Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Mol Cell Neurosci ; 85: 93-104, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28942046

RESUMEN

Dopamine signaling encodes reward learning and motivated behavior through modulation of synaptic signaling in the nucleus accumbens, and aberrations in these processes are thought to underlie obsessive behaviors associated with alcohol abuse. The nucleus accumbens is divided into core and shell sub-regions with overlapping but also divergent contributions to behavior. Here we optogenetically targeted dopamine projections to the accumbens allowing us to isolate stimulation of dopamine terminals ex vivo. We applied 5 pulse (phasic) light stimulations to probe intrinsic differences in dopamine release parameters across regions. Also, we exposed animals to 4weeks of chronic intermittent ethanol vapor and measured phasic release. We found that initial release probability, uptake rate and autoreceptor inhibition were greater in the accumbens core compared to the shell, yet the shell showed greater phasic release ratios. Following chronic ethanol, uptake rates were increased in the core but not the shell, suggesting region-specific neuronal adaptations. Conversely, kappa opioid receptor function was upregulated in both regions to a similar extent, suggesting a local mechanism of kappa opioid receptor regulation that is generalized across the nucleus accumbens. These data suggest that dopamine axons in the nucleus accumbens core and shell display differences in intrinsic release parameters, and that ethanol-induced adaptations to dopamine neuron terminal fields may not be homogeneous. Also, chronic ethanol exposure induces an upregulation in kappa opioid receptor function, providing a mechanism for potential over-inhibition of accumbens dopamine signaling which may negatively impact downstream synaptic function and ultimately bias choice towards previously reinforced alcohol use behaviors.


Asunto(s)
Alcoholismo/metabolismo , Depresores del Sistema Nervioso Central/toxicidad , Dopamina/metabolismo , Etanol/toxicidad , Núcleo Accumbens/metabolismo , Animales , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/efectos de los fármacos , Optogenética , Técnicas de Cultivo de Órganos , Receptores Opioides kappa/biosíntesis , Transmisión Sináptica/efectos de los fármacos
2.
Alcohol Clin Exp Res ; 41(8): 1402-1418, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28425121

RESUMEN

Evidence has demonstrated that dynorphin (DYN) and the kappa opioid receptor (KOR) system contribute to various psychiatric disorders, including anxiety, depression, and addiction. More recently, this endogenous opioid system has received increased attention as a potential therapeutic target for treating alcohol use disorders. In this review, we provide an overview and synthesis of preclinical studies examining the influence of alcohol (ethanol [EtOH]) exposure on DYN/KOR expression and function, as well as studies examining the effects of DYN/KOR manipulation on EtOH's rewarding and aversive properties. We then describe work that has characterized effects of KOR activation and blockade on EtOH self-administration and EtOH dependence/withdrawal-related behaviors. Finally, we address how the DYN/KOR system may contribute to stress-EtOH interactions. Despite an apparent role for the DYN/KOR system in motivational effects of EtOH, support comes from relatively few studies. Nevertheless, review of this literature reveals several common themes: (i) rodent strains genetically predisposed to consume more EtOH generally appear to have reduced DYN/KOR tone in brain reward circuitry; (ii) acute and chronic EtOH exposure typically up-regulate the DYN/KOR system; (iii) KOR antagonists reduce behavioral indices of negative affect associated with stress and chronic EtOH exposure/withdrawal; and (iv) KOR antagonists are effective in reducing EtOH consumption, but are often more efficacious under conditions that engender high levels of consumption, such as dependence or stress exposure. These results support the contention that the DYN/KOR system plays a significant role in contributing to dependence- and stress-induced elevation in EtOH consumption. Overall, more comprehensive analyses (on both behavioral and mechanistic levels) are needed to provide additional insight into how the DYN/KOR system is engaged and adapts to influence the motivation effects of EtOH. This information will be critical for the development of new pharmacological agents targeting KORs as promising novel therapeutics for alcohol use disorders and comorbid affective disorders.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Dinorfinas/biosíntesis , Etanol/administración & dosificación , Motivación/efectos de los fármacos , Receptores Opioides kappa/biosíntesis , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/psicología , Animales , Dinorfinas/genética , Humanos , Motivación/fisiología , Receptores Opioides kappa/genética
3.
Genesis ; 54(1): 29-37, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26575788

RESUMEN

The kappa opioid receptor (KOR) has numerous important roles in the nervous system including the modulation of mood, reward, pain, and itch. In addition, KOR is expressed in many non-neuronal tissues. However, the specific cell types that express KOR are poorly characterized. Here, we report the development of a KOR-Cre knockin allele, which provides genetic access to cells that express KOR. In this mouse, Cre recombinase (Cre) replaces the initial coding sequence of the Opkr1 gene (encoding the kappa opioid receptor). We demonstrate that the KOR-Cre allele mediates recombination by embryonic day 14.5 (E14.5). Within the brain, KOR-Cre shows expression in numerous areas including the cerebral cortex, nucleus accumbens and striatum. In addition, this allele is expressed in epithelium and throughout many regions of the body including the heart, lung, and liver. Finally, we reveal that KOR-Cre mediates recombination of a subset of bipolar and amacrine cells in the retina. Thus, the KOR-Cre mouse line is a valuable new tool for conditional gene manipulation to enable the study of KOR.


Asunto(s)
Técnicas de Transferencia de Gen , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Animales , Encéfalo/metabolismo , Femenino , Integrasas/química , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Receptores Opioides kappa/biosíntesis , Transducción de Señal
4.
J Cell Mol Med ; 19(11): 2587-96, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26153065

RESUMEN

Buprenorphine, a maintenance drug for heroin addicts, exerts its pharmacological function via κ- (KOP), µ-opioid (MOP) and nociceptin/opioid receptor-like 1 (NOP) receptors. Previously, we investigated its effects in an in vitro model expressing human MOP and NOP receptors individually or simultaneously (MOP, NOP, and MOP+NOP) in human embryonic kidney 293 cells. Here, we expanded this cell model by expressing human KOP, MOP and NOP receptors individually or simultaneously (KOP, KOP+MOP, KOP+NOP and KOP+MOP+NOP). Radioligand binding with tritium-labelled diprenorphine confirmed the expression of KOP receptors. Immunoblotting and immunocytochemistry indicated that the expressed KOP, MOP and NOP receptors are N-linked glycoproteins and colocalized in cytoplasmic compartments. Acute application of the opioid receptor agonists- U-69593, DAMGO and nociceptin- inhibited adenylate cyclase (AC) activity in cells expressing KOP, MOP and NOP receptors respectively. Buprenorphine, when applied acutely, inhibited AC activity to ~90% in cells expressing KOP+MOP+NOP receptors. Chronic exposure to buprenorphine induced concentration-dependent AC superactivation in cells expressing KOP+NOP receptors, and the level of this superactivation was even higher in KOP+MOP+NOP-expressing cells. Our study demonstrated that MOP receptor could enhance AC regulation in the presence of coexpressed KOP and NOP receptors, and NOP receptor is essential for concentration-dependent AC superactivation elicited by chronic buprenorphine exposure.


Asunto(s)
Adenilil Ciclasas/metabolismo , Analgésicos Opioides/farmacología , Buprenorfina/farmacología , Receptores Opioides kappa/biosíntesis , Receptores Opioides mu/biosíntesis , Inhibidores de Adenilato Ciclasa/farmacología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Células HEK293 , Humanos , Técnicas Inmunológicas
5.
J Recept Signal Transduct Res ; 34(1): 44-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24219160

RESUMEN

CONTEXT: Our previous study demonstrated that acute and repeated morphine treatment differentially regulated κ-opioid receptor mRNA levels in the rat mesocorticolimbic system. Here, we further investigated the effects of morphine on protein levels of κ-opioid receptor in this reward-related circuitry. METHODS: Three groups of rats received intraperitoneal injection of saline, acute morphine (8.0 mg/kg) and repeated morphine (8.0 mg/kg, once daily for 5 consecutive days) and the κ-receptor protein expression was examined by Western blot analysis. RESULTS: We found that acute morphine treatment did not affect the κ-receptor protein levels in medial prefrontal cortex (mPFC), nucleus accumbens (NAc) and ventral tegmental area (VTA). However, repeated morphine treatment downregulated the κ-receptor protein levels in mPFC and VTA, while there was no significant change in NAc. CONCLUSIONS: These results, along with those reported previously, suggested that morphine dependence may be associated with regionally specific changes in κ-opioid receptor expression in mesocorticolimbic system.


Asunto(s)
Morfina/administración & dosificación , ARN Mensajero/efectos de los fármacos , Receptores Opioides kappa/biosíntesis , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Corteza Prefrontal/efectos de los fármacos , Ratas , Área Tegmental Ventral/efectos de los fármacos
6.
Alcohol Clin Exp Res ; 38(6): 1630-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24796820

RESUMEN

BACKGROUND: Approximately 10 to 15% of women consume alcohol (ethanol [EtOH]) during pregnancy in the United States. Even low amounts of EtOH consumption during pregnancy can elicit long-term consequences. Prenatal experience with as few as 3 drinks has been associated with increase problem drinking in adulthood. Such effects are corroborated in rodents; however, the underlying neural adaptations contributing to this effect are not clear. In the current set of experiments, we investigated whether changes in EtOH responding following prenatal EtOH exposure involved kappa opioid receptor activation and expression. METHODS: Sprague-Dawley rats were prenatally exposed to low levels of alcohol (1.0 g/kg) during late gestation (gestational days 17 to 20 [GD17-20]) via intragastric intubation of pregnant dams. Following birth, EtOH intake, kappa- and mu-opioid-induced place conditioning, and kappa opioid receptor expression in mesolimbic brain regions were assessed in infant rats (postnatal days 14 to 15 [PD14-15]) that were offspring of dams given EtOH, vehicle, or untreated, during pregnancy. RESULTS: Animals exposed to prenatal alcohol drank more alcohol later in life and exhibited significant changes in the kappa opioid system. While control subjects found kappa opioid activation aversive, animals exposed to EtOH prenatally exhibited either no aversion or appetitive responding. Further analysis revealed that synaptosomal kappa opioid receptor expression was significantly decreased in brain areas implicated in responding to EtOH. CONCLUSIONS: Overall, these data suggest that prenatal EtOH affects kappa opioid function and expression and that these changes may be involved in increased drinking later in life.


Asunto(s)
Etanol/farmacología , Naltrexona/análogos & derivados , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Pirrolidinas/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Química Encefálica/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Etanol/administración & dosificación , Femenino , Expresión Génica/efectos de los fármacos , Masculino , Naltrexona/farmacología , Embarazo , Ratas Sprague-Dawley , Receptores Opioides kappa/biosíntesis
7.
J Pharmacol Exp Ther ; 344(2): 522-30, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23230215

RESUMEN

The role of dynorphin A (1-17; Dyn) and its associated kappa opioid receptor (KOR) in nociception represents a longstanding scientific conundrum: Dyn and KOR (Dyn/KOR) have variously been reported to inhibit, facilitate, or have no effect on pain. We investigated whether interactions between sex and pain type (which are usually ignored) influenced Dyn/KOR-mediated antinociception. Blockade of the spinal α(2)-noradrenergic receptor (α(2)-NAR) using yohimbine elicited comparable spinal Dyn release in females and males. Nevertheless, the yohimbine-induced antinociception exhibited sexual dimorphism that depended on the pain test used: in the intraperitoneal acetic acid-induced writhing test, yohimbine produced antinociception only in females, whereas in the intraplantar formalin-induced paw flinch test, antinociception was observed only in males. In females and males, both intrathecal Dyn antibodies and spinal KOR blockade eliminated the yohimbine-induced antinociception, indicating that Dyn/KOR mediated it. However, despite the conditional nature of spinal Dyn/KOR-mediated yohimbine antinociception, both intraplantar formalin and intraperitoneal acetic acid activated spinal Dyn neurons that expressed α(2)-NARs. Moreover, Dyn terminals apposed KOR-expressing spinal nociceptive neurons in both sexes. This similar organization suggests that the sexually dimorphic interdependent effects of sex and pain type may result from the presence of nonfunctional (silent) KORs on nociceptive spinal neurons that are responsive to intraplantar formalin (in females) versus intraperitoneal acetic acid (in males). Our findings that spinal Dyn/KOR-mediated antinociception depends on interactions between sex and pain type underscore the importance of using both sexes and multiple pain models when investigating Dyn/KOR antinociception.


Asunto(s)
Dinorfinas/metabolismo , Dolor Nociceptivo/metabolismo , Dimensión del Dolor , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Opioides kappa/metabolismo , Caracteres Sexuales , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Dinorfinas/biosíntesis , Femenino , Inyecciones Espinales , Masculino , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Dolor Nociceptivo/tratamiento farmacológico , Dolor Nociceptivo/etiología , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 2/biosíntesis , Receptores Opioides kappa/biosíntesis , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Yohimbina/farmacología
8.
Cancer Immunol Immunother ; 61(10): 1699-711, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22392190

RESUMEN

Methionine enkephalin (MENK), the endogenous neuropeptide, is known to exert direct effects on the neuroendocrine and the immune systems and participates in regulation of various functions of cells related to both the innate and adaptive immune systems. Dendritic cells (DCs) play important role in initiating and regulating T cell responses. The aim of this work is to investigate the effects of MENK on differentiation, maturation, and function of DCs derived from murine bone marrow progenitors (BM-derived DCs). Our result showed that MENK could induce BM-derived DCs to polarize predominantly to mDC subtype, rather than pDC both in vivo and in vitro, and this was in favor of Th1 response. BM-derived DCs, after treatment with MENK, up-regulated the expressions of MHC class II and key costimulatory molecules. Result by RT-PCR showed MENK could increase expressions of delta and kappa receptors on BM-derived DCs. Also MENK promoted BM-derived DCs to secret higher levels of proinflammatory cytokines of IL-12p70, TNF-α. Furthermore, differentiated BM-derived DCs treated with MENK displayed higher activity to induce allogeneic T cell proliferation and MENK also inhibited tumor growth in vivo and induced apoptosis of tumor cells in vitro. Thus, it is concluded that MENK could be an effective inducer of BM-derived DCs and might be a new therapeutic agent for cancer, as well as other immune handicapped disease. Also we may consider MENK as a potential adjuvant in vaccine preparation.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Encefalina Metionina/farmacología , Neurotransmisores/farmacología , Células Madre/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Citocinas/metabolismo , Femenino , Genes MHC Clase II/efectos de los fármacos , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Opioides delta/biosíntesis , Receptores Opioides kappa/biosíntesis , Regulación hacia Arriba/efectos de los fármacos
9.
Exp Physiol ; 97(5): 651-62, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22337865

RESUMEN

Both low-frequency electro-acupuncture (EA) and manual acupuncture improve menstrual frequency and decrease circulating androgens in women with polycystic ovary syndrome (PCOS). We sought to determine whether low-frequency EA is more effective than manual stimulation in regulating disturbed oestrous cyclicity in rats with PCOS induced by 5α-dihydrotestosterone. To identify the central mechanisms of the effects of stimulation, we assessed hypothalamic mRNA expression of molecules that regulate reproductive and neuroendocrine function. From age 70 days, rats received 2 Hz EA or manual stimulation with the needles five times per week for 4-5 weeks; untreated rats served as control animals. Specific hypothalamic nuclei were obtained by laser microdissection, and mRNA expression was measured with TaqMan low-density arrays. Untreated rats were acyclic. During the last 2 weeks of treatment, seven of eight (88%) rats in the EA group had epithelial keratinocytes, demonstrating oestrous cycle change (P = 0.034 versus control rats). In the manual group, five of eight (62%) rats had oestrous cycle changes (n.s. versus control animals). The mRNA expression of the opioid receptors Oprk1 and Oprm1 in the hypothalamic arcuate nucleus was lower in the EA group than in untreated control rats. The mRNA expression of the steroid hormone receptors Esr2, Pgr and Kiss1r was lower in the manual group than in the control animals. In rats with 5α-dihydrotestosterone-induced PCOS, low-frequency EA restored disturbed oestrous cyclicity but did not differ from the manual stimulation group, although electrical stimulation lowered serum testosterone in responders, those with restored oestrus cyclicity, and differed from both control animals and the manual stimulation group. Thus, EA cannot in all aspects be considered superior to manual stimulation. The effects of low-frequency EA may be mediated by central opioid receptors, while manual stimulation may involve regulation of steroid hormone/peptide receptors.


Asunto(s)
Terapia por Acupuntura , Electroacupuntura , Ciclo Estral/fisiología , Síndrome del Ovario Poliquístico/fisiopatología , Animales , Núcleo Arqueado del Hipotálamo/fisiopatología , Dihidrotestosterona , Modelos Animales de Enfermedad , Femenino , Síndrome del Ovario Poliquístico/inducido químicamente , Progesterona/sangre , Ratas , Receptores Opioides kappa/biosíntesis , Receptores Opioides mu/biosíntesis , Testosterona/sangre
10.
J Pharmacol Exp Ther ; 332(1): 255-65, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19828880

RESUMEN

Tricyclic antidepressants (TCAs) have been reported to interact with the opioid system, but their pharmacological activity at opioid receptors has not yet been elucidated. In the present study, we investigated the actions of amoxapine, amitriptyline, nortriptyline, desipramine, and imipramine at distinct cloned and native opioid receptors. In Chinese hamster ovary (CHO) cells expressing delta-opioid receptors (CHO/DOR), TCAs displaced [3H]naltrindole binding and stimulated guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS) binding at micromolar concentrations with amoxapine displaying the highest potency and efficacy. Amoxapine and amitriptyline inhibited cyclic AMP formation and induced the phosphorylation of signaling molecules along the extracellular signal-regulated kinase 1/2 (ERK1/2) and phosphatidylinositol-3 kinase pathways. Amoxapine also activated delta-opioid receptors in rat dorsal striatum and nucleus accumbens and human frontal cortex. In CHO cells expressing kappa-opioid receptors (CHO/KOR), TCAs, but not amoxapine, exhibited higher receptor affinity and more potent stimulation of [(35)S]GTPgammaS binding than in CHO/DOR and effectively inhibited cyclic AMP accumulation. Amitriptyline regulated ERK1/2 phosphorylation and activity in CHO/KOR and C6 glioma cells endogenously expressing kappa-opioid receptors, and this effect was attenuated by the kappa-opioid antagonist nor-binaltorphimine. In rat nucleus accumbens, amitriptyline slightly inhibited adenylyl cyclase activity and counteracted the inhibitory effect of the full kappa agonist trans-(-)-3,4dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamide (U50,488). At the cloned mu-opioid receptor, TCAs showed low affinity and no significant agonist activity. These results show that TCAs differentially regulate opioid receptors with a preferential agonist activity on either delta or kappa subtypes and suggest that this property may contribute to their therapeutic and/or side effects.


Asunto(s)
Antidepresivos Tricíclicos/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Adulto , Anciano , Animales , Células CHO , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Unión Proteica , Ensayo de Unión Radioligante , Ratas , Receptores Opioides delta/biosíntesis , Receptores Opioides kappa/biosíntesis , Receptores Opioides mu/biosíntesis
11.
Neurotox Res ; 37(4): 800-814, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32026358

RESUMEN

Disturbances in the function of the mesostriatal dopamine system may contribute to the development and maintenance of chronic pain, including its sensory and emotional/cognitive aspects. In the present study, we assessed the influence of chronic constriction injury (CCI) of the sciatic nerve on the expression of genes coding for dopamine and opioid receptors as well as opioid propeptides in the mouse mesostriatal system, particularly in the nucleus accumbens. We demonstrated bilateral increases in mRNA levels of the dopamine D1 and D2 receptors (the latter accompanied by elevated protein level), opioid propeptides proenkephalin and prodynorphin, as well as delta and kappa (but not mu) opioid receptors in the nucleus accumbens at 7 to 14 days after CCI. These results show that CCI-induced neuropathic pain is accompanied by a major transcriptional dysregulation of molecules involved in dopaminergic and opioidergic signaling in the striatum/nucleus accumbens. Possible functional consequences of these changes include opposite effects of upregulated enkephalin/delta opioid receptor signaling vs. dynorphin/kappa opioid receptor signaling, with the former most likely having an analgesic effect and the latter exacerbating pain and contributing to pain-related negative emotional states.


Asunto(s)
Neuralgia/metabolismo , Dimensión del Dolor/métodos , Prosencéfalo/metabolismo , Receptores Dopaminérgicos/biosíntesis , Receptores Opioides delta/biosíntesis , Receptores Opioides kappa/biosíntesis , Animales , Cuerpo Estriado/metabolismo , Encefalinas/biosíntesis , Encefalinas/genética , Expresión Génica , Masculino , Ratones , Neuralgia/genética , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/genética , Receptores Dopaminérgicos/genética , Receptores Opioides delta/genética , Receptores Opioides kappa/genética , Receptores Opioides mu/biosíntesis , Receptores Opioides mu/genética
12.
Biomed Pharmacother ; 109: 938-944, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30551548

RESUMEN

Social isolation stress (SIS) as a type of chronic stress could induce depressive- and anxiety-like behaviors. Our study evaluates the role of opioid system on negative behavioral impacts of SIS in male NMRI mice. We investigated effects of morphine, a nonselective opioid receptor (OR) agonist, naltrexone (NLX), an OR antagonist, naltrindole (NLT), a delta opioid receptor (DOR) antagonist, SNC80, a DOR agonist, U-69593, a kappa opioid receptor (KOR) agonist, nor-Binaltorphimine, a selective KOR antagonist and cyprodime hydrochloride a selective mu opioid receptor (MOR) antagonist on depressive- and anxiety-like behaviors. Using RT-PCR we evaluated ORs gene expression in mice brain. Our findings showed that SIS induced anxiety- and depressive-like behavior in the forced swimming test, open field test, splash test and hole-board test. Moreover, administration of SNC-80 significantly mitigated anxiety- and depressive-like behaviors. NLT decreased grooming-activity in the splash test. Excitingly, administration of agents affecting KOR failed to alter the negative effects of SIS. RT-PCR demonstrated that MOR and KOR gene expression decreased in socially isolated mice; however, SIS did not affect DORs expression. Our findings suggest that SIS at least in part, probably via altering endogenous opioids particularly MORs and KORs but not DORs mediated negative impacts on behavior; also, it could be concluded that DORs might be considered as a novel target for studying depression and anxiety.


Asunto(s)
Receptores Opioides delta/biosíntesis , Receptores Opioides kappa/biosíntesis , Receptores Opioides mu/biosíntesis , Aislamiento Social/psicología , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología , Analgésicos Opioides/farmacología , Animales , Ansiedad/metabolismo , Ansiedad/psicología , Depresión/metabolismo , Depresión/psicología , Masculino , Ratones , Antagonistas de Narcóticos/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores
13.
Neuroscience ; 151(1): 129-37, 2008 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18082336

RESUMEN

Zebrafish is a novel experimental model that has been used in developmental studies as well as in the study of pathological processes involved in human diseases. It has been demonstrated that the endogenous opioid system is involved in developmental mechanisms. We have studied the relationship between the different embryonic stages and opioid receptor expression for the four known opioid receptors in zebrafish (mu, delta 1, delta 2 and kappa). The mu opioid receptor is detected at higher levels than the other opioid receptors before the midblastula transition and during the segmentation period. The delta duplicate 2 exhibits only one peak of expression at 21 h postfertilization (hpf), when the motor nervous system is forming. The kappa receptor is expressed at very low levels. In situ hybridization studies at 24 hpf show that the opioid receptors are widely distributed in zebrafish CNS and at 48 hpf their localization is detected in more defined structures. Our results support specific implications of the opioid receptors in developmental processes such as morphogenesis of the CNS, neurogenesis, neuroprotection and development of neuromuscular and digestive system. Pain-related alterations can be a consequence of changes in the endogenous opioid system during development, hence we provide important information that might help to solve pain-related pathological situations.


Asunto(s)
Envejecimiento/metabolismo , Receptores Opioides/biosíntesis , Receptores Opioides/metabolismo , Actinas/biosíntesis , Actinas/genética , Animales , Química Encefálica/genética , Química Encefálica/fisiología , Clonación Molecular , ADN Complementario/biosíntesis , ADN Complementario/genética , Embrión no Mamífero , Hibridación in Situ , ARN/biosíntesis , ARN/aislamiento & purificación , Receptores Opioides delta/biosíntesis , Receptores Opioides delta/genética , Receptores Opioides kappa/biosíntesis , Receptores Opioides kappa/genética , Receptores Opioides mu/biosíntesis , Receptores Opioides mu/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular , Pez Cebra
14.
J Neuroimmunol ; 321: 12-23, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29957382

RESUMEN

Regulation of µ-, δ- and κ-opioid receptor protein level in spleen lymphocytes when stimulated by mitogen is not known. To answer the question whether these cells do express opioid receptor (OR) proteins, primary, fresh rat spleen lymphocytes were prepared and stimulated for 48 h with mitogenic dose of Con A. The unstimulated lymphocytes did not express µ- and δ-OR proteins in detectable amounts, however, stimulation with Con A resulted in appearance of clearly detectable immunoblot signals of both µ-OR and δ-OR. κ-OR were detected already in primary cells and increased 2.4-fold in Con A-stimulated cells. These results were supported by data obtained by flow cytometry analysis indicating a dramatic increase in number of µ-, δ- and κ-OR expressing cells after mitogen stimulation. The newly synthesized µ-, δ- and κ-OR in Con A-stimulated spleen lymphocytes were present in the cells interior and not functionally mature, at least in terms of their ability to enhance activity of trimeric G proteins determined by three different protocols of agonist-stimulated, high-affinity [35S]GTPγS binding assay. The up-regulation of µ-, δ- and κ-OR was associated with specific decrease of their cognate trimeric G proteins, Gi1α/Gi2α; the other Gα and Gß subunits were unchanged. The level of ß-arrestin-1/2 was also decreased in Con A-stimulated splenocytes. We conclude that up-regulation of OR expression level in spleen lymphocytes by Con A proceeds in conjunction with down-regulation of their intracellular signaling partners, Gi1α/Gi2α proteins and ß-arrestin-1/2. These regulatory proteins are expressed in high amounts already in unstimulated cells and decreased by mitogen stimulation.


Asunto(s)
Linfocitos/metabolismo , Receptores Opioides delta/biosíntesis , Receptores Opioides kappa/biosíntesis , Receptores Opioides mu/biosíntesis , Bazo/metabolismo , Animales , Concanavalina A/farmacología , Linfocitos/efectos de los fármacos , Masculino , Mitógenos/farmacología , Ratas , Ratas Wistar , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos , Bazo/citología , Bazo/efectos de los fármacos , Regulación hacia Arriba
15.
Neuropharmacology ; 140: 162-173, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30075159

RESUMEN

There is an important emerging role for the endogenous opioid dynorphin (DYN) and the kappa-opioid receptor (KOR) in the treatment of alcohol dependence. Evidence suggests that the DYN/KOR system in the bed nucleus of the stria terminalis (BNST) contributes to maladaptive behavioral regulation during withdrawal in alcohol dependence. The current experiments were designed to assess dysregulation of the BNST DYN/KOR system by evaluating alcohol dependence-induced changes in DYN/KOR gene expression (Pdyn and Oprk1, respectively), and the sensitivity of alcohol self-administration, negative affective-like behavior and physiological withdrawal to intra-BNST KOR antagonism during acute withdrawal. Wistar rats trained to self-administer alcohol, or not trained, were subjected to an alcohol dependence induction procedure (14 h alcohol vapor/10 h air) or air-exposure. BNST micropunches from air- and vapor-exposed animals were analyzed using RT-qPCR to quantify dependence-induced changes in Pdyn and Oprk1 mRNA expression. In addition, vapor- and air-exposed groups received an intra-BNST infusion of a KOR antagonist or vehicle prior to measurement of alcohol self-administration. A separate cohort of vapor-exposed rats was assessed for physiological withdrawal and negative affective-like behavior signs following intra-BNST KOR antagonism. During acute withdrawal, following alcohol dependence induction, there was an upregulation in Oprk1 mRNA expression in alcohol self-administering animals, but not non-alcohol self-administering animals, that confirmed dysregulation of the KOR/DYN system within the BNST. Furthermore, intra-BNST KOR antagonism attenuated escalated alcohol self-administration and negative affective-like behavior during acute withdrawal without reliably impacting physiological symptoms of withdrawal. The results confirm KOR system dysregulation in the BNST in alcohol dependence, illustrating the therapeutic potential of targeting the KOR to treat alcohol dependence.


Asunto(s)
Alcoholismo/metabolismo , Alcoholismo/psicología , Receptores Opioides kappa/biosíntesis , Núcleos Septales/metabolismo , Síndrome de Abstinencia a Sustancias/prevención & control , Alcoholismo/complicaciones , Animales , Condicionamiento Operante/efectos de los fármacos , Dinorfinas/biosíntesis , Etanol/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Naltrexona/análogos & derivados , Naltrexona/farmacología , ARN Mensajero/biosíntesis , Ratas , Receptores Opioides kappa/antagonistas & inhibidores , Autoadministración , Núcleos Septales/efectos de los fármacos , Regulación hacia Arriba , Vocalización Animal/efectos de los fármacos
16.
J Neurosci ; 26(38): 9743-9, 2006 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-16988045

RESUMEN

The expression of kappa opioid receptor (KOR) is subjected to both transcriptional and posttranscriptional controls. We report that KOR translation is regulated by netrin-1 in primary neurons of dorsal root ganglion (DRG) and in P19 embryonal carcinoma cells. Without stimulation, a significant portion of KOR mRNA is maintained in a dormant state and partitions in the translationally inactive, post-polysomal fraction. During netrin-1 stimulation, which activates its downstream target focal adhesion kinase (FAK), KOR mRNA rapidly partitions to the translationally active polysomal fraction. Functionally, the newly synthesized KOR proteins in DRG neurons are able to bind to specific ligands. This report describes the first example of netrin-1 signaling in the translational control of a drug receptor KOR, which involves the mediator of netrin-1, FAK, and a novel mechanism that enhances the association of target mRNA with polysomes for translational activation.


Asunto(s)
Factores de Crecimiento Nervioso/fisiología , Polirribosomas/metabolismo , Biosíntesis de Proteínas/fisiología , Receptores Opioides kappa/biosíntesis , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Línea Celular Tumoral , Ganglios Espinales/metabolismo , Ratones , Netrina-1 , Polirribosomas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Opioides kappa/genética
17.
Neuropharmacology ; 117: 124-133, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28163104

RESUMEN

Anxiety disorders are one of the most common and debilitating mental illnesses worldwide. Growing evidence indicates an age-dependent rise in the incidence of anxiety disorders from adolescence through adulthood, suggestive of underlying neurodevelopmental mechanisms. Kappa opioid receptors (KORs) are known to contribute to the development and expression of anxiety; however, the functional role of KORs in the basolateral amygdala (BLA), a brain structure critical in mediating anxiety, particularly across ontogeny, are unknown. Using whole-cell patch-clamp electrophysiology in acute brain slices from adolescent (postnatal day (P) 30-45) and adult (P60+) male Sprague-Dawley rats, we found that the KOR agonist, U69593, increased the frequency of GABAA-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) in the adolescent BLA, without an effect in the adult BLA or on sIPSC amplitude at either age. The KOR effect was blocked by the KOR antagonist, nor-BNI, which alone did not alter GABA transmission at either age, and the effect of the KOR agonist was TTX-sensitive. Additionally, KOR activation did not alter glutamatergic transmission in the BLA at either age. In contrast, U69593 inhibited sIPSC frequency in the central amygdala (CeA) at both ages, without altering sIPSC amplitude. Western blot analysis of KOR expression indicated that KOR levels were not different between the two ages in either the BLA or CeA. This is the first study to provide compelling evidence for a novel and unique neuromodulatory switch in one of the primary brain regions involved in initiating and mediating anxiety that may contribute to the ontogenic rise in anxiety disorders.


Asunto(s)
Envejecimiento/fisiología , Complejo Nuclear Basolateral/metabolismo , Receptores Opioides kappa/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Complejo Nuclear Basolateral/efectos de los fármacos , Bencenoacetamidas/antagonistas & inhibidores , Bencenoacetamidas/farmacología , Ácido Glutámico/fisiología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Naltrexona/análogos & derivados , Naltrexona/farmacología , Pirrolidinas/antagonistas & inhibidores , Pirrolidinas/farmacología , Ratas , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/biosíntesis , Transmisión Sináptica/efectos de los fármacos
18.
Neuropharmacology ; 50(1): 123-32, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16360182

RESUMEN

The development of neuropathic pain is associated with multiple changes in gene expression occurring in the dorsal root ganglia (DRG) and spinal cord. The goal of this study was to evaluate whether the disruption of CB1 cannabinoid receptor gene modulates the changes induced by neuropathic pain in the expression of mu- (MOR), delta- (DOR) and kappa-opioid receptors (KOR) mRNA levels in the DRG and spinal cord. The induction of c-fos expression in the lumbar and sacral regions of the spinal cord was also evaluated in these animals. Opioid receptors mRNA levels were determined by using real-time PCR and Fos protein levels by immunohistochemistry. Nerve injury significantly reduced the expression of MOR in the DRG and the lumbar section of the spinal cord from CB1 cannabinoid knockout (KO) mice and wild-type littermates (WT). In contrast, mRNA levels of DOR and KOR were not significantly changed in any of the different sections analysed. Furthermore, sciatic nerve injury evoked a similar increase of c-fos expression in lumbar and sacral regions of the spinal cord of both KO and WT. In all instances, no significant differences were observed between WT and KO mice. These data revealed specific changes induced by neuropathic pain in MOR expression and c-fos levels in the DRG and/or spinal cord that were not modified by the genetic disruption of CB1 cannabinoid receptors.


Asunto(s)
Genes fos/genética , Dolor/etiología , Dolor/fisiopatología , Enfermedades del Sistema Nervioso Periférico/etiología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/fisiología , Receptores Opioides/biosíntesis , Traumatismos de la Médula Espinal/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Recuento de Células , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Hiperalgesia/inducido químicamente , Hiperalgesia/psicología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Opioides delta/biosíntesis , Receptores Opioides delta/genética , Receptores Opioides kappa/biosíntesis , Receptores Opioides kappa/genética , Receptores Opioides mu/biosíntesis , Receptores Opioides mu/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/fisiología , Traumatismos de la Médula Espinal/genética
19.
Neurosci Lett ; 405(1-2): 94-9, 2006 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-16842913

RESUMEN

A cDNA that encodes a kappa opioid receptor like from zebrafish (ZFOR3) has been cloned and characterized. The encoded protein is 377 residues long and presents 70% identity with the mammalian kappa receptors, although less homology is found in the amino- and carboxyl-terminus as well as in the extracellular loops. In situ hybridization studies have revealed that ZFOR3 mRNA is highly expressed in particular brain areas that coincide with the expression of the kappa opioid receptor in other species. When ZFOR3 is stably expressed in HEK293 cells, [(3)H]-diprenorphine binds with high affinity (K(D)=1.05+/-0.26 nM), being this value on the same range as those reported for mammalian kappa opioid receptors. On the other hand, the selective agonist for mammalian kappa receptors U69,593 does not bind to ZFOR3. [(3)H]-diprenorphine binding is readily displaced by the peptidic ligand dynorphin A and by the non-endogenous compounds bremazocine, naloxone and morphine, although with different affinities. Our results demonstrate that ZFOR3 is a unique model to study the kappa opioid receptor functionality.


Asunto(s)
Receptores Opioides kappa/genética , Proteínas de Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Unión Competitiva , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Línea Celular , Humanos , Hibridación in Situ , Ligandos , Datos de Secuencia Molecular , Ensayo de Unión Radioligante , Receptores Opioides kappa/biosíntesis , Receptores Opioides kappa/aislamiento & purificación , Homología de Secuencia de Aminoácido , Pez Cebra , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/aislamiento & purificación
20.
FEBS Lett ; 435(1): 65-8, 1998 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-9755860

RESUMEN

KOR-3, also known as ORL-1, is a member of the opioid receptor family, encoding the murine receptor for orphanin FQ/nociceptin. In the current studies we have identified five different splice variants of KOR-3 in mouse brain, three of which have not been previously reported. In addition to variants with a 15 bp deletion at the 3'-end of the first coding exon (KOR-3d) and an 81 bp insertion between the second and third coding exons (KOR-3e), three new variants with insertions of 34 (KOR-3a), 98 (KOR-3b), and 139 bp (KOR-3c) between the first and second coding exons have been obtained. The expression of the three variants in mouse brain varies markedly among brain regions with a distribution which is quite distinct from KOR-3 itself. Of greatest interest was the presence of high levels of KOR-3a in the striatum, a region with no demonstrable KOR-3, and in the cortex. KOR-3c was seen in the periaqueductal gray and hypothalamus, regions where KOR-3 predominated. The brainstem had similar levels of KOR-3, KOR-3a, and KOR-3d. In contrast, KOR-3d was most prominent in the cerebellum. KOR-3b levels were very low throughout.


Asunto(s)
Empalme Alternativo , Encéfalo/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores Opioides kappa/genética , Receptores Opioides , Animales , Secuencia de Bases , Northern Blotting , Química Encefálica/genética , Clonación Molecular , ADN Complementario/aislamiento & purificación , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/biosíntesis , Especificidad de Órganos/genética , Receptores Opioides kappa/biosíntesis , Receptor de Nociceptina
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda