Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 225
Filtrar
1.
Nat Neurosci ; 27(9): 1844-1857, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39009835

RESUMEN

Neuropeptides are ubiquitous in the nervous system. Research into neuropeptides has been limited by a lack of experimental tools that allow for the precise dissection of their complex and diverse dynamics in a circuit-specific manner. Opioid peptides modulate pain, reward and aversion and as such have high clinical relevance. To illuminate the spatiotemporal dynamics of endogenous opioid signaling in the brain, we developed a class of genetically encoded fluorescence sensors based on kappa, delta and mu opioid receptors: κLight, δLight and µLight, respectively. We characterized the pharmacological profiles of these sensors in mammalian cells and in dissociated neurons. We used κLight to identify electrical stimulation parameters that trigger endogenous opioid release and the spatiotemporal scale of dynorphin volume transmission in brain slices. Using in vivo fiber photometry in mice, we demonstrated the utility of these sensors in detecting optogenetically driven opioid release and observed differential opioid release dynamics in response to fearful and rewarding conditions.


Asunto(s)
Técnicas Biosensibles , Optogenética , Animales , Técnicas Biosensibles/métodos , Ratones , Optogenética/métodos , Neuronas/metabolismo , Humanos , Dinorfinas/metabolismo , Dinorfinas/genética , Masculino , Péptidos Opioides/metabolismo , Péptidos Opioides/genética , Células HEK293 , Ratones Endogámicos C57BL , Encéfalo/metabolismo , Neuropéptidos/metabolismo , Neuropéptidos/genética , Receptores Opioides/metabolismo , Receptores Opioides/genética , Estimulación Eléctrica , Recompensa
2.
Pain Res Manag ; 2021: 9981732, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336071

RESUMEN

Objective: To determine behavioral testing after experimental tooth movement in rats and to explore the role of nociceptin/orphanin FQ and its receptor (the N/OFQ-NOP system) in pain induced by experimental tooth movement. Design: The mouth-wiping behavior of rats was assessed by studying behavioral responses after experimental tooth movement. The distribution of N/OFQ in the periodontal ligament, the trigeminal ganglion (TG), and the caudal one-third of the trigeminal subnucleus caudalis (Vc) was assessed by immunohistochemistry. The variations in N/OFQ expression in the TG and Vc were measured by Western blotting. The ongoing changes in the gene expression of the prepronociceptin gene and opioid receptor-like 1 receptor were assessed in the TG and Vc by real-time polymerase chain reaction (RT-PCR). Results: Overall, the mouth-wiping behavior increased significantly. The behavior first increased and then gradually decreased to a low level, showing cyclical variation. N/OFQ immunoreactivity increased in the periodontal ligament after tooth movement. ppN/OFQ mRNA and protein levels showed a time-dependent increase in the TG and were positively correlated with pain stimulus. NOP gene levels showed large fluctuations. In the Vc, the expression and changes in the N/OFQ-NOP system showed the opposite trend as those noted in TG and the periodontal membrane. Conclusion: The N/OFQ system may have a complex regulatory function in the pain induced by tooth movement and may be related to inflammation caused by orthodontic tooth movement and periodontal damage. The specific mechanism remains to be further studied.


Asunto(s)
Receptores Opioides , Técnicas de Movimiento Dental , Animales , Péptidos Opioides/genética , Dolor , Ratas , Receptores Opioides/genética , Nociceptina
3.
Int J Mol Sci ; 22(2)2021 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-33435320

RESUMEN

Nicotine addiction is a severe public health problem. The aim of this study was to investigate the alterations in key neurotransmissions after 60 days of withdrawal from seven weeks of intermittent cigarette smoke, e-cigarette vapours, or an e-cigarette vehicle. In the nicotine withdrawal groups, increased depressive and anxiety/obsessive-compulsive-like behaviours were demonstrated in the tail suspension, sucrose preference and marble burying tests. Cognitive impairments were detected in the spatial object recognition test. A significant increase in Corticotropin-releasing factor (Crf) and Crf1 mRNA levels was observed, specifically after cigarette withdrawal in the caudate-putamen nucleus (CPu). The nociceptin precursor levels were reduced by cigarette (80%) and e-cigarette (50%) withdrawal in the CPu. The delta opioid receptor showed a significant reduction in the hippocampus driven by the exposure to an e-cigarette solubilisation vehicle, while the mRNA levels doubled in the CPu of mice that had been exposed to e-cigarettes. Withdrawal after exposure to e-cigarette vapour induced a 35% Bdnf mRNA decrease in the hippocampus, whereas Bdnf was augmented by 118% by cigarette withdrawal in the CPu. This study shows that long-term withdrawal-induced affective and cognitive symptoms associated to lasting molecular alterations in peptidergic signalling may determine the impaired neuroplasticity in the hippocampal and striatal circuitry.


Asunto(s)
Cigarrillo Electrónico a Vapor/efectos adversos , Hipocampo/efectos de los fármacos , ARN Mensajero/genética , Síndrome de Abstinencia a Sustancias/genética , Contaminación por Humo de Tabaco/efectos adversos , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Núcleo Caudado/efectos de los fármacos , Núcleo Caudado/metabolismo , Núcleo Caudado/fisiopatología , Hormona Liberadora de Corticotropina/genética , Regulación hacia Abajo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Endogámicos BALB C , Péptidos Opioides/genética , Orexinas/genética , Putamen/efectos de los fármacos , Putamen/metabolismo , Putamen/fisiopatología , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores Opioides/genética , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Regulación hacia Arriba/efectos de los fármacos
4.
Int J Mol Sci ; 22(2)2021 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-33429857

RESUMEN

Opioid peptides and their receptors are expressed in the mammalian retina; however, little is known about how they might affect visual processing. The melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs), which mediate important non-image-forming visual processes such as the pupillary light reflex (PLR), express ß-endorphin-preferring, µ-opioid receptors (MORs). The objective of the present study was to elucidate if opioids, endogenous or exogenous, modulate pupillary light reflex (PLR) via MORs expressed by ipRGCs. MOR-selective agonist [D-Ala2, MePhe4, Gly-ol5]-enkephalin (DAMGO) or antagonist D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP) was administered via intravitreal injection. PLR was recorded in response to light stimuli of various intensities. DAMGO eliminated PLR evoked by light with intensities below melanopsin activation threshold but not that evoked by bright blue irradiance that activated melanopsin signaling, although in the latter case, DAMGO markedly slowed pupil constriction. CTAP or genetic ablation of MORs in ipRGCs slightly enhanced dim-light-evoked PLR but not that evoked by a bright blue stimulus. Our results suggest that endogenous opioid signaling in the retina contributes to the regulation of PLR. The slowing of bright light-evoked PLR by DAMGO is consistent with the observation that systemically applied opioids accumulate in the vitreous and that patients receiving chronic opioid treatment have slow PLR.


Asunto(s)
Péptidos Opioides/genética , Receptores Opioides mu/genética , Retina/metabolismo , Percepción Visual/genética , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Encefalinas/antagonistas & inhibidores , Encefalinas/genética , Humanos , Luz , Ratones , Péptidos/farmacología , Receptores Opioides/genética , Receptores Opioides mu/antagonistas & inhibidores , Reflejo/genética , Retina/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/fisiología , Transducción de Señal/efectos de los fármacos , Percepción Visual/efectos de los fármacos , betaendorfina/genética
5.
Mol Pharmacol ; 98(2): 96-108, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32487735

RESUMEN

In the mid-1970s, an intense race to identify endogenous substances that activated the same receptors as opiates resulted in the identification of the first endogenous opioid peptides. Since then, >20 peptides with opioid receptor activity have been discovered, all of which are generated from three precursors, proenkephalin, prodynorphin, and proopiomelanocortin, by sequential proteolytic processing by prohormone convertases and carboxypeptidase E. Each of these peptides binds to all three of the opioid receptor types (µ, δ, or κ), albeit with differing affinities. Peptides derived from proenkephalin and prodynorphin are broadly distributed in the brain, and mRNA encoding all three precursors are highly expressed in some peripheral tissues. Various approaches have been used to explore the functions of the opioid peptides in specific behaviors and brain circuits. These methods include directly administering the peptides ex vivo (i.e., to excised tissue) or in vivo (in animals), using antagonists of opioid receptors to infer endogenous peptide activity, and genetic knockout of opioid peptide precursors. Collectively, these studies add to our current understanding of the function of endogenous opioids, especially when similar results are found using different approaches. We briefly review the history of identification of opioid peptides, highlight the major findings, address several myths that are widely accepted but not supported by recent data, and discuss unanswered questions and future directions for research. SIGNIFICANCE STATEMENT: Activation of the opioid receptors by opiates and synthetic drugs leads to central and peripheral biological effects, including analgesia and respiratory depression, but these may not be the primary functions of the endogenous opioid peptides. Instead, the opioid peptides play complex and overlapping roles in a variety of systems, including reward pathways, and an important direction for research is the delineation of the role of individual peptides.


Asunto(s)
Péptidos Opioides/genética , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Animales , Encéfalo/metabolismo , Carboxipeptidasa H/metabolismo , Encefalinas/química , Encefalinas/genética , Humanos , Proopiomelanocortina/química , Proopiomelanocortina/genética , Proproteína Convertasas/metabolismo , Precursores de Proteínas/química , Precursores de Proteínas/genética
6.
Peptides ; 128: 170307, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32217145

RESUMEN

Opioid peptides, derived from PENK, POMC, PDYN and PNOC precursors, together with their receptors (DOR, MOR, KOR and ORL1), constitute the opioid system and are suggested to participate in multiple physiological/pathological processes in vertebrates. However, the question whether an opioid system exists and functions in non-mammalian vertebrates including birds remains largely unknown. Here, we cloned genes encoding opioid system from the chicken brain and examined their functionality and tissue expression. As in mammals, 6 opioid peptides encoded by PENK (Met-enkephalin and Leu-enkephalin), POMC (ß-endorphin), PDYN (dynorphin-A and dynorphin-B) and PNOC (nociceptin) precursors and four opioid receptors were found to be highly conserved in chickens. Using pGL3-CRE-luciferase and pGL4-SRE-luciferase reporter systems, we demonstrated that chicken opioid receptors (cDOR, cMOR, cKOR and cORL1) expressed in CHO cells, could be differentially activated by chicken opioid peptides, and resulted in the inhibition of cAMP/PKA and activation of MAPK/ERK signaling pathways. cDOR is potently activated by Met-enkephalin and Leu-enkephalin, and cKOR is potently activated by dynorphin-A, dynorphin-B and nociceptin, whereas cORL1 is specifically activated by nociceptin. Unlike cDOR, cKOR and cORL1, cMOR is moderately/weakly activated by enkephalins and other opioid peptides. These findings suggest the ligand-receptor pair in chicken opioid system is similar, but not identical to, that in mammals. Quantitative real-time PCR revealed that the opioid system is mainly expressed in chicken central nervous system including the hypothalamus. Collectively, our data will help to facilitate the better understanding of the conserved roles of opioid system across vertebrates.


Asunto(s)
Encéfalo/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Sistema de Señalización de MAP Quinasas , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Pollos , Clonación Molecular/métodos , Cricetinae , Proteínas Quinasas Dependientes de AMP Cíclico/genética , ADN Complementario/genética , Péptidos Opioides/química , Péptidos Opioides/genética , Receptores Opioides/química , Receptores Opioides/genética , Homología de Secuencia , Distribución Tisular
7.
Int J Mol Sci ; 20(20)2019 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-31635381

RESUMEN

A wide variety of peptides not only interact with the cell surface, but govern complex signaling from inside the cell. This has been referred to as an "intracrine" action, and the orchestrating molecules as "intracrines". Here, we review the intracrine action of dynorphin B, a bioactive end-product of the prodynorphin gene, on nuclear opioid receptors and nuclear protein kinase C signaling to stimulate the transcription of a gene program of cardiogenesis. The ability of intracrine dynorphin B to prime the transcription of its own coding gene in isolated nuclei is discussed as a feed-forward loop of gene expression amplification and synchronization. We describe the role of hyaluronan mixed esters of butyric and retinoic acids as synthetic intracrines, controlling prodynorphin gene expression, cardiogenesis, and cardiac repair. We also discuss the increase in prodynorphin gene transcription and intracellular dynorphin B afforded by electromagnetic fields in stem cells, as a mechanism of cardiogenic signaling and enhancement in the yield of stem cell-derived cardiomyocytes. We underline the possibility of using the diffusive features of physical energies to modulate intracrinergic systems without the needs of viral vector-mediated gene transfer technologies, and prompt the exploration of this hypothesis in the near future.


Asunto(s)
Diferenciación Celular/genética , Encefalinas/genética , Encefalinas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Animales , Butiratos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos Opioides/genética , Péptidos Opioides/metabolismo , Organogénesis/genética , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Tretinoina/metabolismo
8.
Vitam Horm ; 111: 1-16, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31421696

RESUMEN

Proopiomelanocortin (POMC) belongs to the opioid/orphanin gene family whose peptide precursors include either opioid (YGGF) or the orphanin/nociceptin core sequences (FGGF). In addition to POMC the family includes the proenkephalin (PENK), prodynorphin (PDYN), and nociceptin/proorphanin (PNOC) precursors. The opioid core sequence in POMC is incorporated by the ß-endorphin that occupies the C-terminal region but this propeptide also exhibits at least two "alien" melanocortin core sequences (HFRW). An ACTH/MSH fragment merged into the opioid fragment not earlier than the two tetraploidizations of the vertebrate genome. Therefore, POMC exhibit a complex "evolutionary life" since the gene has coevolved together with two different receptor systems, i.e., opioid and melanocortin following a horse trading system. In this article, we summarize the different evolutionary hypotheses proposed for POMC evolution.


Asunto(s)
Evolución Molecular , Proopiomelanocortina/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Humanos , Melanocortinas/química , Melanocortinas/genética , Hormonas Estimuladoras de los Melanocitos , Péptidos Opioides/genética , Filogenia , Proopiomelanocortina/química , Nociceptina
9.
Genes Brain Behav ; 18(8): e12595, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31216095

RESUMEN

Hypothalamic histaminergic neurons regulate a variety of homeostatic, metabolic and cognitive functions. Recent data have suggested a modulatory role of histamine and histamine receptors in shaping striatal activity and connected the histaminergic system to neuropsychiatric disorders. We characterized exploratory behavior and striatal neurotransmission in mice lacking the histamine producing enzyme histidine decarboxylase (Hdc). The mutant mice showed a distinct behavioral pattern during exploration of novel environment, specifically, increased frequency of rearing seated against the wall, jumping and head/body shakes. This behavioral phenotype was associated with decreased levels of striatal dopamine and serotonin and increased level of dopamine metabolite DOPAC. Gene expression levels of dynorphin and enkephalin, opioids released by medium spiny neurons of striatal direct and indirect pathways respectively, were lower in Hdc mutant mice than in control animals. A low dose of amphetamine led to similar behavioral and biochemical outcomes in both genotypes. Increased striatal dopamine turnover was observed in Hdc KO mice after treatment with dopamine precursor l-Dopa. Overall, our study suggests a role for striatal dopamine and opioid peptides in formation of distinct behavioral phenotype of Hdc KO mice.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Histamina/metabolismo , Histidina Descarboxilasa/genética , Movimiento , Péptidos Opioides/genética , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Cuerpo Estriado/fisiopatología , Histamina/deficiencia , Histidina Descarboxilasa/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos Opioides/metabolismo , Serotonina/metabolismo
10.
Peptides ; 116: 42-62, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31047940

RESUMEN

This review is part of a special issue dedicated to Opioid addiction, and examines the influential role of opioid peptides, opioid receptors and opiate drugs in mediating food intake and body weight control in rodents. This review postulates that opioid mediation of food intake was an example of "positive addictive" properties that provide motivational drives to maintain opioid-seeking behavior and that are not subject to the "negative addictive" properties associated with tolerance, dependence and withdrawal. Data demonstrate that opiate and opioid peptide agonists stimulate food intake through homeostatic activation of sensory, metabolic and energy-related In contrast, general, and particularly mu-selective, opioid receptor antagonists typically block these homeostatically-driven ingestive behaviors. Intake of palatable and hedonic food stimuli is inhibited by general, and particularly mu-selective, opioid receptor antagonists. The selectivity of specific opioid agonists to elicit food intake was confirmed through the use of opioid receptor antagonists and molecular knockdown (antisense) techniques incapacitating specific exons of opioid receptor genes. Further extensive evidence demonstrated that homeostatic and hedonic ingestive situations correspondingly altered the levels and expression of opioid peptides and opioid receptors. Opioid mediation of food intake was controlled by a distributed brain network intimately related to both the appetitive-consummatory sites implicated in food intake as well as sites intimately involved in reward and reinforcement. This emergent system appears to sustain the "positive addictive" properties providing motivational drives to maintain opioid-seeking behavior.


Asunto(s)
Ingestión de Alimentos/genética , Antagonistas de Narcóticos/uso terapéutico , Péptidos Opioides/genética , Receptores Opioides/genética , Animales , Peso Corporal/genética , Encéfalo/metabolismo , Conducta Alimentaria/efectos de los fármacos , Humanos , Motivación/genética
11.
Drug Alcohol Depend ; 197: 127-133, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30818133

RESUMEN

BACKGROUND: Early-life stressful events affect the neurobiological maturation of cerebral circuitries including the endogenous opioid system and the effects elicited by adolescent cocaine exposure on this system have been poorly investigated. Here, we evaluated whether cocaine exposure during adolescence causes short- or long-term alterations in mRNAs codifying for selected elements belonging to the opioid system. Moreover, since brain-derived neurotrophic factor (BDNF) may undergo simultaneous alterations with the opioid peptide dynorphin, we also evaluated its signaling pathway as well. METHODS: Adolescent male rats were exposed to cocaine (20 mg/kg/day) from post-natal day (PND) 28 to PND42, approximately corresponding to human adolescence. After short- (PND45) or long-term (PND90) abstinence, prodynorphin-κ-opioid receptor (pDYN-KOP) and pronociceptin-nociceptin receptor (pN/OFQ-NOP) gene expression were evaluated in the nucleus accumbens (NAc) and hippocampus (Hip) together with the analysis of BDNF signaling pathways. RESULTS: In the NAc of PND45 rats, pDYN mRNA levels were up-regulated, an effect paralled by increased BDNF signaling. Differently from NAc, pDYN mRNA levels were down-regulated in the Hip of PND45 rats without significant changes of BDNF pathway. At variance from PND45 rats, we did not find any significant alteration of the investigated parameters either in NAc and Hip of PND90 rats. CONCLUSIONS: Our results indicate that the short-term withdrawal from adolescent cocaine exposure is characterized by a parallel pDYN mRNA and BDNF signaling increase in the NAc. Given the depressive-like state experienced during short abstinence in humans, we hypothesize that such changes may contribute to promote the risk of cocaine abuse escalation and relapse.


Asunto(s)
Trastornos Relacionados con Cocaína/genética , Dinorfinas/genética , Núcleo Accumbens/metabolismo , ARN Mensajero/metabolismo , Síndrome de Abstinencia a Sustancias/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Encefalinas/metabolismo , Expresión Génica , Masculino , Núcleo Accumbens/efectos de los fármacos , Péptidos Opioides/genética , Precursores de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides , Receptores Opioides kappa/genética , Transducción de Señal , Receptor de Nociceptina
12.
Mol Pain ; 15: 1744806919828921, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30665329

RESUMEN

Little is known about the mechanisms involved in the regulation of nociceptin and its receptor (nociceptin opioid peptide receptor, NOP) in response to inflammation and pain in humans. In this study, specific signaling pathways contributing to the regulation of nociceptin and NOP in human peripheral blood leukocytes were investigated. After approval by the ethics committee, peripheral blood obtained from healthy donors was cultured with or without phorbol-12-myristate-13-acetate (PMA). Prepronociceptin (ppNOC) and NOP mRNA were analyzed by real-time quantitative polymerase chain reaction, and nociceptin concentrations in culture supernatants by fluorescent enzyme immunoassay. Nociceptin and NOP protein levels in blood leukocyte subsets were determined using flow cytometry. To examine the contribution of signaling pathways to ppNOC and NOP regulation, blood was pre-treated with kinase inhibitors specific for ERK, JNK, p38, and NFκB pathways prior to culturing with or without PMA. PMA dose-dependently upregulated ppNOC mRNA but downregulated NOP mRNA in human peripheral blood leukocytes. PMA 10 ng/ml increased ppNOC after 6 h and suppressed NOP after 3 h compared to controls (both P <0.005). Nociceptin concentrations were increased in supernatants of PMA-induced blood samples after 24 h ( P <0.005), whereas expression of cell-membrane NOP was decreased by PMA in blood leukocyte subsets (all P <0.05). Blockade of ERK or p38 pathways partially prevented PMA effects on ppNOC and NOP mRNA (all P <0.05). The combination of ERK and p38 inhibitors completely reversed the effects of PMA ( P <0.05). ERK and p38 are two major signaling pathways regulating nociceptin and its receptor in human peripheral blood leukocytes under inflammatory conditions.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/fisiología , Leucocitos/metabolismo , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adolescente , Adulto , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Leucocitos/efectos de los fármacos , Persona de Mediana Edad , Péptidos Opioides/genética , Ésteres del Forbol/farmacología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores Opioides/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Tiempo , Adulto Joven , Receptor de Nociceptina , Nociceptina
13.
Mol Cell Proteomics ; 17(9): 1737-1749, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29895708

RESUMEN

The cerebellum is a brain structure involved in motor and cognitive functions. The development of the cerebellar cortex (the external part of the cerebellum) is under the control of numerous factors. Among these factors, neuropeptides including PACAP or somatostatin modulate the survival, migration and/or differentiation of cerebellar granule cells. Interestingly, such peptides contributing to cerebellar ontogenesis usually exhibit a specific transient expression profile with a low abundance at birth, a high expression level during the developmental processes, which take place within the first two postnatal weeks in rodents, and a gradual decline toward adulthood. Thus, to identify new peptides transiently expressed in the cerebellum during development, rat cerebella were sampled from birth to adulthood, and analyzed by a semi-quantitative peptidomic approach. A total of 33 peptides were found to be expressed in the cerebellum. Among these 33 peptides, 8 had a clear differential expression pattern during development, 4 of them i.e. cerebellin 2, nociceptin, somatostatin and VGF [353-372], exhibiting a high expression level during the first two postnatal weeks followed by a significative decrease at adulthood. A focus by a genomic approach on nociceptin, confirmed that its precursor mRNA is transiently expressed during the first week of life in granule neurons within the internal granule cell layer of the cerebellum, and showed that the nociceptin receptor is also actively expressed between P8 and P16 by the same neurons. Finally, functional studies revealed a new role for nociceptin, acting as a neurotrophic peptide able to promote the survival and differentiation of developing cerebellar granule neurons.


Asunto(s)
Corteza Cerebelosa/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Péptidos Opioides/metabolismo , Péptidos/metabolismo , Proteómica/métodos , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cerebelo/metabolismo , Femenino , Peróxido de Hidrógeno/toxicidad , Masculino , Factores de Crecimiento Nervioso/química , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Péptidos Opioides/química , Péptidos Opioides/genética , Péptidos/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Receptores Opioides/metabolismo , Receptor de Nociceptina , Nociceptina
14.
Brain Res ; 1695: 78-83, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29852138

RESUMEN

The endogenous opioid system (EOS) controls the processing of nociceptive stimuli and is a pharmacological target for opioids. Alterations in expression of the EOS genes under neuropathic pain condition may account for low efficacy of opioid drugs. We here examined whether EOS expression patterns are altered in the lumbar spinal cord of the rats with spinal nerve ligation (SNL) as a neuropathic pain model. Effects of the left- and right-side SNL on expression of EOS genes in the ipsi- and contralateral spinal domains were analysed. The SNL-induced changes were complex and different between the genes; between the dorsal and ventral spinal domains; and between the left and right sides of the spinal cord. Prodynorphin (Pdyn) expression was upregulated in the ipsilateral dorsal domains by each the left and right-side SNL, while changes in expression of µ-opioid receptor (Oprm1) and proenkephalin (Penk) genes were dependent on the SNL side. Changes in expression of the Pdyn and κ-opioid receptor (Oprk1) genes were coordinated between the ipsi- and contralateral sides. Withdrawal response thresholds, indicators of mechanical allodynia correlated negatively with Pdyn expression in the right ventral domain after right side SNL. These findings suggest multiple roles of the EOS gene products in spinal sensitization and changes in motor reflexes, which may differ between the left and right sides.


Asunto(s)
Analgésicos Opioides/farmacología , Expresión Génica/efectos de los fármacos , Neuralgia/tratamiento farmacológico , Péptidos Opioides/genética , Médula Espinal/efectos de los fármacos , Animales , Expresión Génica/genética , Neuralgia/metabolismo , Péptidos Opioides/metabolismo , Umbral del Dolor/efectos de los fármacos , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo , Médula Espinal/metabolismo , Nervios Espinales/metabolismo
15.
Peptides ; 99: 205-216, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29038035

RESUMEN

In an attempt to design opioid-nociceptin hybrid peptides, three novel bivalent ligands, H-YGGFGGGRYYRIK-NH2, H-YGGFRYYRIK-NH2 and Ac-RYYRIKGGGYGGFL-OH were synthesized and studied by biochemical, pharmacological, biophysical and molecular modelling tools. These chimeric molecules consist of YGGF sequence, a crucial motif in the N-terminus of natural opioid peptides, and Ac-RYYRIK-NH2, which was isolated from a combinatorial peptide library as an antagonist or partial agonist that inhibits the biological activity of the endogenously occurring heptadecapeptide nociceptin. Solution structures for the peptides were studied by analysing their circular dichroism spectra. Receptor binding affinities were measured by equilibrium competition experiments using four highly selective radioligands. G-protein activating properties of the multitarget peptides were estimated in [35S]GTPγS binding tests. The three compounds were also measured in electrically stimulated mouse vas deferens (MVD) bioassay. H-YGGFGGGRYYRIK-NH2 (BA55), carrying N-terminal opioid and C-terminal nociceptin-like sequences interconnected with GGG tripeptide spacer displayed a tendency of having either unordered or ß-sheet structures, was moderately potent in MVD and possessed a NOP/KOP receptor preference. A similar peptide without spacer H-YGGFRYYRIK-NH2 (BA62) exhibited the weakest effect in MVD, more α-helical periodicity was present in its structure and it exhibited the most efficacious agonist actions in the G-protein stimulation assays. The third hybrid peptide Ac-RYYRIKGGGYGGFL-OH (BA61) unexpectedly displayed opioid receptor affinities, because the opioid message motif is hidden within the C-terminus. The designed chimeric peptide ligands presented in this study accommodate well into a group of multitarget opioid compounds that include opioid-non-opioid peptide dimer analogues, dual non-peptide dimers and mixed peptide- non-peptide bifunctional ligands.


Asunto(s)
Modelos Moleculares , Péptidos Opioides , Ingeniería de Proteínas/métodos , Receptores Acoplados a Proteínas G/agonistas , Animales , Femenino , Masculino , Ratones , Péptidos Opioides/química , Péptidos Opioides/genética , Péptidos Opioides/farmacología , Estructura Secundaria de Proteína , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Nociceptina
16.
Eur Neuropsychopharmacol ; 27(12): 1298-1307, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29102248

RESUMEN

The endogenous neuropeptide nociceptin (N/OFQ), which mediates its actions via the nociceptin receptor (NOP), is implicated in multiple behavioural and physiological functions. This study examined the effects of the NOP agonists N/OFQ and the synthetic agonist Ro 64-6198, the antagonists NNN and NalBzoH, as well as deletion of the Pronociceptin gene on emotional memory in mice. The animals were tested in the passive avoidance (PA) task, dependent on hippocampal and amygdala functions. N/OFQ injected intraventricularly (i.c.v.) prior to training produced a biphasic effect on PA retention; facilitation at a low dose and impairment at higher doses. Ro 64-6198 also displayed a biphasic effect with memory facilitation at lower doses and impairment at a high dose. None of the agonists influenced PA training latencies. NNN did not significantly modulate retention in the PA task but antagonized the inhibitory effects of N/OFQ. NalBzoH facilitated memory retention in a dose-dependent manner and blocked the impairing effects of N/OFQ. However, neither NNN nor NalBzoH blocked the memory-impairing effects of Ro 64-6198. Finally, the Pnoc knockout mice exhibited enhanced PA retention latencies compared to the wild type mice. The biphasic effect of the natural ligand and Ro 64-6198 and the failure of the antagonists to block the action of Ro 64-6198 indicate complexity in ligand-receptor interaction. These results indicate that brain nociceptin and its NOP has a subtle role in regulation of mechanisms of relevance for treatment of disorders with processing disturbances of aversive events e.g. Alzheimer's disease, anxiety, depression and PTSD.


Asunto(s)
Reacción de Prevención/fisiología , Péptidos Opioides/metabolismo , Receptores Opioides/deficiencia , Animales , Aprendizaje por Asociación/efectos de los fármacos , Reacción de Prevención/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Imidazoles/farmacología , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Naloxona/análogos & derivados , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/genética , Péptidos Opioides/farmacología , Fragmentos de Péptidos/farmacología , Receptores Opioides/agonistas , Receptores Opioides/genética , Retención en Psicología/efectos de los fármacos , Compuestos de Espiro/farmacología , Receptor de Nociceptina , Nociceptina
17.
Peptides ; 95: 116-123, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28782637

RESUMEN

Morphiceptin (Tyr-Pro-Phe-Pro-NH2) is a selective ligand of the mu opioid receptor, an important target in pain regulation. In this study, morphiceptin was modified at positions 2 or 3 by introduction of ß2- or ß3-amino acids and additionally in position 1 by replacing Tyr by Dmt (2',6'-dimethyltyrosine), which resulted in obtaining enzymatically stable analogs with mixed opioid receptor affinity profiles. An analog of the sequence Dmt-d-Ala-(R)-ß2-1-Nal-Pro-NH2 [Nal=3-(1-naphthyl)-alanine] showed very high activity at the mu and delta receptors in the calcium mobilization functional test but did not cross the artificial membrane imitating the blood-brain barrier. In the in vivo test this analog induced strong antinociceptive effect in the writhing test in mice after intraperitioneal but also oral administration and inhibited diarrhea similarly to loperamide. Therefore, it may become an interesting lead compound in the development of peripherally restricted drugs for the treatment of gastrointestinal disorders.


Asunto(s)
Endorfinas/química , Péptidos Opioides/genética , Dolor/tratamiento farmacológico , Peptidomiméticos/uso terapéutico , Secuencia de Aminoácidos/genética , Analgésicos/química , Analgésicos/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Endorfinas/genética , Endorfinas/uso terapéutico , Humanos , Ratones , Péptidos Opioides/química , Péptidos Opioides/uso terapéutico , Dolor/genética , Peptidomiméticos/química , Receptores Opioides mu/química , Receptores Opioides mu/genética
18.
Brain Res ; 1668: 36-45, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28511993

RESUMEN

The young brain is highly sensitive to environmental influences that can cause long-term changes in neuronal function, possibly through altered gene expression. The endogenous opioid system continues to mature after birth and because of its involvement in reward, an inadequate maturation of this system could lead to enhanced susceptibility for alcohol use disorder. Recent studies show that the classical reward areas nucleus accumbens and ventral tegmental area are less affected by early life stress whereas endogenous opioids in non-classical areas, e.g. dorsal striatum and amygdala, are highly responsive. The aim was to investigate the interaction between early life conditions and adult voluntary ethanol intake on opioid gene expression. Male Wistar rats were exposed to conventional rearing, 15, or 360min of daily maternal separation (MS) postnatal day 1-21, and randomly assigned to ethanol or water drinking postnatal week 10-16. Rats exposed to early life stress (MS360) had increased opioid receptor gene (Oprm1, Oprd1 and Oprk1) expression in the dorsal striatum. Ethanol drinking was associated with lower striatal Oprd1 and Oprk1 expression solely in rats exposed to early life stress. Furthermore, rats exposed to early life stress had high inherent Pomc expression in the amygdala but low expression after ethanol intake. Thus, adverse events early in life induced changes in opioid gene expression and also influenced the central molecular response to ethanol intake. These long-term consequences of early life stress can contribute to the enhanced risk for excessive ethanol intake and alcohol use disorder seen after exposure to childhood adversity.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Alcoholismo/genética , Etanol/efectos adversos , Péptidos Opioides/genética , Receptores Opioides/genética , Alcoholismo/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Femenino , Masculino , Privación Materna , Péptidos Opioides/metabolismo , Ratas Wistar , Receptores Opioides/metabolismo , Recompensa , Tiempo
19.
Psychopharmacology (Berl) ; 234(9-10): 1603-1614, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28280884

RESUMEN

RATIONALE: Mood disorders can be triggered by stress and are characterized by deficits in reward processing, including disrupted reward learning (the ability to modulate behavior according to past rewards). Reward learning is regulated by the anterior cingulate cortex (ACC) and striatal circuits, both of which are implicated in the pathophysiology of mood disorders. OBJECTIVES: Here, we assessed in rats the effects of a potent stressor (social defeat) on reward learning and gene expression in the ACC, ventral tegmental area (VTA), and striatum. METHODS: Adult male Wistar rats were trained on an operant probabilistic reward task (PRT) and then exposed to 3 days of social defeat before assessment of reward learning. After testing, the ACC, VTA, and striatum were dissected, and expression of genes previously implicated in stress was assessed. RESULT: Social defeat blunted reward learning (manifested as reduced response bias toward a more frequently rewarded stimulus) and was associated with increased nociceptin/orphanin FQ (N/OFQ) peptide mRNA levels in the striatum and decreased Fos mRNA levels in the VTA. Moreover, N/OFQ peptide and nociceptin receptor mRNA levels in the ACC, VTA and striatum were inversely related to reward learning. CONCLUSIONS: The behavioral findings parallel previous data in humans, suggesting that stress similarly disrupts reward learning in both species. Increased striatal N/OFQ mRNA in stressed rats characterized by impaired reward learning is consistent with accumulating evidence that antagonism of nociceptin receptors, which bind N/OFQ, has antidepressant-like effects. These results raise the possibility that nociceptin systems represent a molecular substrate through which stress produces reward learning deficits in mood disorders.


Asunto(s)
Cuerpo Estriado/metabolismo , Aprendizaje/fisiología , Péptidos Opioides/biosíntesis , ARN Mensajero/biosíntesis , Recompensa , Estrés Psicológico/metabolismo , Animales , Femenino , Humanos , Relaciones Interpersonales , Masculino , Trastornos del Humor/genética , Trastornos del Humor/metabolismo , Trastornos del Humor/psicología , Péptidos Opioides/genética , ARN Mensajero/genética , Ratas , Ratas Long-Evans , Ratas Wistar , Estrés Psicológico/psicología , Área Tegmental Ventral/metabolismo , Nociceptina
20.
Genes Brain Behav ; 16(5): 537-545, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28000999

RESUMEN

Several studies showed that chronic pain causes reorganization and functional alterations of supraspinal brain regions. The nociceptin-NOP receptor system is one of the major systems involved in pain control and much evidence also suggested its implication in stress, anxiety and depression. Therefore, we investigated the nociceptin-NOP system alterations in selected brain regions in a neuropathic pain murine model. Fourteen days after the common sciatic nerve ligature, polymerase chain reaction (PCR) analysis indicated a significant decrease of pronociceptin and NOP receptor mRNA levels in the thalamus; these alterations could contribute to the decrease of the thalamic inhibitory function reported in neuropathic pain condition. Nociceptin peptide and NOP mRNA increased in the anterior cingulate cortex (ACC) and not in the somatosensory cortex, suggesting a peculiar involvement of this system in pain regulating circuitry. Similarly to the ACC, an increase of nociceptin peptide levels was observed in the amygdala. Finally, the pronociceptin and NOP mRNAs decrease observed in the hypothalamus reflects the lack of hypothalamus-pituitary-adrenal axis activation, already reported in neuropathic pain models. Our data indicate that neuropathic pain conditions affect the supraspinal nociceptin-NOP system which is also altered in regions known to play a role in emotional aspects of pain.


Asunto(s)
Giro del Cíngulo/metabolismo , Neuralgia/metabolismo , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Nervio Ciático/lesiones , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiología , Animales , Giro del Cíngulo/fisiología , Masculino , Ratones , Neuralgia/fisiopatología , Péptidos Opioides/genética , Receptores Opioides/genética , Corteza Somatosensorial/metabolismo , Corteza Somatosensorial/fisiología , Tálamo/metabolismo , Tálamo/fisiología , Receptor de Nociceptina , Nociceptina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA