Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurophysiol ; 124(6): 1605-1614, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32966754

RESUMEN

The vestibular system is modulated by various neuromodulators including opioid peptides. The current study was conducted to determine whether activation of nociceptin/orphanin FQ peptide (NOP) receptors modulates voltage-gated calcium currents and action potential discharge of rat vestibular afferent neurons. We performed whole cell patch-clamp recordings on cultured vestibular afferent neurons from P7-P10 Long-Evans rats. Application of nociceptin/orphanin FQ (N/OFQ), a 17-amino acid neuropeptide that is the endogenous ligand for NOP receptor, inhibits the high-voltage activated (HVA) component of the calcium current in a concentration-dependent manner with a half inhibitory concentration of 26 nM. Said inhibitory action on the calcium current is voltage-dependent, which was made clear by the fact that it was reverted in 80% by a depolarizing prepulse. Furthermore, the effect of N/OFQ was blocked by application of the specific NOP-antagonist UFP101, by preincubation with G-protein blocker pertussis toxin, and by coapplication of the specific N-type calcium-current blocker ω-conotoxin-MVIIA. N/OFQ application causes an increase in the duration and maximum rate of repolarization of action potentials. It also decreases repetitive discharge and discharge elicited by sinusoidal stimulation. These results show that in vestibular afferents, NOP receptor activation inhibits N-type calcium current by activating G proteins, mostly through the Gßγ subunit. This suggests that NOP activation produces a presynaptic modulation of primary vestibular afferent neurons' output into the vestibular nuclei, thus taking part in the integration and gain setting of vestibular information in second-order vestibular nucleus neurons.NEW & NOTEWORTHY Our results show that in primary vestibular afferent neurons, activation of the nociceptin/orphanin FQ peptide receptor inhibits the N-type calcium current by a mechanism mediated by G proteins. We propose that calcium current inhibition modulates neurotransmitter release from vestibular afferents, producing a presynaptic modulation of vestibular input to vestibular nuclei, thus contributing to gain control in the vestibular afferent input.


Asunto(s)
Canales de Calcio Tipo N/fisiología , Neuronas/fisiología , Péptidos Opioides/fisiología , Receptores Opioides/fisiología , Vestíbulo del Laberinto/fisiología , Animales , Células Cultivadas , Femenino , Masculino , Potenciales de la Membrana , Neuronas Aferentes/fisiología , Ratas Long-Evans , Receptor de Nociceptina , Nociceptina
2.
Molecules ; 24(22)2019 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-31752279

RESUMEN

The discovery of endogenous peptide ligands for morphine binding sites occurred in parallel with the identification of three subclasses of opioid receptor (OR), traditionally designated as µ, δ, and κ, along with the more recently defined opioid-receptor-like (ORL1) receptor. Early efforts in opioid receptor radiochemistry focused on the structure of the prototype agonist ligand, morphine, although N-[methyl-11C]morphine, -codeine and -heroin did not show significant binding in vivo. [11C]Diprenorphine ([11C]DPN), an orvinol type, non-selective OR antagonist ligand, was among the first successful PET tracers for molecular brain imaging, but has been largely supplanted in research studies by the µ-preferring agonist [11C]carfentanil ([11C]Caf). These two tracers have the property of being displaceable by endogenous opioid peptides in living brain, thus potentially serving in a competition-binding model. Indeed, many clinical PET studies with [11C]DPN or [11C]Caf affirm the release of endogenous opioids in response to painful stimuli. Numerous other PET studies implicate µ-OR signaling in aspects of human personality and vulnerability to drug dependence, but there have been very few clinical PET studies of µORs in neurological disorders. Tracers based on naltrindole, a non-peptide antagonist of the δ-preferring endogenous opioid enkephalin, have been used in PET studies of δORs, and [11C]GR103545 is validated for studies of κORs. Structures such as [11C]NOP-1A show selective binding at ORL-1 receptors in living brain. However, there is scant documentation of δ-, κ-, or ORL1 receptors in healthy human brain or in neurological and psychiatric disorders; here, clinical PET research must catch up with recent progress in radiopharmaceutical chemistry.


Asunto(s)
Imagen Molecular , Receptores Opioides/metabolismo , Animales , Biomarcadores , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encefalopatías/diagnóstico por imagen , Encefalopatías/etiología , Encefalopatías/metabolismo , Humanos , Ligandos , Trastornos Mentales/diagnóstico por imagen , Trastornos Mentales/etiología , Trastornos Mentales/metabolismo , Imagen Molecular/métodos , Neuroimagen/métodos , Péptidos/química , Péptidos/metabolismo , Tomografía de Emisión de Positrones , Trazadores Radiactivos , Receptores Opioides/agonistas , Receptores Opioides/química
3.
Biopolymers ; 106(4): 460-9, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27271345

RESUMEN

We previously showed that an antagonist-based peptide ligand, H-Cys(Npys)-Arg-Tyr-Tyr-Arg- Ile-Lys-NH2 , captures the free thiol groups in the ligand-binding site of the nociceptin receptor ORL1. However, the exact receptor sites of this thiol-disulfide exchange reaction have not been uncovered, although such identification would help to clarify the ligand recognition site. Since the Cys→Ala substitution prevents the reaction, we performed the so-called Ala scanning for all the Cys residues in the transmembrane (TM) domains of the ORL1 receptor. Seven different mutant receptors were soundly expressed in the COS-7 cells and examined for their specific affinity labeling by a competitive binding assay using nociceptin and [(3) H]nociceptin. The results of in vitro Ala scanning analyses revealed that the labeled residues were Cys59 in TM1, Cys215 and Cys231 in TM5, and Cys310 in TM7. The present study has provided a novel method of Cys(Npys)-affinity labeling for identification of the ligand-binding sites in the ORL1 receptor. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 106: 460-469, 2016.


Asunto(s)
Péptidos/química , Receptores Opioides , Coloración y Etiquetado/métodos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Humanos , Ligandos , Mutación Missense , Receptores Opioides/biosíntesis , Receptores Opioides/química , Receptores Opioides/genética , Receptor de Nociceptina
4.
Alcohol Clin Exp Res ; 40(5): 945-54, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27084498

RESUMEN

BACKGROUND: The nociceptin/orphanin-FQ (or opioid receptor-like [ORL1]) receptor (NOP) is localized in the mesolimbic reward pathway and has been suggested to play a role in feeding, mood, stress, and addiction. Since its deorphanization in 1995, there has been a clear dichotomy in the literature regarding whether an agonist or antagonist would provide therapeutic benefit. Specifically, the literature reports indicate that NOP receptor antagonists produce efficacy in animal models of hyperphagia and antidepressant-like activity, whereas NOP agonists produce anxiolytic-like effects and dampen reward/addiction behaviors including ethanol consumption. METHODS: We characterize here the potent, orally bioavailable NOP antagonist, LY2940094, in rodent models of ethanol consumption, including ethanol self-administration, progressive ratio operant self-administration, stress-induced reinstatement of ethanol seeking, and in vivo microdialysis in the nucleus accumbens. RESULTS: LY2940094 dose dependently reduced homecage ethanol self-administration in Indiana alcohol-preferring (P) and Marchigian Sardinian alcohol-preferring (msP) rats, without affecting food/water intake or locomotor activity. Reduced ethanol intake in P rats did not show significant tolerance over 4 days of subchronic dosing. LY2940094 attenuated progressive ratio operant responding and break points for ethanol in P rats. Moreover, stress-induced reinstatement of ethanol seeking in msP rats was completely blocked by LY2940094. Furthermore, LY2940094 blocked ethanol-stimulated dopamine release in response to ethanol challenge (1.1 g/kg, intraperitoneally). CONCLUSIONS: Our findings demonstrate for the first time that blockade of NOP receptors attenuates ethanol self-administration and ethanol-motivated behaviors, stress-induced ethanol seeking, and ethanol-induced stimulation of brain reward pathways in lines of rats that exhibit excessive ethanol consumption. Results suggest that LY2940094 may have potential therapeutic utility in treating alcohol addiction.


Asunto(s)
Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Etanol/antagonistas & inhibidores , Piranos/farmacología , Receptores Opioides/efectos de los fármacos , Compuestos de Espiro/farmacología , Administración Oral , Animales , Condicionamiento Operante/efectos de los fármacos , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Etanol/administración & dosificación , Femenino , Masculino , Microdiálisis , Antagonistas de Narcóticos/farmacología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Piranos/administración & dosificación , Ratas , Ratas Endogámicas , Autoadministración , Compuestos de Espiro/administración & dosificación , Receptor de Nociceptina
5.
J Allergy Clin Immunol ; 133(1): 139-46.e1-10, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24055295

RESUMEN

BACKGROUND: Nonsense mutations in filaggrin (FLG) represent a significant genetic factor in the cause of atopic dermatitis (AD). OBJECTIVE: It is of great importance to find drug candidates that upregulate FLG expression and to determine whether increased FLG expression controls the development of AD. METHODS: We screened a library of bioactives by using an FLG reporter assay to find candidates that promoted FLG mRNA expression using a human immortalized keratinocyte cell line (HaCaT). We studied the effect of the compound on keratinocytes using the human skin equivalent model. We examined the effect of the compound on AD-like skin inflammation in NC/Nga mice. RESULTS: JTC801 promoted FLG mRNA and protein expression in both HaCaT and normal human epidermal keratinocytes. Intriguingly, JTC801 promoted the mRNA and protein expression levels of FLG but not the mRNA levels of other makers for keratinocyte differentiation, including loricrin, keratin 10, and transglutaminase 1, in a human skin equivalent model. In addition, oral administration of JTC801 promoted the protein level of Flg and suppressed the development of AD-like skin inflammation in NC/Nga mice. CONCLUSION: This is the first observation that the compound, which increased FLG expression in human and murine keratinocytes, attenuated the development of AD-like skin inflammation in mice. Our findings provide evidence that modulation of FLG expression can be a novel therapeutic target for AD.


Asunto(s)
Aminoquinolinas/administración & dosificación , Benzamidas/administración & dosificación , Dermatitis Atópica/genética , Proteínas de Filamentos Intermediarios/metabolismo , Queratinocitos/efectos de los fármacos , Aminoquinolinas/farmacología , Animales , Benzamidas/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada , Codón sin Sentido/genética , Dermatitis Atópica/terapia , Proteínas Filagrina , Humanos , Proteínas de Filamentos Intermediarios/genética , Queratina-10/metabolismo , Queratinocitos/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos , Terapia Molecular Dirigida , Péptidos Opioides/antagonistas & inhibidores , Transglutaminasas/metabolismo , Regulación hacia Arriba , Nociceptina
6.
Bioorg Med Chem Lett ; 24(22): 5219-23, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25442316

RESUMEN

Herein we report the identification of (+)-N-(2-((1H-pyrazol-1-yl)methyl)-3-((1R,3r,5S)-6'-fluoro-8-azaspiro[bicyclo[3.2.1]octane-3,1'-isochroman]-8-yl)propyl)-N-[(3)H]-methylacetamide {[(3)H]PF-7191 [(+)-11]} as a promising radiotracer for the nociceptin opioid peptide (NOP) receptor. (+)-11 demonstrated high NOP binding affinity (Ki = 0.1 nM), excellent selectivity over other opioid receptors (>1000×) and good brain permeability in rats (C(b,u)/C(p,u) = 0.29). Subsequent characterization of [(3)H](+)-11 showed a high level of specific binding and a brain bio-distribution pattern consistent with known NOP receptor expression. Furthermore, the in vivo brain binding of [(3)H](+)-11 in rats was inhibited by a selective NOP receptor antagonist in a dose-responsive manner. This overall favorable profile indicated that [(3)H](+)-11 is a robust radiotracer for pre-clinical in vivo receptor occupancy (RO) measurements and a possible substrate for carbon-11 labeling for positron emission tomography (PET) imaging in higher species.


Asunto(s)
Encéfalo/metabolismo , Diseño de Fármacos , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Tritio/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Péptidos Opioides/química , Unión Proteica/fisiología , Ratas , Tritio/química , Receptor de Nociceptina
7.
Clin Pharmacol Drug Dev ; 13(7): 790-800, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38476082

RESUMEN

Sunobinop is an investigational, potent, selective partial agonist at the nociceptin/orphanin FQ peptide receptor in vitro. Three phase 1 studies were conducted to evaluate the safety, tolerability, and pharmacokinetics (PK) of escalating single- and multiple-dose administration of sunobinop in healthy participants. Study 1 was a randomized, double-blind, placebo-controlled, single-ascending dose study. Study 2 was a randomized, double-blind, placebo-controlled, multiple-ascending dose study. Study 3 was a randomized, open-label, single-dose, 4-way crossover study of oral and sublingual sunobinop comparing morning (AM) and bedtime (PM) administration. Seventy participants were included. Systemic exposure (peak plasma concentration [Cmax], area under the plasma concentration-time curve from time 0 to the time of last quantifiable concentration [AUC0-t], and area under the plasma concentration-time curve from time 0 extrapolated to infinity [AUCinf]) of sunobinop was characterized by dose proportionality from 0.6 to 2 mg and increased less than proportionally from 3 to 30 mg. The PKs of sunobinop were similar, regardless of AM or PM administration, for both the oral and sublingual formulations. The majority of absorbed sunobinop was excreted unchanged in the urine within 8 hours of dosing, thereby showing rapid elimination with no appreciable accumulation following 14 consecutive days of once-daily dosing and suggesting exclusive renal elimination. Most treatment-emergent adverse events (TEAEs) were mild in severity; 1 severe TEAE occurred and all TEAEs resolved by the end of the studies. Sunobinop was generally well-tolerated and safe across the range of doses evaluated and presents a clinical profile suitable for continued development.


Asunto(s)
Área Bajo la Curva , Estudios Cruzados , Voluntarios Sanos , Humanos , Masculino , Adulto , Método Doble Ciego , Femenino , Persona de Mediana Edad , Adulto Joven , Administración Oral , Relación Dosis-Respuesta a Droga , Administración Sublingual , Esquema de Medicación , Receptor de Nociceptina , Receptores Opioides/metabolismo , Adolescente , Morfinanos/farmacocinética , Morfinanos/administración & dosificación , Morfinanos/efectos adversos , Naltrexona/análogos & derivados
8.
Res Sq ; 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39070633

RESUMEN

Purpose: The number of ischemic heart failure (HF) patients is growing dramatically worldwide. However, there are at present no preventive treatments for HF. Our previous study showed that Gata4 overexpression improved cardiac function after myocardial infarction in the rat heart. We also found that Gata4 overexpression significantly increased a Pnoc gene expression, an endogenous ligand for cell membrane receptor, ORL1. We hypothesized that an activation of ORL1 receptor would suppress HF in a rat ischemic heart model. Method: Adult Sprague Dawley rats (8 weeks old, 6 males and 6 females) underwent left anterior descending coronary artery ligation. Three weeks later, normal saline or MCOPPB (ORL1 activator, 2.5mg/kg/day) intraperitoneal injection was started, and continued 5 days a week, for 3 months. Echocardiography was performed six times, pre-operative, 3 days after coronary artery ligation, pre-MCOPPB or saline injection, and 1, 2, and 3 months after saline or MCOPPB injection started. Animals were euthanized after 3 months follow up and the heart was harvested for histological analysis. Results: ORL1 activator, MCOPPB, significantly improved cardiac function after myocardial infarction in rat (Ejection fraction, MCOPPB vs saline at euthanasia, 67 ± 3 vs 43 ± 2, p < 0.001). MCOPPB also decreased fibrosis and induced angiogenesis. Conclusion: ORL1 activator, MCOPPB, may be a novel treatment for preventing HF progression.

9.
Peptides ; 169: 171095, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37704079

RESUMEN

This paper is the forty-fifth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2022 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).


Asunto(s)
Analgesia , Analgésicos no Narcóticos , Animales , Humanos , Femenino , Embarazo , Péptidos Opioides/farmacología , Analgésicos Opioides , Tolerancia a Medicamentos
10.
Life Sci ; 328: 121892, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37364634

RESUMEN

The apelin receptor (APJ) and the opioid-related nociceptin receptor 1 (ORL1) are family A G protein-coupled receptors that participate in a variety of physiological processes. The distribution and function of APJ and ORL1 in the nervous system and peripheral tissues are similar; however, the detailed mechanism of how these two receptors modulate signaling and physiological effects remains unclear. Here, we examined whether APJ and ORL1 form dimers, and investigated signal transduction pathways. The endogenous co-expression of APJ and ORL1 in SH-SY5Y cells was confirmed by western blotting and RT-PCR. Bioluminescence and fluorescence resonance energy transfer assays, as well as a proximity ligation assay and co-immunoprecipitation experiments, demonstrated that APJ and ORL1 heterodimerize in HEK293 cells. We found that the APJ-ORL1 heterodimer is selectively activated by apelin-13, which causes the dimer to couple to Gαi proteins and reduce the recruitment of GRKs and ß-arrestins to the dimer. We showed that the APJ-ORL1 dimer exhibits biased signaling, in which G protein-dependent signaling pathways override ß-arrestin-dependent signaling pathways. Our results demonstrate that the structural interface of the APJ-ORL1 dimer switches from transmembrane domain TM1/TM2 in the inactive state to TM5 in the active state. We used mutational analysis and BRET assays to identify key residues in TM5 (APJ L2185.55, APJ I2245.61, and ORL1 L2295.52) responsible for the receptor-receptor interaction. These results provide important information on the APJ-ORL1 heterodimer and may assist the design of new drugs targeting biased signaling pathways for treatment of pain and cardiovascular and metabolic diseases.


Asunto(s)
Neuroblastoma , Humanos , Apelina/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides/genética , Receptores Opioides/metabolismo , Transducción de Señal
11.
Mol Brain ; 15(1): 95, 2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36434658

RESUMEN

Activation of nociceptin opioid peptide receptors (NOP, a.k.a. opioid-like receptor-1, ORL-1) by the ligand nociceptin/orphanin FQ, leads to G protein-dependent regulation of Cav2.2 (N-type) voltage-gated calcium channels (VGCCs). This typically causes a reduction in calcium currents, triggering changes in presynaptic calcium levels and thus neurotransmission. Because of the widespread expression patterns of NOP and VGCCs across multiple brain regions, the dorsal horn of the spinal cord, and the dorsal root ganglia, this results in the alteration of numerous neurophysiological features. Here we review the regulation of N-type calcium channels by the NOP-nociceptin system in the context of neurological conditions such as anxiety, addiction, and pain.


Asunto(s)
Canales de Calcio Tipo N , Enfermedades del Sistema Nervioso , Humanos , Analgésicos Opioides , Calcio , Receptor de Nociceptina
12.
Front Psychiatry ; 9: 430, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30283364

RESUMEN

A series of 14ß-acyl substituted 17-cyclopropylmethyl-7,8-dihydronoroxymorphinone compounds has been synthesized and evaluated for affinity and efficacy for mu (MOP), kappa (KOP), and delta (DOP) opioid receptors and nociceptin/orphanin FQ peptide (NOP) receptors. The majority of the new ligands displayed high binding affinities for the three opioid receptors, and moderate affinity for NOP receptors. The affinities for NOP receptors are of particular interest as most classical opioid ligands do not bind to NOP receptors. The predominant activity in the [35S]GTPγS assay was partial agonism at each receptor. The results are consistent with our prediction that an appropriate 14ß side chain would access a binding site within the NOP receptor and result in substantially higher affinity than displayed by the parent compound naltrexone. Molecular modeling studies, utilizing the recently reported structure of the NOP receptor, are also consistent with this interpretation.

13.
Phytomedicine ; 50: 285-299, 2018 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-30466988

RESUMEN

BACKGROUND: Curcumin (CC) exerts polyvalent pharmacological actions and multi-target effects, including pain relief and anti-nociceptive activity. In combination with Boswellia serrata extract (BS), curcumin shows greater efficacy in knee osteoarthritis management, presumably due to synergistic interaction of the ingredients. AIM: To elucidate the molecular mechanisms underlying the analgesic activity of curcumin and its synergistic interaction with BS. METHODS: We performed gene expression profiling by transcriptome-wide mRNA sequencing in human T98G neuroglia cells treated with CC (Curamed), BS, and the combination of CC and BS (CC-BS; Curamin), followed by interactive pathways analysis of the regulated genes. RESULTS: Treatment with CC and with CC-BS selectively downregulated opioid-related nociceptin receptor 1 gene (OPRL1) expression by 5.9-fold and 7.2-fold, respectively. No changes were detected in the other canonical opioid receptor genes: OPRK1, OPRD1, and OPRM1. Nociceptin reportedly increases the sensation of pain in supra-spinal pain transduction pathways. Thus, CC and CC-BS may downregulate OPRL1, consequently inhibiting production of the nociception receptor NOP, leading to pain relief. In neuroglia cells, CC and CC-BS inhibited signaling pathways related to opioids, neuropathic pain, neuroinflammation, osteoarthritis, and rheumatoid diseases. CC and CC-BS also downregulated ADAM metallopeptidase gene ADAMTS5 expression by 11.2-fold and 13.5-fold, respectively. ADAMTS5 encodes a peptidase that plays a crucial role in osteoarthritis development via inhibition of a corresponding signaling pathway. CONCLUSION: Here, we report for the first time that CC and CC-BS act as nociceptin receptor antagonists, selectively downregulating opioid-related nociceptin receptor 1 gene (OPRL1) expression, which is associated with pain relief. BS alone did not affect OPRL1 expression, but rather appears to potentiate the effects of CC via multiple mechanisms, including synergistic interactions of molecular networks.


Asunto(s)
Analgésicos/farmacología , Curcumina/farmacología , Neuroglía/efectos de los fármacos , Extractos Vegetales/farmacología , Receptores Opioides/metabolismo , Proteína ADAMTS5/metabolismo , Analgésicos Opioides , Boswellia/química , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Antagonistas de Narcóticos/farmacología , Receptor de Nociceptina
14.
J Mol Neurosci ; 66(1): 10-16, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30074175

RESUMEN

Opioid receptor like 1 (ORL1) receptor activation displayed an anti-nociceptive effect at spinal level for acute and neuropathic pain. SCH221510, an orally active non-peptide ORL1 agonist, was reported to be effective in treating neuropathic pain. The present study used ORL1 antagonist and siRNA to investigate that ORL1 activation mediates intrathecal SCH221510 analgesia in neuropathic pain induced by chronic constrictive injury (CCI) to rat sciatic nerve. Paw withdrawal latency and 50% mechanical threshold were measured for thermal and mechanical hypersensitivity in rats. CCI significantly decreased paw withdrawal latency and mechanical threshold. SCH221510 (3, 10, 30 µg) or ORL1 antagonist ([Nphe1]nociceptin(1-13)NH2, 10 µg) was intrathecally injected to test the behavioral effects on neuropathic pain. Intrathecal siRNA was started on 1 day before CCI surgery and maintained for 7 days. L4-L5 spinal cord ORL1 mRNA and protein were measured by real-time PCR and Western blot. The effect of intrathecal siRNA on SCH2210510 was tested in CCI rats on day 7. Intrathecal SCH221510 dose-dependently reduced thermal and mechanical hypersensitivity induced by CCI. [Nphe1]nociceptin(1-13)NH2 blocked SCH221510 analgesia in CCI rats. Intrathecal siRNA blocked ORL1 mRNA and protein increase induced by CCI. Intrathecal ORL1 siRNA did not change thermal and mechanical hypersensitivity induced by nerve injury. Intrathecal siRNA blocked SCH221510 analgesia in neuropathic pain at spinal level. Conclusively, ORL1 activation mediates SCH221510 analgesia in neuropathic pain at spinal level. The results warrant a potential clinically applicable drug in treating neuropathic pain.


Asunto(s)
Analgésicos/farmacología , Compuestos de Azabiciclo/farmacología , Neuralgia/metabolismo , Receptores Opioides/metabolismo , Analgésicos/uso terapéutico , Animales , Compuestos de Azabiciclo/uso terapéutico , Masculino , Neuralgia/tratamiento farmacológico , Péptidos Opioides/farmacología , Fragmentos de Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides/agonistas , Receptores Opioides/genética , Nervio Ciático/efectos de los fármacos , Nervio Ciático/lesiones , Receptor de Nociceptina
15.
Peptides ; 88: 126-188, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28012859

RESUMEN

This paper is the thirty-eighth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2015 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.


Asunto(s)
Conducta Animal , Péptidos Opioides/metabolismo , Dolor/metabolismo , Receptores Opioides/metabolismo , Animales , Ingestión de Alimentos/genética , Humanos , Aprendizaje/fisiología , Memoria/fisiología , Enfermedades del Sistema Nervioso , Péptidos Opioides/genética , Péptidos Opioides/fisiología , Dolor/genética , Receptores Opioides/genética , Receptores Opioides/fisiología , Conducta Sexual/fisiología
16.
Clin Pharmacol Drug Dev ; 6(6): 584-591, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28032481

RESUMEN

The purpose of the present trial was to evaluate safety, tolerability, and effect on systolic blood pressure (SBP) of SER100 in a small group of patients with isolated systolic hypertension (ISH) in treatment with at least 1 antihypertensive drug. Eligible patients were randomized to either SER100 (10 mg) or placebo in a crossover design, and 2 doses were given subcutaneously (SC), 8 hours apart, on 2 consecutive days. On all treatment days patients were monitored with an ambulatory blood pressure measurement device for 12 daytime hours. Seventeen patients completed treatment. There were no serious or severe adverse events. Relative to placebo SER100 induced an average reduction of SBP during the 2 treatment days of 7.0 mm Hg (P = 0.0032), whereas the average reduction of diastolic blood pressure (DBP) over the same period was 3.8 mm Hg (P = 0.0011). For patients with ISH, this short-term cross-over study of SC SER100 demonstrated an acceptable safety profile and consistent, significant lowering of SBP and DBP. As initial clinical proof of concept for a new class of drugs, a nociceptin agonist peptide, the results were encouraging and warrant further research.


Asunto(s)
Antihipertensivos/administración & dosificación , Presión Sanguínea/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Oligopéptidos/administración & dosificación , Anciano , Antihipertensivos/efectos adversos , Antihipertensivos/farmacología , Monitoreo Ambulatorio de la Presión Arterial , Estudios Cruzados , Método Doble Ciego , Femenino , Humanos , Inyecciones Subcutáneas , Masculino , Persona de Mediana Edad , Oligopéptidos/efectos adversos , Oligopéptidos/farmacología , Receptores Opioides/agonistas , Receptor de Nociceptina
17.
Eur J Med Chem ; 114: 345-64, 2016 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-27043173

RESUMEN

Nociceptin/orphanin FQ (N/OFQ) and N/OFQ peptide (NOP) receptor are expressed and distributed in various regions such as central nervous system (CNS), peripheral nervous system, immune system, and peripheral tissues. N/OFQ and NOP receptor have important roles on a variety of physiological, pathophysiological, regulatory, and dysregulatory mechanisms in the living body. Both activation and blockade of NOP receptor function have displayed clinical potential of NOP receptor agonists and antagonists for the treatment of various diseases or pathophysiological conditions, respectively. Potent and selective NOP receptor agonists/antagonists are also useful tools to investigate the various mechanisms mediated by NOP receptor-N/OFQ system. As the present study, a series of (4-arylpiperidine substituted-methyl)-[bicyclic (hetero)cycloalkanobenzene] analogs was designed, synthesized, and biologically evaluated in vitro to seek and identify potent and selective, small-molecules of nonpeptide NOP receptor antagonists, which resulted in the discovery of novel potent small-molecule 15 with high human NOP receptor selectivity over human µ receptor. The structure-activity relationship (SAR) of the potency and selectivity, structure-metabolic stability relationship (SMR), and SAR of hERG (human ether-a-go-go related gene) potassium ion channel binding affinity for the analogs in the present studies in vitro provided or suggested significant and/or useful structural determinants and insights for the respective purposes. The superior profiles of compound 15 are discussed with a viewpoint of multisite interactions between ligand and NOP receptor, together with the results of previous NOP receptor agonist/antagonist studies.


Asunto(s)
Descubrimiento de Drogas , Hidrocarburos Acíclicos/farmacología , Piperidinas/farmacología , Receptores Opioides/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Hidrocarburos Acíclicos/síntesis química , Hidrocarburos Acíclicos/química , Estructura Molecular , Piperidinas/síntesis química , Piperidinas/química , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Receptor de Nociceptina
18.
Chem Biol Drug Des ; 86(4): 447-58, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25546401

RESUMEN

Several spiroxatrine derivatives were synthesized and evaluated as potential NOP receptor ligands. Structural modifications of the 1,4-benzodioxane moiety of spiroxatrine have been the focus of this research project. The structure-activity relationships that emerged indicate that the presence of an H-bond donor group (hydroxyl group) is more favorable for NOP activity when it is positioned α with respect to the CH2 linked to the 1-phenyl-1,3,8-triaza-spiro[4.5]decan-4-one portion. Moreover, cis diastereoisomers of the hydroxyl derivatives4 and 22 show a moderately higher degree of stereoselectivity than trans isomers. In particular, the spiropiperidine derivative cis-4 has submicromolar agonistic activity, and it will be the reference compound for the design and synthesis of new NOP agonists.


Asunto(s)
Compuestos Aza , Receptores Opioides/agonistas , Compuestos de Espiro , Animales , Compuestos Aza/síntesis química , Compuestos Aza/química , Humanos , Ligandos , Estructura Molecular , Compuestos de Espiro/síntesis química , Compuestos de Espiro/química , Relación Estructura-Actividad , Receptor de Nociceptina
19.
Peptides ; 72: 20-33, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25843025

RESUMEN

Functional elucidation of the endogenous opioid system temporally paralleled the creation and growth of the journal, Peptides, under the leadership of its founding editor, Dr. Abba Kastin. He was prescient in publishing annual and uninterrupted reviews on Endogenous Opiates and Behavior that served as a microcosm for the journal under his stewardship. This author published a 2004 review, "Endogenous opioids and feeding behavior: a thirty-year historical perspective", summarizing research in this field between 1974 and 2003. The present review "closes the circle" by reviewing the last 10 years (2004-2014) of research examining the role of endogenous opioids and feeding behavior. The review summarizes effects upon ingestive behavior following administration of opioid receptor agonists, in opioid receptor knockout animals, following administration of general opioid receptor antagonists, following administration of selective mu, delta, kappa and ORL-1 receptor antagonists, and evaluating opioid peptide and opioid receptor changes in different food intake models.


Asunto(s)
Conducta Alimentaria/fisiología , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Animales , Conducta Alimentaria/efectos de los fármacos , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/historia , Receptores Opioides/historia
20.
Structure ; 23(12): 2291-2299, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26526853

RESUMEN

Understanding the mechanism by which ligands affect receptor conformational equilibria is key in accelerating membrane protein structural biology. In the case of G protein-coupled receptors (GPCRs), we currently pursue a brute-force approach for identifying ligands that stabilize receptors and facilitate crystallogenesis. The nociceptin/orphanin FQ peptide receptor (NOP) is a member of the opioid receptor subfamily of GPCRs for which many structurally diverse ligands are available for screening. We observed that antagonist potency is correlated with a ligand's ability to induce receptor stability (Tm) and crystallogenesis. Using this screening strategy, we solved two structures of NOP in complex with top candidate ligands SB-612111 and C-35. Docking studies indicate that while potent, stabilizing antagonists strongly favor a single binding orientation, less potent ligands can adopt multiple binding modes, contributing to their low Tm values. These results suggest a mechanism for ligand-aided crystallogenesis whereby potent antagonists stabilize a single ligand-receptor conformational pair.


Asunto(s)
Receptores Opioides/química , Secuencia de Aminoácidos , Sitios de Unión , Cicloheptanos/farmacología , Células HEK293 , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Piperidinas/farmacología , Unión Proteica , Receptores Opioides/metabolismo , Receptor de Nociceptina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA