Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Handb Exp Pharmacol ; 247: 87-114, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28993838

RESUMEN

The functional diversity of primary afferent neurons of the dorsal root ganglia (DRG) generates a variety of qualitatively and quantitatively distinct somatosensory experiences, from shooting pain to pleasant touch. In recent years, the identification of dozens of genetic markers specifically expressed by subpopulations of DRG neurons has dramatically improved our understanding of this diversity and provided the tools to manipulate their activity and uncover their molecular identity and function. Opioid receptors have long been known to be expressed by discrete populations of DRG neurons, in which they regulate cell excitability and neurotransmitter release. We review recent insights into the identity of the DRG neurons that express the delta opioid receptor (DOR) and the ion channel mechanisms that DOR engages in these cells to regulate sensory input. We highlight recent findings derived from DORGFP reporter mice and from in situ hybridization and RNA sequencing studies in wild-type mice that revealed DOR presence in cutaneous mechanosensory afferents eliciting touch and implicated in tactile allodynia. Mechanistically, we describe how DOR modulates opening of voltage-gated calcium channels (VGCCs) to control glutamatergic neurotransmission between somatosensory neurons and postsynaptic neurons in the spinal cord dorsal horn. We additionally discuss other potential signaling mechanisms, including those involving potassium channels, which DOR may engage to fine tune somatosensation. We conclude by discussing how this knowledge may explain the analgesic properties of DOR agonists against mechanical pain and uncovers an unanticipated specialized function for DOR in cutaneous mechanosensation.


Asunto(s)
Neuronas Aferentes/metabolismo , Receptores Opioides delta/biosíntesis , Células Receptoras Sensoriales/metabolismo , Animales , Ganglios Espinales/metabolismo , Humanos , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Receptores Opioides delta/agonistas
2.
Handb Exp Pharmacol ; 247: 227-260, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28035528

RESUMEN

Delta opioid receptor (DOR) displays a unique, highly conserved, structure and an original pattern of distribution in the central nervous system, pointing to a distinct and specific functional role among opioid peptide receptors. Over the last 15 years, in vivo pharmacology and genetic models have allowed significant advances in the understanding of this role. In this review, we will focus on the involvement of DOR in modulating different types of hippocampal- and striatal-dependent learning processes as well as motor function, motivation, and reward. Remarkably, DOR seems to play a key role in balancing hippocampal and striatal functions, with major implications for the control of cognitive performance and motor function under healthy and pathological conditions.


Asunto(s)
Aprendizaje/fisiología , Motivación/fisiología , Receptores Opioides delta/fisiología , Animales , Humanos , Aprendizaje/efectos de los fármacos , Motivación/efectos de los fármacos , Receptores Opioides delta/biosíntesis , Receptores Opioides delta/efectos de los fármacos , Recompensa
3.
J Neurosci Res ; 94(12): 1531-1545, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27661001

RESUMEN

Brain injuries, such as cerebral hypoxia-ischemia (H-I), induce a regenerative response from the neural stem/progenitors (NSPs) of the subventricular zone (SVZ); however, the mechanisms that regulate this expansion have not yet been fully elucidated. The Notch- Delta-Serrate-Lag2 (DSL) signaling pathway is considered essential for the maintenance of neural stem cells, but it is not known if it is necessary for the expansion of the NSPs subsequent to perinatal H-I injury. Therefore, the aim of this study was to investigate whether this pathway contributes to NSP expansion in the SVZ after H-I and, if so, to establish whether this pathway is directly induced by H-I or regulated by paracrine factors. Here we report that Notch1 receptor induction and one of its ligands, Delta-like 1, precedes NSP expansion after perinatal H-I in P6 rat pups and that this increase occurs specifically in the most medial cell layers of the SVZ where the stem cells reside. Pharmacologically inhibiting Notch signaling in vivo diminished NSP expansion. With an in vitro model of H-I, Notch1 was not induced directly by hypoxia, but was stimulated by soluble factors, specifically leukemia inhibitory factor, produced by astrocytes within the SVZ. These data confirm the importance both of the Notch-DSL signaling pathway in the expansion of NSPs after H-I and in the role of the support cells in their niche. They further support the body of evidence that indicates that leukemia inhibitory factor is a key injury-induced cytokine that is stimulating the regenerative response of the NSPs. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Astrocitos/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Factor Inhibidor de Leucemia/biosíntesis , Regeneración Nerviosa , Células-Madre Neurales , Animales , Citocinas/metabolismo , Diaminas/farmacología , Femenino , Ventrículos Laterales/patología , Embarazo , Ratas , Ratas Wistar , Receptor Notch1/biosíntesis , Receptor Notch1/genética , Receptores Opioides delta/biosíntesis , Transducción de Señal , Tiazoles/farmacología
4.
J Neurochem ; 124(6): 808-20, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23286559

RESUMEN

Although biochemical and physiological evidence suggests a strong interaction between striatal CB1 cannabinoid (CB1 R) and D2 dopamine (D2 R) receptors, the mechanisms are poorly understood. We targeted medium spiny neurons of the indirect pathway using shRNA to knockdown either CB1 R or D2 R. Chronic reduction in either receptor resulted in deficits in gene and protein expression for the alternative receptor and concomitantly increased expression of the cannabinoid receptor interacting protein 1a (CRIP1a), suggesting a novel role for CRIP1a in dopaminergic systems. Both CB1 R and D2 R knockdown reduced striatal dopaminergic-stimulated [(35) S]GTPγS binding, and D2 R knockdown reduced pallidal WIN55212-2-stimulated [(35) S]GTPγS binding. Decreased D2 R and CB1 R activity was associated with decreased striatal phosphoERK. A decrease in mRNA for opioid peptide precursors pDYN and pENK accompanied knockdown of CB1 Rs or D2 Rs, and over-expression of CRIP1a. Down-regulation in opioid peptide mRNAs was followed in time by increased DOR1 but not MOR1 expression, leading to increased [D-Pen2, D-Pen5]-enkephalin-stimulated [(35) S]GTPγS binding in the striatum. We conclude that mechanisms intrinsic to striatal medium spiny neurons or extrinsic via the indirect pathway adjust for changes in CB1 R or D2 R levels by modifying the expression and signaling capabilities of the alternative receptor as well as CRIP1a and the DELTA opioid system.


Asunto(s)
Proteínas Portadoras/biosíntesis , Cuerpo Estriado/metabolismo , Receptor Cannabinoide CB1/fisiología , Receptores de Dopamina D2/fisiología , Receptores Opioides delta/biosíntesis , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Antagonistas de los Receptores de Dopamina D2 , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptores Opioides delta/genética
5.
Cereb Cortex ; 22(5): 1215-23, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21810780

RESUMEN

Certain cognitive deficits in schizophrenia have been linked to dysfunction of prefrontal cortical (PFC) γ-aminobutyric acid (GABA) neurons and appear neurodevelopmental in nature. Since opioids suppress GABA neuron activity, we conducted the first study to determine 1) whether the µ opioid receptor (MOR), δ opioid receptor (DOR), and opioid ligand proenkephalin are altered in the PFC of a large cohort of schizophrenia subjects and 2) the postnatal developmental trajectory in monkey PFC of opioid markers that are altered in schizophrenia. We used quantitative polymerase chain reaction to measure mRNA levels from 42 schizophrenia and 42 matched healthy comparison subjects; 18 monkeys chronically exposed to haloperidol, olanzapine, or placebo; and 49 monkeys aged 1 week-11.5 years. We found higher levels for MOR mRNA (+27%) in schizophrenia but no differences in DOR or proenkephalin mRNAs. Elevated MOR mRNA levels in schizophrenia did not appear to be explained by substance abuse, psychotropic medications, or illness chronicity. Finally, MOR mRNA levels declined through early postnatal development, stabilized shortly before adolescence and increased across adulthood in monkey PFC. In schizophrenia, higher MOR mRNA levels may contribute to suppressed PFC GABA neuron activity and might be attributable to alterations in the postnatal developmental trajectory of MOR signaling.


Asunto(s)
Encefalinas/biosíntesis , Corteza Prefrontal/metabolismo , Precursores de Proteínas/biosíntesis , Receptores Opioides delta/biosíntesis , Receptores Opioides mu/biosíntesis , Esquizofrenia/metabolismo , Animales , Antipsicóticos/farmacología , Benzodiazepinas/farmacología , Femenino , Haloperidol/farmacología , Humanos , Macaca fascicularis , Masculino , Persona de Mediana Edad , Olanzapina , Reacción en Cadena de la Polimerasa , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/crecimiento & desarrollo , ARN Mensajero/análisis , Esquizofrenia/fisiopatología
6.
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
7.
J Neurosci ; 30(13): 4735-45, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20357124

RESUMEN

By sustained activation of mu-opioid receptors (MORs), chronic opioids cause analgesic tolerance, physical dependence, and opioid addiction, common clinical problems for which an effective treatment is still lacking. Chronic opioids recruit delta-opioid receptors (DORs) to plasma membrane through exocytotic trafficking, but the role of this new DOR and its interaction with existing MOR in brain functions and in these clinical problems remain largely unknown. In this study, we investigated the mechanisms underlying synaptic and behavioral actions of chronic morphine-induced DORs and their interaction with MORs in nucleus raphe magnus (NRM) neurons important for opioid analgesia. We found that the emerged DOR inhibited GABAergic IPSCs through both the phospholipase A(2) (PLA(2)) and cAMP/protein kinase A (PKA) signaling pathways. MOR inhibition of IPSCs, normally mediated predominantly by the PLA(2) pathway, was additionally mediated by the cAMP/PKA pathway, with MOR potency significantly increased after chronic morphine treatment. Isobologram analysis revealed a synergistic DOR-MOR interaction in their IPSC inhibition, which was dependent on upregulated activities of both the PLA(2) and cAMP/PKA pathways. Furthermore, DOR and MOR agonists microinjected into the NRM in vivo also produced a PLA(2)-dependent synergism in their antinociceptive effects. These findings suggest that the cAMP/PKA pathway, upregulated by chronic opioids, becomes more important in the mechanisms of both MOR and DOR inhibition of GABA synaptic transmission after chronic opioid exposure, and DORs and MORs are synergic both synaptically and behaviorally in producing analgesic effects in a PLA(2)-dependent fashion, supporting the potential therapeutic use of DOR agonists in pain management under chronic opioid conditions.


Asunto(s)
Receptores Opioides delta/fisiología , Receptores Opioides mu/fisiología , Sinapsis/fisiología , Analgésicos Opioides/farmacología , Animales , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/metabolismo , AMP Cíclico/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Sinergismo Farmacológico , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Morfina/farmacología , Técnicas de Placa-Clamp , Fosfolipasas A2/metabolismo , Fosfolipasas A2/fisiología , Terminales Presinápticos/fisiología , Ratas , Ratas Wistar , Receptores Opioides delta/agonistas , Receptores Opioides delta/biosíntesis , Receptores Opioides mu/agonistas , Transducción de Señal , Sinapsis/efectos de los fármacos , Regulación hacia Arriba , Ácido gamma-Aminobutírico/metabolismo
8.
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
9.
J Pharmacol Exp Ther ; 334(3): 887-96, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20498253

RESUMEN

Both delta-opioid receptor (DOPr) and cannabinoid-2 receptor (CB2R) agonists attenuate neuropathic pain, but the precise mechanism implicated in these effects is not completely elucidated. We investigated whether nitric oxide synthesized by neuronal (NOS1) or inducible (NOS2) nitric-oxide synthases could modulate DOPr and/or CB2R antiallodynic and antihyperalgesic effects through the peripheral nitric oxide-cGMP-protein kinase G (PKG) pathway activation and affect their expression during neuropathic pain. In wild-type (WT) mice at 21 days after chronic constriction of sciatic nerve, we evaluated the effects of [d-Pen(2),d-Pen(5)]-enkephalin (DPDPE); (2-methyl-1-propyl-1H-indol-3-yl)-1-naphthalenylmethanone (JWH-015); and a NOS1 [N-[(4S)-4-amino-5-[(2-aminoethyl)amino]pentyl]-N'-nitroguanidine tris(trifluoroacetate) salt; NANT], NOS2 [l-N(6)-(1-iminoethyl)-lysine; l-NIL], l-guanylate cyclase [1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one; ODQ], or PKG [(Rp)-8-(para-chlorophenylthio)guanosine-3',5'-cyclic monophosphorothioate; Rp-8-pCPT-cGMPs] inhibitor administered alone or combined. Expression of DOPr and CB2R mRNA in the spinal cord and dorsal root ganglia of naive and nerve-injured WT, NOS1-knockout (KO), and NOS2-KO mice, also was assessed. The subplantar administration of NANT, l-NIL, ODQ, or Rp-8-pCPT-cGMPs dose-dependently inhibited neuropathic pain and enhanced the local effects of DPDPE or JWH-015. Moreover, although the basal levels of DOPr and CB2R mRNA were similar between WT and NOS-KO animals, nerve injury only decreased (DOPr) or increased (CB2R) their expression in the dorsal root ganglia of WT and NOS2-KO mice, and not in NOS1-KO mice. Results suggest that inactivation of the nitric oxide-cGMP-PKG peripheral pathway triggered by NOS1 and NOS2 enhanced the peripheral actions of DOPr and CB2R agonists and that nitric oxide synthesized by NOS1 is implicated in the peripheral regulation of DOPr and CB2R gene transcription during neuropathic pain.


Asunto(s)
Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Óxido Nítrico/fisiología , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/metabolismo , Receptor Cannabinoide CB2/biosíntesis , Receptores Opioides delta/biosíntesis , Animales , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Ganglios Espinales/metabolismo , Calor , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo II/genética , Dimensión del Dolor/efectos de los fármacos , Estimulación Física , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/antagonistas & inhibidores , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neuropatía Ciática/patología , Transducción de Señal/efectos de los fármacos , Médula Espinal/metabolismo
10.
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
11.
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
12.
J Pharmacol Exp Ther ; 326(3): 725-31, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18541716

RESUMEN

Pharmacological and behavioral studies suggest that spinal delta- and kappa-opioid antinociceptive systems are functionally associated with ovarian sex steroids. These interactions can be demonstrated specifically during pregnancy or hormone-simulated pregnancy (HSP). The analgesia associated with both conditions can be abolished by blockade of either spinal kappa-opioid receptors or delta-opioid receptors (DOR). Furthermore, both dynorphin (DYN) release (J Pharmacol Exp Ther 298:1213-1220, 2001) and the processing of the DYN precursor (J Neurochem 65:1374-1380, 1995) are significantly increased in the spinal cord during HSP. We undertook the current study to determine whether DYN, DOR, and estrogen receptor alpha (ERalpha) share anatomical relationships that permit their direct interaction. Coexpression of DOR or ERalpha by DYN neurons was assessed using fluorescence immunohistochemistry and a synaptosomal release assay. Findings show that ERalpha and DYN are coexpressed. Moreover, in the spinal cord of HSP animals, there were significant increases in the number of DYN-immunoreactive (DYN-ir) cells, ERalpha-ir cells, cells double-labeled for DYN-ir and ERalpha-ir and the proportion of DYN-ir cells coexpressing ERalpha. Some varicose fibers in the spinal cord dorsal horn and intermediate gray matter that expressed DYN-ir also expressed DOR-ir. Activation of DORs located on DYN terminals was sufficient to inhibit K(+)-evoked DYN release. These data define, at least in part, the anatomical substrates that may be relevant to the antinociception of gestation and its hormonal simulation. Furthermore, they provide a framework for understanding sex-based nociception and antinociception and suggest novel strategies for treating pain.


Asunto(s)
Dinorfinas/biosíntesis , Receptor alfa de Estrógeno/biosíntesis , Hormonas Esteroides Gonadales/biosíntesis , Ovario/anatomía & histología , Ovario/metabolismo , Receptores Opioides delta/biosíntesis , Médula Espinal/anatomía & histología , Médula Espinal/metabolismo , Analgésicos Opioides/metabolismo , Animales , Dinorfinas/análisis , Dinorfinas/genética , Receptor alfa de Estrógeno/análisis , Receptor alfa de Estrógeno/genética , Femenino , Hormonas Esteroides Gonadales/genética , Neuronas/química , Neuronas/metabolismo , Ovario/química , Dolor/genética , Dolor/metabolismo , Dolor/patología , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/análisis , Receptores Opioides delta/genética , Médula Espinal/química
13.
J Neuroimmunol ; 197(1): 21-8, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18486241

RESUMEN

Endogenous opioid peptides are locally produced at the inflammatory site where antigens are captured and processed by dendritic cells (DCs). Subsequently, maturing DCs migrate towards draining lymph nodes to initiate T cell response. Given the primordial role of DCs in adaptive immune response, we examined whether opioids may affect the migratory response of DCs. We found that the delta opioid receptor (DOR) mRNA was expressed at low level in bone marrow-derived immature DCs and up-regulated upon DC maturation. Moreover, DOR agonists triggered DC chemotaxis in vitro. In vivo, enkephalins prevented the egress of mature DCs injected into the peritoneal cavity of normal mice. This effect was inhibited by blocking opioid receptors on mature DCs. The cross-talk between CCR7 and DOR receptors that are both up-regulated during DC maturation was then examined. Whereas opioids did not alter the migratory responsiveness to CCR7 ligands, DOR-mediated mobilization of mature DCs was inhibited by CCL19 and CCL21 suggesting that the opioid chemotactic activity decreases as the concentration of the chemokines increases.


Asunto(s)
Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Quimiotaxis de Leucocito/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Receptores Opioides delta/fisiología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Quimiocina CCL19/fisiología , Quimiocina CCL21/fisiología , Células Dendríticas/citología , Células Dendríticas/trasplante , Encefalina D-Penicilamina (2,5)/agonistas , Encefalina D-Penicilamina (2,5)/biosíntesis , Encefalina D-Penicilamina (2,5)/fisiología , Mediadores de Inflamación/fisiología , Ratones , Ratones Endogámicos BALB C , Receptor Cross-Talk/inmunología , Receptores CCR7/fisiología , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides delta/biosíntesis , Regulación hacia Arriba/inmunología
14.
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
15.
Br J Pharmacol ; 175(13): 2622-2634, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29579315

RESUMEN

BACKGROUND AND PURPOSE: To better understand opioid signalling in visceral nociceptors, we examined the expression and selective activation of µ and δ opioid receptors by dorsal root ganglia (DRG) neurons innervating the mouse colon. EXPERIMENTAL APPROACH: DRG neurons projecting to the colon were identified by retrograde tracing. δ receptor-GFP reporter mice, in situ hybridization, single-cell RT-PCR and µ receptor-specific antibodies were used to characterize expression of µ and δ receptors. Voltage-gated Ca2+ currents and neuronal excitability were recorded in small diameter nociceptive neurons (capacitance <30 pF) by patch clamp and ex vivo single-unit afferent recordings were obtained from the colon. KEY RESULTS: In situ hybridization of oprm1 expression in Fast Blue-labelled DRG neurons was observed in 61% of neurons. µ and δ receptors were expressed by 36-46% of colon DRG neurons, and co-expressed by ~25% of neurons. µ and δ receptor agonists inhibited Ca2+ currents in DRG, effects blocked by opioid antagonists. One or both agonists inhibited action potential firing by colonic afferent endings. Incubation of neurons with supernatants from inflamed colon segments inhibited Ca2+ currents and neuronal excitability. Antagonists of µ, but not δ receptors, inhibited the effects of these supernatant on Ca2+ currents, whereas both antagonists inhibited their actions on neuronal excitability. CONCLUSIONS AND IMPLICATIONS: A significant number of small diameter colonic nociceptors co-express µ and δ receptors and are inhibited by agonists and endogenous opioids in inflamed tissues. Thus, opioids that act at µ or δ receptors, or their heterodimers may be effective in treating visceral pain.


Asunto(s)
Colon/metabolismo , Nociceptores/metabolismo , Receptores Opioides delta/biosíntesis , Receptores Opioides mu/biosíntesis , Animales , Perfilación de la Expresión Génica , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Opioides delta/genética , Receptores Opioides mu/genética
16.
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
17.
Neuron ; 98(1): 90-108.e5, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29576387

RESUMEN

Cellular interactions between delta and mu opioid receptors (DORs and MORs), including heteromerization, are thought to regulate opioid analgesia. However, the identity of the nociceptive neurons in which such interactions could occur in vivo remains elusive. Here we show that DOR-MOR co-expression is limited to small populations of excitatory interneurons and projection neurons in the spinal cord dorsal horn and unexpectedly predominates in ventral horn motor circuits. Similarly, DOR-MOR co-expression is rare in parabrachial, amygdalar, and cortical brain regions processing nociceptive information. We further demonstrate that in the discrete DOR-MOR co-expressing nociceptive neurons, the two receptors internalize and function independently. Finally, conditional knockout experiments revealed that DORs selectively regulate mechanical pain by controlling the excitability of somatostatin-positive dorsal horn interneurons. Collectively, our results illuminate the functional organization of DORs and MORs in CNS pain circuits and reappraise the importance of DOR-MOR cellular interactions for developing novel opioid analgesics.


Asunto(s)
Células del Asta Anterior/metabolismo , Red Nerviosa/metabolismo , Dolor/metabolismo , Células del Asta Posterior/metabolismo , Receptores Opioides delta/biosíntesis , Receptores Opioides mu/biosíntesis , Animales , Células del Asta Anterior/química , Células del Asta Anterior/patología , Sistema Nervioso Central/química , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/química , Red Nerviosa/patología , Dolor/patología , Dimensión del Dolor/métodos , Células del Asta Posterior/química , Células del Asta Posterior/patología , Receptores Opioides delta/genética , Receptores Opioides mu/genética
18.
J Neurosci ; 26(3): 953-62, 2006 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-16421315

RESUMEN

The present study demonstrates that perikaryaldelta-opioid receptors (deltaORs) in rat dorsal root ganglion (DRG) neurons bind and internalize opioid ligands circulating in the CSF. Using confocal and electron microscopy, we found that prolonged morphine treatment increased the cell surface density of these perikaryal deltaORs and, by way of consequence, receptor-mediated internalization of the fluorescent deltorphin (DLT) analog omega-Bodipy 576/589 deltorphin-I 5-aminopentylamide (Fluo-DLT) in all three types of DRG neurons (small, medium, and large). In contrast, chronic inflammatory pain induced by the injection of complete Freund's adjuvant (CFA) into one hindpaw selectively increased Fluo-DLT internalization in small and medium-sized DRG neurons ipsilateral to the inflammation. Based on our previous studies in the spinal cord of mu-opioid receptor (muOR) knock-out mice, it may be assumed that the enhanced membrane recruitment of deltaORs observed after sustained morphine is attributable to stimulation of muORs. However, the selectivity of the effect induced by inflammatory pain suggests that it involves a different mechanism, namely a modality-specific and pain-related activation of C and Adelta fibers. Indeed, stimulation by capsaicin of transient receptor potential vanilloid 1 receptors, which are selectively expressed by small diameter (< 600 microm2) DRG neurons, increased Fluo-DLT internalization exclusively in this cell population. The present results, therefore, demonstrate that DRG neurons express perikaryal deltaORs accessible to CSF-circulating ligands and that the density and, hence, presumably also the responsiveness, of these receptors may be modulated by both pain-related stimuli and sustained exposure to muOR agonists.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Morfina/farmacología , Dimensión del Dolor/efectos de los fármacos , Receptores Opioides delta/análisis , Receptores Opioides delta/biosíntesis , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ganglios Espinales/citología , Masculino , Morfina/uso terapéutico , Dolor/tratamiento farmacológico , Dolor/metabolismo , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/fisiología
19.
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
20.
J Med Chem ; 49(14): 4044-7, 2006 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-16821764

RESUMEN

We report the computer-aided design, chemical synthesis, and biological evaluation of a novel family of delta opioid receptor (DOR) antagonists containing a 1,2,4-triazole core structure that are structurally distinct from other known opioid receptor active ligands. Among those delta antagonists sharing this core structure, 8 exhibited strong binding affinity (K(i) = 50 nM) for the DOR and appreciable selectivity for delta over mu and kappa opioid receptors (delta/mu = 80; delta/kappa > 200).


Asunto(s)
Receptores Opioides delta/antagonistas & inhibidores , Triazoles/síntesis química , Línea Celular , Humanos , Ligandos , Ensayo de Unión Radioligante , Receptores Opioides delta/biosíntesis , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad , Triazoles/química , Triazoles/farmacología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA