of pancreatic cancer Panc-1 cells in a dose- and time-dependent manner. Dot-blot results showed that emodin combined with 5-Aza-CdR significantly suppressed the expression of genome 5mC in PANC-1 cells. In order to verify the effect of methylation, methylation-specific PCR (MSP) and bisulfite genomic sequencing PCR (BSP) combined with TA were selected for the cloning and sequencing. Results of MSP and BSP confirmed that emodin caused faint demethylation, and 5-Aza-CdR had a certain degree of demethylation. When emodin was combined with 5-Aza-CdR, the demethylation was more significant. At the same time, fluorescent quantitative PCR and western blot analysis results confirmed that when emodin was combined with 5-Aza-CdR, the expression levels of P16, RASSF1A and ppENK were increased more significantly compared to either treatment alone. In contrast, the expression levels of DNA methyltransferase 1 (DNMT1) and DNMT3a were more significantly reduced with the combination treatment than the control or either agent alone, further proving that emodin in combination with 5-Aza-CdR enhanced the demethylation effect of 5-Aza-CdR by reducing the expression of methyltransferases. In conclusion, the present study confirmed that emodin in combination with 5-Aza-CdR enhanced the demethylation by 5-Aza-CdR of tumor-suppressor genes p16, RASSF1A and ppENK by reducing the expression of methyltransferases DNMT1 and DNMT3a.


Asunto(s)
Azacitidina/análogos & derivados , Metilación de ADN/efectos de los fármacos , Emodina/farmacología , Encefalinas/genética , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Precursores de Proteínas/genética , Proteínas Supresoras de Tumor/genética , Azacitidina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Decitabina , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Regiones Promotoras Genéticas/efectos de los fármacos , Análisis de Secuencia de ADN/métodos
7.
Cell Metab ; 22(6): 1059-67, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26655697

RESUMEN

In this report we evaluated the functions of hypothalamic amylin in vivo and in vitro. Profiling of hypothalamic neurons revealed that islet amyloid polypeptide (Iapp, precursor to amylin) is expressed in neurons in the lateral hypothalamus, arcuate nucleus, medial preoptic area, and elsewhere. Hypothalamic expression of lapp is markedly decreased in ob/ob mice and normalized by exogenous leptin. In slices, amylin and leptin had similar electrophysiologic effects on lateral hypothalamic leptin receptor ObRb-expressing neurons, while the amylin antagonist AC187 inhibited their activity and blunted the effect of leptin. Finally, i.c.v. infusion of AC187 acutely reduced the anorectic effects of leptin. These data show that hypothalamic amylin is transcriptionally regulated by leptin, that it can act directly on ObRb neurons in concert with leptin, and that it regulates feeding. These findings provide a potential mechanism for the increased efficacy of a metreleptin/pramlintide combination therapy for obesity.


Asunto(s)
Hipotálamo/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Leptina/metabolismo , Animales , Ingestión de Alimentos/efectos de los fármacos , Encefalinas/genética , Encefalinas/metabolismo , Femenino , Hipotálamo/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/antagonistas & inhibidores , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Leptina/sangre , Leptina/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Microscopía Confocal , Neuronas/metabolismo , Técnicas de Placa-Clamp , Fragmentos de Péptidos/farmacología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
8.
J Tradit Chin Med ; 34(1): 90-5, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25102697

RESUMEN

OBJECTIVE: To observe the influence of different moxibustion durations on hypothalamic pro-opiomelanocortin (POMC) and prodynorphin (PDYN) mRNA expressions and plasma beta-endorphin (beta-EP) content in rheumatoid arthritis (RA) rats, to understand the mechanism of moxibustion analgesia and its dose-effect relationship. METHODS: Twelve male Wistar rats were randomly selected from 48 male Wistar rats as a normal control group. The RA model was created by raising rats in a windy (blowing with electric fan), cold (6 degrees C +/- 2 degrees C), and wet (80%-90% humidity) environment for 20 days, 12 h each day. This was followed by injection of Freund's complete adjuvant (0.15 mL) into the ankle. Then, rats were randomly divided into a model group, moxibustion group I, and moxibustion group II, with 12 rats in each group. In moxibustion groups I and II, moxibustion was given at Shenshu (BL 23) and Zusanli (ST 36) for 20 and 40 min, respectively, once daily for 15 days. Hypothalamic POMC and PDYN mRNA expression levels and plasma beta-EP content were determined. RESULTS: Compared with the normal group, the pressure pain threshold decreased, while the hypothalamic POMC and PDYN mRNA expression levels and plasma beta-EP content increased in the moxibustion groups (P < 0.01). Compared with the model group, the pressure pain threshold, hypothalamic POMC and PDYN mRNA expression levels and plasma beta-EP content in the moxibustion groups increased significantly (P < 0.01). Compared the moxibustion group I, the pain threshold, hypothalamic POMC and PDYN mRNA expression levels and plasma beta-EP content in moxibustion group II significantly increased (P < 0.01). CONCLUSION: Moxibustion has an analgesic effect and increases hypothalamic POMC and PDYN mRNA expression levels and plasma beta-EP content in RA rats.The analgesic effect in moxibustion group II is betterthan that in moxibustion group I.


Asunto(s)
Artralgia/terapia , Artritis Reumatoide/terapia , Moxibustión , Puntos de Acupuntura , Analgesia , Animales , Artralgia/genética , Artralgia/metabolismo , Artritis Reumatoide/genética , Artritis Reumatoide/metabolismo , Modelos Animales de Enfermedad , Encefalinas/genética , Encefalinas/metabolismo , Humanos , Hipotálamo/metabolismo , Masculino , Umbral del Dolor , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Ratas , Ratas Wistar , betaendorfina/sangre
9.
Genes Brain Behav ; 13(6): 550-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24804898

RESUMEN

Enhanced stress reactivity or sensitivity to chronic stress increases the susceptibility to mood pathologies such as major depression. The opioid peptide enkephalin is an important modulator of the stress response. Previous studies using preproenkephalin knockout (PENK KO) mice showed that these animals exhibit abnormal stress reactivity and show increased anxiety behavior in acute stress situations. However, the consequence of enkephalin deficiency in the reactivity to chronic stress conditions is not known. In this study, we therefore submitted wild-type (WT) and PENK KO male mice to chronic stress conditions, using the chronic mild stress (CMS) protocol. Subsequently, we studied the CMS effects on the behavioral and hormonal level and also performed gene expression analyses. In WT animals, CMS increased the expression of the enkephalin gene in the paraventricular nucleus (PVN) of the hypothalamus and elevated the corticosterone levels. In addition, WT mice exhibited enhanced anxiety in the zero-maze test and depression-related behaviors in the sucrose preference and forced swim tests. Surprisingly, in PENK KO mice, we did not detect anxiety and depression-related behavioral changes after the CMS procedure, and even measured a decreased hormonal stress response. These results indicate that PENK KO mice are resistant to the CMS effects, suggesting that enkephalin enhances the reactivity to chronic stress.


Asunto(s)
Ansiedad/genética , Depresión/genética , Encefalinas/genética , Precursores de Proteínas/genética , Estrés Psicológico/genética , Animales , Corticosterona/sangre , Encefalinas/metabolismo , Hipotálamo/metabolismo , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Núcleo Hipotalámico Paraventricular/metabolismo , Precursores de Proteínas/metabolismo
10.
J Neurosci ; 33(34): 13600-11, 2013 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-23966683

RESUMEN

Animal and clinical studies show that gestational exposure to nicotine increases the propensity of offspring to consume nicotine, but the precise mechanism mediating this behavioral phenomenon is unclear. The present study in Sprague Dawley rats examined the possibility that the orexigenic peptide systems, enkephalin (ENK) and orexin (OX), which are stimulated by nicotine in adult animals and promote consummatory behavior, may be similarly responsive to nicotine's stimulatory effect in utero while having long-term behavioral consequences. The results demonstrated that nicotine exposure during gestation at low doses (0.75 or 1.5 mg/kg/d) significantly increased mRNA levels and density of neurons that express ENK in the hypothalamic paraventricular nucleus and central nucleus of the amygdala, OX, and another orexigenic peptide, melanin-concentrating hormone, in the perifornical lateral hypothalamus in preweanling offspring. These effects persisted in the absence of nicotine, at least until puberty. Colabeling of the cell proliferation marker BrdU with the neuronal marker NeuN and peptides revealed a marked stimulatory effect of prenatal nicotine on neurogenesis, but not gliogenesis, and also on the number of newly generated neurons expressing ENK, OX, or melanin-concentrating hormone. During adolescence, offspring also exhibited significant behavioral changes, increased consumption of nicotine and other substances of abuse, ethanol and a fat-rich diet, with no changes in chow and water intake or body weight. These findings reveal a marked sensitivity during gestation of the orexigenic peptide neurons to low nicotine doses that may increase the offspring's propensity to overconsume substances of abuse during adolescence.


Asunto(s)
Amígdala del Cerebelo/citología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipotálamo/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neurogénesis/efectos de los fármacos , Neuropéptidos/metabolismo , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Factores de Edad , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/crecimiento & desarrollo , Amígdala del Cerebelo/metabolismo , Animales , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Encefalinas/genética , Encefalinas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Hormonas Hipotalámicas/genética , Hormonas Hipotalámicas/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Etiquetado Corte-Fin in Situ , Péptidos y Proteínas de Señalización Intracelular/genética , Melaninas/genética , Melaninas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptidos/genética , Orexinas , Fosfopiruvato Hidratasa , Hormonas Hipofisarias/genética , Hormonas Hipofisarias/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Zhongguo Zhen Jiu ; 33(5): 433-7, 2013 May.
Artículo en Chino | MEDLINE | ID: mdl-23885620

RESUMEN

OBJECTIVE: To explore the central mechanism of moxibustion on analgesic effect. METHODS: Male Wistar rats were screened by pain threshold value before making model, and 48 rats whose pain threshold was (250 +/- 25) g were selected. Twelve male Wistar rats were randomly selected as a normal group. For the rest rats the rheumatoid arthritis (RA) model was duplicated by raising in a windy, cold and wet environment combined with injection of Freund's complete adjuvant (FCA), and then they were randomly divided into a model group, a moxibustion group and a moxa volatile oil group, 12 rats in each group. The moxibustion and the moxa volatile oil igroup were treated with moxibustion and moxa volatile oil at "Shenshu"(BL 23) and "Zusanli"(ST 36), respectively, for 15 days. No interventions were added on the model group and the normal group. The pain threshold in Iinjured foot and the expression of hypothalamic POMC mRNA and PDYN mRNA in rats were observed. RESULTS: Compared with the normal group, the pain threshold and the expression of hypothalamic POMC mRNA and PDYN mRNA in the model group were increased (all P < 0.01). Compared with the model group, the pain threshold and the expression of hypothalamic POMC mRNA and PDYN mRNA in the moxibustion group were increased significantly (all P < 0.01), but no statistically significance in the moxa volatile oil group (P > 0.05). Compared with the moxa volatile oil group, the above-mentioned observative indices in moxibustion group were all increased significantly (all P < 0.01). CONCLUSION: Moxibustion has obvious analgesic effect and its mechanism may be related to the increasing expression of hypothalamic POMC and PDYN mRNA through the warming effect of moxibustion.


Asunto(s)
Artritis Reumatoide/genética , Artritis Reumatoide/terapia , Encefalinas/genética , Hipotálamo/metabolismo , Moxibustión , Proopiomelanocortina/genética , Precursores de Proteínas/genética , Animales , Artritis Reumatoide/metabolismo , Encefalinas/metabolismo , Humanos , Masculino , Proopiomelanocortina/metabolismo , Precursores de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
12.
Eur J Pharmacol ; 711(1-3): 10-8, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23624329

RESUMEN

Despite their widespread use in opioid maintenance treatment and pain management, little is known about the intracellular effectors of methadone and buprenorphine and the transcriptional responses they induce. We therefore studied the acute effects of these two opioids in rats, comparing our observations with those for the reference molecule, morphine. We determined the analgesic ED50 of the three molecules in the tail flick test, to ensure that transcriptional effects were compared between doses of equivalent analgesic effect. We analysed changes in gene expression over time in three cerebral structures involved in several opioid behaviours-the dorsal striatum, thalamus and nucleus accumbens-by real-time quantitative PCR. We analysed the expression of genes encoding proteins of the endogenous opioid system in parallel with that of Fos, a marker of neuronal activation. The acute transcriptional effects of methadone resembled those of morphine more closely than did those of buprenorphine, in terms of kinetics and intensities. Our results provide the first evidence that these two drugs widely used in pain management and opioid maintenance treatment can disturb the regulation of endogenous opioid system genes and induce molecular outcomes different from those observed with morphine.


Asunto(s)
Analgésicos Opioides/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Buprenorfina/farmacología , Encefalinas/genética , Masculino , Metadona/farmacología , Morfina/farmacología , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Proopiomelanocortina/genética , Precursores de Proteínas/genética , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides/genética , Receptores Opioides/metabolismo , Tálamo/efectos de los fármacos , Tálamo/metabolismo , Receptor de Nociceptina
13.
In Vitro Cell Dev Biol Anim ; 48(9): 562-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23054438

RESUMEN

Stem cell therapy is one of the most promising treatments in neuroregenerative medicine. Considering the role of the endogenous opioid system in controlling the pathophysiology of neurological disorders and behavioral aberrations, current studies have focused on enkephalins as a part of the opioid system. Due to high capability of unrestricted somatic stem cells (USSCs) and human mesenchymal stem cells (hMSCs) for cell therapy and transplantation; here, we examined their enkephalinergic differentiation potential through Ikaros-related pathways in order to develop in vitro models to help drug screening and stem cell therapy for the opioid-related disorders. The authenticity of the stem cells was verified by differentiation experiments along with flow cytometry for surface markers. Later, we confirmed their neurogenic differentiation with semiquantitative and quantitative transcriptional and translational evaluations of the enkephalinergic-related genes such as proenkephalin, CREBZF, Ikaros, and prodynorphin. Our findings supported the enkephalinergic differentiation of these stem cells. Noteworthy, USSCs showed higher potential for differentiating into enkephalinergic neurons under Ikaros activation than hMSCs, which makes them appropriate for neurological therapeutic applications. In conclusion, this study suggests a powerful in vitro model for neurogenesis that may help clarification of enkephalinergic differentiation and related signaling networks along with neural drug screening. Such investigations may be beneficial to ameliorate the neural-related therapeutic approaches.


Asunto(s)
Dinorfinas/metabolismo , Encefalinas/metabolismo , Neurogénesis , Neuronas/citología , Precursores de Proteínas/metabolismo , Células Madre/citología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Biomarcadores/metabolismo , Evaluación Preclínica de Medicamentos , Encefalinas/genética , Citometría de Flujo , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Factor de Transcripción Ikaros/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neuronas/metabolismo , Precursores de Proteínas/genética , Transducción de Señal , Trasplante de Células Madre , Células Madre/metabolismo
14.
Neurosci Lett ; 477(2): 61-5, 2010 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-20399834

RESUMEN

Electroacupuncture (EA) causes prolonged suppression of reflex elevations in blood pressure for at least 60min in anesthetized preparations. Thus, EA can modify sympathetic outflow and elevated blood pressure through actions in a number of hind brain regions, including the rostral ventrolateral medulla (rVLM). Since our previous data show that the opioid system plays a role in EA-related prolonged inhibition of presympathetic neuronal activity in the rVLM, we postulated that EA increases preproenkephalin (PPE) mRNA in this region, possibly for prolonged periods of time. Under alpha-chloralose anesthesia, rats received EA (1-2mA, 2Hz, 0.5ms) at P5-P6 acupoints (overlying median nerves) or sham (needle placement without electrical stimulation) for 30min. PPE mRNA in the rVLM also was evaluated in control rats that received surgery but no EA, or sham treatment. 20min, 1.5h or 4h following EA or sham treatment, PPE mRNA in the rVLM was analyzed by reverse transcription and quantitative real-time PCR. Relative ratios of PPE mRNA levels (normalized with 18s house keeping gene) were increased 1.5h after EA stimulation (7.77+/-1.39, n=6) relative to sham (2.84+/-0.37, n=5) but were unchanged both 20min and 4h after EA, compared to the sham or surgery groups at the same time points. Thus, 30min of EA transiently stimulates the production of enkephalin in a region of the brain that importantly regulates sympathetic outflow suggesting that even a single brief acupuncture treatment can increase the expression of this modulatory neuropeptide.


Asunto(s)
Electroacupuntura , Encefalinas/biosíntesis , Bulbo Raquídeo/metabolismo , Precursores de Proteínas/biosíntesis , ARN Mensajero/biosíntesis , Animales , Encefalinas/genética , Masculino , Precursores de Proteínas/genética , Ratas , Ratas Sprague-Dawley
15.
Zhen Ci Yan Jiu ; 35(5): 354-8, 393, 2010 Oct.
Artículo en Chino | MEDLINE | ID: mdl-21235063

RESUMEN

OBJECTIVE: To observe the effect of acupotomy lysis (AL) on hypothalamic proopiomelanocortin (POMO) mRNA and preproenkephalin (PPE) mRNA expression in rats with the third lumbar vertebrae transverse process syndrome (TLVTPS) so as to study its underlying mechanism in relieving symptoms of lumbar muscle strain. METHODS: Twenty-four SD rats were randomly divided into normal control group, model group, AL group and electroacupunture (EA) group. The TLVTPS model was established by inserting a piece of gelatin sponge into the posterior of the left 3rd lumbar vertebrae transverse process. AL and EA were applied to the left "Shenshu"(BL 23) and "Yaoyangguan" (GV 3) respectively. The POMC mRNA and PPE mRNA expression levels in the hypothalamus were detected by in situ hybridization. RESULTS: In comparison with the normal group, the integrated optical density (IOD) values of hypothalamic POMC mRNA and PPE mRNA positive cells in the model group were increased significantly (P < 0.01); while compared with the model group, those of POMC mRNA and PPE mRNA positive cells in both left and right hypothalamus were increased further considerably in both AL and EA groups (P < 0.01). No significant differences were found between AL and EA groups in POMC mRNA and PPE mRNA expression levels (P > 0.05). CONCLUSION: AL and EA therapies can increase the expression of POMC mRNA and PPE mRNA in hypothalamus in rats with TLVTPS, which may contribute to its effect in relieving pain in the treatment of lumbar muscle strain.


Asunto(s)
Electroacupuntura , Encefalinas/genética , Expresión Génica , Hipotálamo/metabolismo , Vértebras Lumbares/metabolismo , Manejo del Dolor , Proopiomelanocortina/genética , Precursores de Proteínas/genética , Enfermedades de la Médula Espinal/terapia , Analgesia por Acupuntura , Puntos de Acupuntura , Animales , Encefalinas/metabolismo , Humanos , Masculino , Dolor/genética , Dolor/metabolismo , Proopiomelanocortina/metabolismo , Precursores de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Enfermedades de la Médula Espinal/genética , Enfermedades de la Médula Espinal/metabolismo
16.
Peptides ; 30(12): 2423-31, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19782113

RESUMEN

The peptides galanin (GAL) and orexin (OX) share common features with the opioid enkephalin (ENK) in their relationship to ingestive behavior, stimulating consumption of a fat-rich diet and ethanol when injected into the hypothalamus. Since receptors for GAL and OX are dense in areas where ENK-expressing neurons are concentrated, these non-opioid peptides may exert their effects, in part, through the stimulation of endogenous ENK. This study was conducted to determine whether injection of GAL or OX affects the expression of ENK in hypothalamic and mesolimbic nuclei involved in consummatory behavior. Rats were injected with GAL (1 microg), OX-A (1 microg), or saline vehicle just dorsal to the hypothalamic paraventricular nucleus (PVN). They were sacrificed 1h later for analysis of ENK mRNA levels in the PVN, ventral tegmental area (VTA), central nucleus of the amygdala (CeA), and nucleus accumbens (NAc). Both GAL and OX had similar effects, significantly increasing ENK mRNA expression in each of these areas, except for the NAc. This enhanced ENK expression in the PVN, VTA and CeA was demonstrated with real-time quantitative polymerase chain reaction and confirmed in separate groups using radiolabeled and digoxigenin-labeled in situ hybridization. These findings demonstrate that the non-opioid peptides, GAL or OX, which have similar effects on consummatory behavior, are also similar in their effect on endogenous ENK. In light of published findings showing an opioid antagonist to block GAL- and OX-induced feeding, these results provide additional evidence that ENK is involved in mediating the common behavioral effects of these peptides.


Asunto(s)
Núcleo Celular/metabolismo , Encefalinas/genética , Galanina/farmacología , Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/farmacología , Neuropéptidos/farmacología , Animales , Núcleo Celular/efectos de los fármacos , Galanina/administración & dosificación , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular/administración & dosificación , Masculino , Neuropéptidos/administración & dosificación , Neurotransmisores/farmacología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Orexinas , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
17.
Behav Brain Res ; 197(1): 90-6, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-18762218

RESUMEN

L-DOPA-induced dyskinesia (LID) is among the motor complications that arise in Parkinson patients after a prolonged treatment with levodopa (L-DOPA). Since previous transcriptome and proteomic studies performed in the rat model of LID suggested important changes in striatal energy-related components, we hypothesize that oral creatine supplementation could prevent or attenuate the occurrence of LID. In this study, 6-hydroxydopamine-lesioned rats received a 2% creatine-supplemented diet for 1 month prior to L-DOPA therapy. During the 21 days of L-DOPA treatment, significant reductions in abnormal involuntary movements (AIMs) have been observed in the creatine-supplemented group, without any worsening of parkinsonism. In situ hybridization histochemistry and immunohistochemistry analysis of the striatum also showed a reduction in the levels of prodynorphin mRNA and FosB/DeltaFosB-immunopositive cells in creatine-supplemented diet group, an effect that was dependant on the development of AIMs. Further investigation of the bioenergetics' status of the denervated striatum revealed significant changes in the levels of creatine both after L-DOPA alone and with the supplemented diet. In conclusion, we demonstrated that combining L-DOPA therapy with a diet enriched in creatine could attenuate LID, which may represent a new way to control the motor complications associated with L-DOPA therapy.


Asunto(s)
Creatina/metabolismo , Discinesia Inducida por Medicamentos/metabolismo , Metabolismo Energético/fisiología , Neostriado/metabolismo , Fármacos Neuroprotectores/metabolismo , Administración Oral , Análisis de Varianza , Animales , Creatina/administración & dosificación , Suplementos Dietéticos , Modelos Animales de Enfermedad , Discinesia Inducida por Medicamentos/complicaciones , Discinesia Inducida por Medicamentos/prevención & control , Encefalinas/genética , Encefalinas/metabolismo , Femenino , Técnicas In Vitro , Levodopa/efectos adversos , Levodopa/uso terapéutico , Neostriado/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Oxidopamina , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/complicaciones , Trastornos Parkinsonianos/tratamiento farmacológico , Fosfocreatina/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Estadísticas no Paramétricas
18.
Neuropsychopharmacology ; 34(3): 775-85, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18800067

RESUMEN

Stress and anxiety are mainly regulated by amygdala and hypothalamic circuitries involving several neurotransmitter systems and providing physiological responses to peripheral organs via the hypothalamic-pituitary-adrenal axis and other pathways. The role of endogenous opioid peptides in this process is largely unknown. Here we show for the first time that anxiolytic parameters of explorative behavior in mice lacking prodynorphin were increased 2-4-fold in the open field, the elevated plus maze and the light-dark test. Consistent with this, treatment of wild-type mice with selective kappa-opioid receptor antagonists GNTI or norbinaltorphimine showed the same effects. Furthermore, treatment of prodynorphin knockout animals with U-50488H, a selective kappa-opioid receptor agonist, fully reversed their anxiolytic phenotype. These behavioral data are supported by an approximal 30% reduction in corticotropin-releasing hormone (CRH) mRNA expression in the hypothalamic paraventricular nucleus and central amygdala and an accompanying 30-40% decrease in corticosterone serum levels in prodynorphin knockout mice. Although stress-induced increases in corticosterone levels were attenuated in prodynorphin knockout mice, they were associated with minor increases in depression-like behavior in the tail suspension and forced swim tests. Taken together, our data suggest a pronounced impact of endogenous prodynorphin-derived peptides on anxiety, but not stress coping ability and that these effects are mediated via kappa-opioid receptors. The delay in the behavioral response to kappa-opioid receptor agonists and antagonist treatment suggests an indirect control level for the action of dynorphin, probably by modulating the expression of CRH or neuropeptide Y, and subsequently influencing behavior.


Asunto(s)
Ansiedad/metabolismo , Corticosterona , Encefalinas/fisiología , Neuropéptidos/metabolismo , Precursores de Proteínas/fisiología , Receptores Opioides kappa , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/fisiopatología , Tronco Encefálico/metabolismo , Corticosterona/sangre , Hormona Liberadora de Corticotropina/metabolismo , Dinorfinas/fisiología , Encefalinas/genética , Encefalinas/metabolismo , Conducta Exploratoria , Femenino , Guanidinas , Hipotálamo/metabolismo , Masculino , Aprendizaje por Laberinto , Mesencéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/metabolismo , Morfinanos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Neuropéptido Y/fisiología , Neuropéptidos/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Núcleos del Rafe/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Estrés Psicológico/fisiopatología
19.
J Neuroendocrinol ; 20(12): 1376-81, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19094085

RESUMEN

Human menopause is characterised by ovarian failure, gonadotrophin hypersecretion and hypertrophy of neurones expressing neurokinin B (NKB), kisspeptin (KiSS)-1 and oestrogen receptor (ER) alpha gene transcripts within the hypothalamic infundibular (arcuate) nucleus. In the arcuate nucleus of experimental animals, dynorphin, an opioid peptide, is colocalised with NKB, kisspeptin, ER alpha and progesterone receptors. Moreover, ovariectomy decreases the expression of prodynorphin gene transcripts in the arcuate nucleus of the ewe. Therefore, we hypothesised that the hypertrophied neurones in the infundibular nucleus of postmenopausal women would express prodynorphin mRNA and that menopause would be accompanied by changes in prodynorphin gene transcripts. In the present study, in situ hybridisation was performed on hypothalamic sections from premenopausal and postmenopausal women using a radiolabelled cDNA probe targeted to prodynorphin mRNA. Autoradiography and computer-assisted microscopy were used to map and count labelled neurones, measure neurone size and compare prodynorphin gene expression between premenopausal and postmenopausal groups. Neurones expressing dynorphin mRNA in the infundibular nucleus of the postmenopausal women were larger and exhibited hypertrophied morphological features. Moreover, there were fewer neurones labelled with the prodynorphin probe in the infundibular nucleus of the postmenopausal group compared to the premenopausal group. The number of dynorphin mRNA-expressing neurones was also reduced in the medial preoptic/anterior hypothalamic area of postmenopausal women without changes in cell size. No differences in cell number or size of dynorphin mRNA-expressing neurones were observed in any other hypothalamic region. Previous studies using animal models provide strong evidence that the changes in prodynorphin neuronal size and gene expression in postmenopausal women are secondary to the ovarian failure of menopause. Given the inhibitory effect of dynorphin on the reproductive axis, decreased dynorphin gene expression could play a role in the elevation in luteinising hormone secretion that occurs in postmenopausal women.


Asunto(s)
Encefalinas/genética , Expresión Génica , Hipotálamo/citología , Neuronas , Posmenopausia/fisiología , Precursores de Proteínas/genética , Adolescente , Adulto , Anciano , Animales , Autorradiografía , Forma de la Célula , Encefalinas/metabolismo , Femenino , Humanos , Hipotálamo/metabolismo , Persona de Mediana Edad , Neuronas/citología , Neuronas/fisiología , Precursores de Proteínas/metabolismo , Adulto Joven
20.
Domest Anim Endocrinol ; 34(4): 440-50, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18308503

RESUMEN

Testosterone and season influence mRNA expression for the opioid, enkephalin, in the preoptic area and hypothalamus of rams. Dynorphin is another opioid which has been shown to play a role in the control of reproductive function in females. We now report effects of season and testosterone on the expression of prodynorphin mRNA in the hypothalamus of the ram. Castrated adult Romney Marsh rams (5/group) received vehicle or testosterone propionate (i.m.) during either the 'breeding' season or 'non-breeding' season. Prodynorphin mRNA expression was quantified in the hypothalami by in situ hybridisation. Testosterone treatment increased prodynorphin mRNA expression in the supraoptic nucleus and the bed nucleus of the stria terminalis in the breeding season but not during the non-breeding season. Prodynorphin mRNA expression was also higher in the breeding season than in the non-breeding season in the caudal preoptic area, paraventricular nucleus and accessory supraoptic nucleus, irrespective of treatment. No effects of treatment were observed in any other regions of the hypothalamus. We conclude that testosterone and season regulate prodynorphin mRNA expression in a region-specific manner, which may influence seasonal changes in reproductive function.


Asunto(s)
Encefalinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Área Preóptica/efectos de los fármacos , Precursores de Proteínas/genética , Estaciones del Año , Ovinos/genética , Testosterona/farmacología , Animales , Encefalinas/metabolismo , Hormonas/sangre , Hipotálamo/metabolismo , Masculino , Área Preóptica/metabolismo , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ovinos/sangre , Ovinos/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA