Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
1.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884757

RESUMEN

Nociceptin/orphanin FQ (N/OFQ) is a 17-residue neuropeptide that binds the nociceptin opioid-like receptor (NOP). N/OFQ exhibits nucleotidic and aminoacidics sequence homology with the precursors of other opioid neuropeptides but it does not activate either MOP, KOP or DOP receptors. Furthermore, opioid neuropeptides do not activate the NOP receptor. Generally, activation of N/OFQ system exerts anti-opioids effects, for instance toward opioid-induced reward and analgesia. The NOP receptor is widely expressed throughout the brain, whereas N/OFQ localization is confined to brain nuclei that are involved in stress response such as amygdala, BNST and hypothalamus. Decades of studies have delineated the biological role of this system demonstrating its involvement in significant physiological processes such as pain, learning and memory, anxiety, depression, feeding, drug and alcohol dependence. This review discusses the role of this peptidergic system in the modulation of stress and stress-associated psychiatric disorders in particular drug addiction, mood, anxiety and food-related associated-disorders. Emerging preclinical evidence suggests that both NOP agonists and antagonists may represent a effective therapeutic approaches for substances use disorder. Moreover, the current literature suggests that NOP antagonists can be useful to treat depression and feeding-related diseases, such as obesity and binge eating behavior, whereas the activation of NOP receptor by agonists could be a promising tool for anxiety.


Asunto(s)
Péptidos Opioides/fisiología , Receptores Opioides/fisiología , Estrés Fisiológico/fisiología , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/fisiopatología , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Trastornos de Alimentación y de la Ingestión de Alimentos/tratamiento farmacológico , Trastornos de Alimentación y de la Ingestión de Alimentos/fisiopatología , Humanos , Modelos Neurológicos , Trastornos del Humor/tratamiento farmacológico , Trastornos del Humor/fisiopatología , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Recompensa , Estrés Fisiológico/efectos de los fármacos , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Trastornos Relacionados con Sustancias/fisiopatología , Receptor de Nociceptina , Nociceptina
2.
Turk J Med Sci ; 51(4): 2185-2192, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-33862670

RESUMEN

Background/aim: Physical exercise is a state of physiological stress that requires adaptation of the organism to physical activity. Glycogen is an important and essential energy source for muscle contraction. Skeletal muscle and liver are two important glycogen stores, and the energy required to maintain exercise in rodents are provided by destruction of this glycogen depot. In this study, the effects of endogenous opioid peptide antagonism at the central nervous system level on tissue glycogen content after exhaustive exercise were investigated. Materials and methods: Rats had intracerebroventricularly (icv) received nonspecific opioid peptide receptor antagonist, naloxone (50 µg/10 µL in saline) and δ-opioid receptor-selective antagonist naltrindole (50 µg/10 µL in saline) and then exercised till exhaustion. After exhaustion, skeletal muscle, heart, and liver were excised immediately. Results: Both opioid peptide antagonists decreased glycogen levels in skeletal muscle. Although, in soleus muscle, this decrease was not statistically significant (p > 0.05), in gastrocnemius muscle, it was significant in the icv naloxone administered group compared with control (p < 0.05). Heart glycogen levels increased significantly in both naloxone and naltrindole groups compared to control and sham-operated groups (p < 0.05). Heart glycogen levels were higher in the naloxone group than naltrindole (p < 0.05). Liver glycogen levels were elevated significantly with icv naloxone administration compared with the control group (p < 0.05). Glycogen levels in the naloxone group was also significantly higher than the naltrindole group (p < 0.05). Conclusion: Our findings indicate that icv administered opioid peptide antagonists may play a role in glycogen metabolism in peripheral tissues such as skeletal muscle, heart, and liver.


Asunto(s)
Glucógeno/sangre , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/antagonistas & inhibidores , Condicionamiento Físico Animal , Animales , Encefalinas/administración & dosificación , Encefalinas/farmacología , Infusiones Intraventriculares , Naloxona/farmacología , Antagonistas de Narcóticos/administración & dosificación , Neurotransmisores/administración & dosificación , Neurotransmisores/farmacología , Péptidos Opioides/administración & dosificación , Ratas , Receptores Opioides delta
3.
Psychopharmacology (Berl) ; 237(10): 2943-2958, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32588078

RESUMEN

RATIONALE: Evaluation of pharmacotherapies for acute stress disorder (ASD) or post-traumatic stress disorder (PTSD) is challenging due to robust heterogeneity of trauma histories and limited efficacy of any single candidate to reduce all stress-induced effects. Pursuing novel mechanisms, such as the nociceptin/orphanin FQ (NOP) system, may be a viable path for therapeutic development and of interest as it is involved in regulation of relevant behaviors and recently implicated in PTSD and ASD. OBJECTIVES: First, we evaluated NOP receptor antagonism on general behavioral performance and again following a three-species predator exposure model (Experiment 1). Then, we evaluated effects of NOP antagonism on fear memory expression (Experiment 2). METHODS: Adult, male rats underwent daily administration of NOP antagonists (J-113397 or SB-612,111; 0-20 mg/kg, i.p.) and testing in acoustic startle, elevated plus maze, tail-flick, and open field tests. Effects of acute NOP antagonism on behavioral performance following predator exposure were then assessed. Separately, rats underwent fear conditioning and were later administered SB-612,111 (0-3 mg/kg, i.p.) prior to fear memory expression tests. RESULTS: J-113397 and SB-612,111 did not significantly alter most general behavioral performance measures alone, suggesting minimal off-target behavioral effects of NOP antagonism. J-113397 and SB-612,111 restored performance in measures of exploratory behavior (basic movements on the elevated plus maze and total distance in the open field) following predator exposure. Additionally, SB-612,111 significantly reduced freezing behavior relative to control groups across repeated fear memory expression tests, suggesting NOP antagonism may be useful in dampening fear responses. Other measures of general behavioral performance were not significantly altered following predator exposure. CONCLUSIONS: NOP antagonists may be useful as pharmacotherapeutics for dampening fear responses to trauma reminders, and the present results provide supporting evidence for the implication of the NOP system in the neuropathophysiology of dysregulations in fear learning and memory processes observed in trauma- and stress-related disorders.


Asunto(s)
Bencimidazoles/administración & dosificación , Cicloheptanos/administración & dosificación , Miedo/psicología , Péptidos Opioides/antagonistas & inhibidores , Piperidinas/administración & dosificación , Receptores Opioides , Trastornos por Estrés Postraumático/tratamiento farmacológico , Animales , Relación Dosis-Respuesta a Droga , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Miedo/efectos de los fármacos , Miedo/fisiología , Masculino , Memoria/efectos de los fármacos , Memoria/fisiología , Péptidos Opioides/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/psicología , Receptor de Nociceptina , Nociceptina
4.
Peptides ; 132: 170348, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32574695

RESUMEN

This paper is the forty-first consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2018 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 (2), the roles of these opioid peptides and receptors in pain and analgesia in animals (3) and humans (4), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (5), opioid peptide and receptor involvement in tolerance and dependence (6), stress and social status (7), learning and memory (8), eating and drinking (9), drug abuse and alcohol (10), sexual activity and hormones, pregnancy, development and endocrinology (11), mental illness and mood (12), seizures and neurologic disorders (13), electrical-related activity and neurophysiology (14), general activity and locomotion (15), gastrointestinal, renal and hepatic functions (16), cardiovascular responses (17), respiration and thermoregulation (18), and immunological responses (19).


Asunto(s)
Analgésicos Opioides/farmacología , Conducta/efectos de los fármacos , Aprendizaje/fisiología , Trastornos Mentales/tratamiento farmacológico , Péptidos Opioides/metabolismo , Dolor/tratamiento farmacológico , Receptores Opioides/agonistas , Animales , Humanos , Trastornos Mentales/metabolismo , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Péptidos Opioides/farmacología , Dolor/metabolismo
5.
Peptides ; 124: 170223, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31805297

RESUMEN

This paper is the fortieth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2017 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)
Analgésicos Opioides/farmacología , Péptidos Opioides/farmacología , Péptidos Opioides/fisiología , Receptores Opioides/metabolismo , Trastornos Relacionados con Sustancias/etiología , Animales , Dolor en Cáncer/tratamiento farmacológico , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/genética , Ingestión de Alimentos/efectos de los fármacos , Emociones , Femenino , Humanos , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Trastornos Relacionados con Opioides/etiología , Trastornos Relacionados con Opioides/prevención & control , Embarazo , Estrés Fisiológico , Receptor de Nociceptina
6.
Vitam Horm ; 111: 247-279, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31421703

RESUMEN

More than 40years ago, the endogenous opioids were first described. Their role as important neuromodulators of pain and their influence on a variety of neuroendocrine control systems within the central nervous system has been recognized. More recently, endogenous opioids and their receptor have been identified in a variety of reproductive and non-reproductive tissues outside the central nervous system. What role the opioid system plays in these peripheral tissues and organs is not completely understood and thus the subjects of current research. In the central nervous system, endogenous opioids inhibit pulsatile Gonadotropin Releasing Hormone (GnRH) release, affecting the release of gonadotropins from the pituitary, and thus mediating stress response within the central nervous-pituitary-gonadal axes in both women and men-Peripherally, endogenous opioids have been demonstrated to be present-among other organs-in the pancreas and in the ovary, where they are produced by granulosa cells and may influence oocyte maturation. In men, endogenous opioids play a role in sperm production within the testis. Opioid antagonists such as naltrexone have been used to restore cyclicity in women through improvement in insulin resistance, GnRH-pulsatility and hyperandrogenemia stemming from specific pathophysiological conditions such as hypothalamic amenorrhea, polycystic ovarian syndrome, hyperinsulinemia, ovarian hyperstimulation syndrome. Opioid antagonists have also been used to treat male sexual disorders and male infertility. In summary, endogenous opioids exert a variety of actions within the reproductive system which are reviewed in this chapter.


Asunto(s)
Analgésicos Opioides/farmacología , Péptidos Opioides/fisiología , Reproducción/efectos de los fármacos , Reproducción/fisiología , Amenorrea/etiología , Amenorrea/fisiopatología , Animales , Endorfinas/fisiología , Femenino , Humanos , Enfermedades Hipotalámicas/complicaciones , Enfermedades Hipotalámicas/fisiopatología , Masculino , Péptidos Opioides/antagonistas & inhibidores , Oxitocina/fisiología , Síndrome del Ovario Poliquístico/fisiopatología , Embarazo , Prolactina/fisiología , Receptores Opioides/fisiología
7.
J Psychopharmacol ; 33(1): 51-61, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30407114

RESUMEN

BACKGROUND: There is a controversy regarding the key role played by opioid peptide neurotransmission in the modulation of panic-attack-related responses. AIMS: Using a prey versus rattlesnakes paradigm, the present work investigated the involvement of the endogenous opioid peptide-mediated system of the inferior colliculus in the modulation of panic attack-related responses. METHODS: Wistar rats were pretreated with intracollicular administration of either physiological saline or naloxone at different concentrations and confronted with rattlesnakes ( Crotalus durissus terrificus). The prey versus rattlesnake confrontations were performed in a polygonal arena for snakes. The defensive behaviors displayed by prey (defensive attention, defensive immobility, escape response, flat back approach and startle) were recorded twice: firstly, over a period of 15 min the presence of the predator and a re-exposure was performed 24 h after the confrontation, when animals were exposed to the experimental enclosure without the rattlesnake. RESULTS: The intramesencephalic non-specific blockade of opioid receptors with microinjections of naloxone at higher doses decreased both anxiety- (defensive attention and flat back approach) and panic attack-like (defensive immobility and escape) behaviors, evoked in the presence of rattlesnakes and increased non-defensive responses. During the exposure to the experimental context, there was a decrease in duration of defensive attention. CONCLUSIONS: These findings suggest a panicolytic-like effect of endogenous opioid receptors antagonism in the inferior colliculus on innate (panic attack) and conditioned (anticipatory anxiety) fear in rats threatened by rattlesnakes.


Asunto(s)
Miedo/efectos de los fármacos , Colículos Inferiores/efectos de los fármacos , Naloxona/farmacología , Péptidos Opioides/fisiología , Trastorno de Pánico/tratamiento farmacológico , Animales , Reacción de Prevención/efectos de los fármacos , Crotalus , Mecanismos de Defensa , Reacción de Fuga/efectos de los fármacos , Miedo/psicología , Colículos Inferiores/fisiología , Masculino , Péptidos Opioides/antagonistas & inhibidores , Ratas , Ratas Wistar
8.
Br J Pharmacol ; 175(13): 2662-2675, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29582417

RESUMEN

BACKGROUND AND PURPOSE: The nociceptin/orphanin FQ opioid peptide (NOP) receptor system plays a significant role in the regulation of pain. This system functions differently in the spinal cord and brain. The mechanism by which the NOP receptor agonists regulate pain transmission in these regions is not clearly understood. Here, we investigate the peripheral and spinal NOP receptor distribution and antinociceptive effects of intrathecal nociceptin/orphanin FQ (N/OFQ) in chronic neuropathic pain. EXPERIMENTAL APPROACH: We used immunohistochemistry to determine changes in NOP receptor distribution triggered by spinal nerve ligation (SNL) using NOP-eGFP knock-in mice. Antinociceptive effects of intrathecal N/OFQ on SNL-mediated allodynia and heat/cold hyperalgesia were assessed in wild-type mice. KEY RESULTS: NOP-eGFP immunoreactivity was decreased by SNL in the spinal laminae I and II outer, regions that mediate noxious heat stimuli. In contrast, immunoreactivity of NOP-eGFP was unchanged in the ventral border of lamina II inner, which is an important region for the development of allodynia. NOP-eGFP expression was also decreased in a large number of primary afferents in the L4 dorsal root ganglion (DRG) of SNL mice. However, SNL mice showed increased sensitivity, compared to sham animals to the effects of i.t administered N/OFQ with respect to mechanical as well as thermal stimuli. CONCLUSIONS AND IMPLICATIONS: Our findings suggest that the spinal NOP receptor system attenuates injury-induced hyperalgesia by direct inhibition of the projection neurons in the spinal cord that send nociceptive signals to the brain and not by inhibiting presynaptic terminals of DRG neurons in the superficial lamina.


Asunto(s)
Dolor Crónico/tratamiento farmacológico , Modelos Animales de Enfermedad , Péptidos Opioides/antagonistas & inhibidores , Receptores Opioides/análisis , Médula Espinal/química , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Animales , Dolor Crónico/metabolismo , Femenino , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/antagonistas & inhibidores , Proteínas Fluorescentes Verdes/metabolismo , Inyecciones Espinales , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Médula Espinal/efectos de los fármacos , Receptor de Nociceptina
9.
Behav Pharmacol ; 28(7): 521-530, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28704271

RESUMEN

The nociceptin/orphanin FQ peptide (NOP) receptor is believed to have an integral modulatory function in the stress response system. We evaluated the highly selective NOP antagonist J-113397 (7.5 and 20.0 mg/kg), using a predator exposure in which rats were exposed to predator cats as a stressor. A single dose of J-113397 or vehicle was administered (intraperitoneally) shortly before exposure to the predators or a sham exposure. Behavioral impact was measured using elevated plus maze (EPM), open field activity (OFA), and an olfactory discrimination (OD). The predator exposure produced a relatively long-lasting deficit (decreased time in open arms, decreased basic activity) on the EPM while having little effect on performance on the OFA or OD. J-113397 mitigated the performance deficits on the EPM in a dose-dependent manner while having little effect on performance on the OFA or OD. The largest dose of J-113397, administered with a sham exposure, was essentially devoid of effects on the EPM, OFA, and OD. These results demonstrate that J-113397 can significantly and selectively mitigate the effects of a stressor typically used in a preclinical model of post-traumatic stress disorder. Furthermore, these results are consistent with and extend previous results showing that the NOP receptor has an important role in the response to stress and that NOP antagonism may, potentially, have therapeutic benefit in stress disorders.


Asunto(s)
Bencimidazoles/farmacología , Piperidinas/farmacología , Receptores Opioides/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Animales , Ansiedad/tratamiento farmacológico , Bencimidazoles/metabolismo , Gatos , Masculino , Antagonistas de Narcóticos/uso terapéutico , Péptidos Opioides/antagonistas & inhibidores , Péptidos Opioides/metabolismo , Fragmentos de Péptidos/metabolismo , Piperidinas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Trastornos por Estrés Postraumático/tratamiento farmacológico , Estrés Fisiológico/fisiología , Estrés Psicológico/fisiopatología , Receptor de Nociceptina , Nociceptina
10.
Nat Commun ; 8: 15495, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28541285

RESUMEN

Many fearful expectations are shaped by observation of aversive outcomes to others. Yet, the neurochemistry regulating social learning is unknown. Previous research has shown that during direct (Pavlovian) threat learning, information about personally experienced outcomes is regulated by the release of endogenous opioids, and activity within the amygdala and periaqueductal gray (PAG). Here we report that blockade of this opioidergic circuit enhances social threat learning through observation in humans involving activity within the amygdala, midline thalamus and the PAG. In particular, anticipatory responses to learned threat cues (CS) were associated with temporal dynamics in the PAG, coding the observed aversive outcomes to other (observational US). In addition, pharmacological challenge of the opioid receptor function is classified by distinct brain activity patterns during the expression of conditioned threats. Our results reveal an opioidergic circuit that codes the observed aversive outcomes to others into threat responses and long-term memory in the observer.


Asunto(s)
Miedo/fisiología , Péptidos Opioides/fisiología , Sustancia Gris Periacueductal/fisiología , Aprendizaje Social/fisiología , Adulto , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Método Doble Ciego , Miedo/efectos de los fármacos , Neuroimagen Funcional , Humanos , Imagen por Resonancia Magnética , Masculino , Núcleos Talámicos de la Línea Media/efectos de los fármacos , Núcleos Talámicos de la Línea Media/fisiología , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Péptidos Opioides/antagonistas & inhibidores , Sustancia Gris Periacueductal/efectos de los fármacos , Receptores Opioides/fisiología , Aprendizaje Social/efectos de los fármacos
11.
Peptides ; 91: 40-48, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28363796

RESUMEN

We previously reported a series of novel endomorphin analogs with unnatural amino acid modifications. These analogs display good binding affinity and functional activity toward the µ opioid receptor (MOP). In the present study, we further investigated the spinal antinociceptive activity of these compounds. The analogs were potent in several nociceptive models. Opioid antagonists and antibodies against several endogenous opioid peptides were used to determine the mechanisms of action of these peptides. Intrathecal pretreatment with naloxone and ß-funaltrexamine (ß-FNA) effectively inhibited analog-induced analgesia, demonstrating that activity of the analogs is regulated primarily through MOP. Antinociception induced by analog 2 through 4 was not reversed by δ opioid receptor (DOP) or κ opioid receptor (KOP) antagonist; antibodies against dynorphin-A (1-17), dynorphin-B (1-13), and Leu5/Met5-enkephalin had no impact on the antinociceptive effects of these analogs. In contrast, antinociceptive effects induced by a spinal injection of the fluorine substituted analog 1 were significantly reversed by KOP antagonism. Furthermore, intrathecal pretreatment with antibodies against dynorphin-B (1-13) attenuated the antinociceptive effect of analog 1. These results indicate that the antinociceptive activity exerted by intrathecally-administered analog 1 is mediated, in part, through KOP with increased release of dynorphin-B (1-13). The chemical modifications used in the present study may serve as a useful tool to gain insight into the mechanisms of endomorphins activity.


Asunto(s)
Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Oligopéptidos/química , Oligopéptidos/farmacología , Péptidos Opioides/química , Péptidos Opioides/farmacología , Analgesia , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/antagonistas & inhibidores , Análisis de Varianza , Animales , Anticuerpos/inmunología , Dinorfinas/administración & dosificación , Dinorfinas/antagonistas & inhibidores , Dinorfinas/química , Dinorfinas/farmacología , Encefalina Leucina/administración & dosificación , Encefalina Leucina/antagonistas & inhibidores , Encefalina Leucina/química , Encefalina Leucina/farmacología , Encefalina Metionina/administración & dosificación , Encefalina Metionina/antagonistas & inhibidores , Encefalina Metionina/química , Encefalina Metionina/farmacología , Flúor/química , Inyecciones Espinales , Masculino , Ratones , Naloxona/administración & dosificación , Naloxona/farmacología , Naltrexona/administración & dosificación , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Oligopéptidos/administración & dosificación , Oligopéptidos/antagonistas & inhibidores , Péptidos Opioides/administración & dosificación , Péptidos Opioides/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Dolor/metabolismo , Dimensión del Dolor , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores , Receptores sigma/antagonistas & inhibidores
12.
Psychopharmacology (Berl) ; 234(9-10): 1371-1394, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28285326

RESUMEN

BACKGROUND: Nicotine addiction continues to be a health challenge across the world. Despite several approved medications, smokers continue to relapse. Several human and animal studies have evaluated the role of the endogenous opioid system as a potential target for smoking cessation medications. METHODS: In this review, studies that have elucidated the role of the mu (MORs), delta (DORs), and kappa (KORs) opioid receptors in nicotine reward, nicotine withdrawal, and reinstatement of nicotine seeking will be discussed. Additionally, the review will discuss discrepancies in the literature and therapeutic potential of the endogenous opioid system, and suggest studies to address gaps in knowledge with respect to the role of the opioid receptors in nicotine dependence. RESULTS: Data available till date suggest that blockade of the MORs and DORs decreased the rewarding effects of nicotine, while activation of the MORs and DORs decreased nicotine withdrawal-induced aversive effects. In contrast, activation of the KORs decreased the rewarding effects of nicotine, while blockade of the KORs decreased nicotine withdrawal-induced aversive effects. Interestingly, blockade of the MORs and KORs attenuated reinstatement of nicotine seeking. In humans, MOR antagonists have shown benefits in select subpopulations of smokers and further investigation is required to realize their full therapeutic potential. CONCLUSION: Future work must assess the influence of polymorphisms in opioid receptor-linked genes in nicotine dependence, which will help in both identifying individuals vulnerable to nicotine addiction and the development of opioid-based smoking cessation medications. Overall, the endogenous opioid system continues to be a promising target for future smoking cessation medications.


Asunto(s)
Analgésicos Opioides/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Receptores Opioides/metabolismo , Cese del Hábito de Fumar , Tabaquismo/metabolismo , Analgésicos Opioides/administración & dosificación , Animales , Sistemas de Liberación de Medicamentos/métodos , Predicción , Humanos , Nicotina/administración & dosificación , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Péptidos Opioides/metabolismo , Receptores Opioides/agonistas , Recompensa , Cese del Hábito de Fumar/métodos , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/metabolismo , Tabaquismo/tratamiento farmacológico
13.
Pain ; 158(3): 505-515, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28135212

RESUMEN

Drugs able to treat both nociceptive and neuropathic pain effectively without major side effects are lacking. We developed a bifunctional peptide-based hybrid (KGNOP1) that structurally combines a mu-opioid receptor agonist (KGOP1) with antinociceptive activity and a weak nociceptin receptor antagonist (KGNOP3) with anti-neuropathic pain activity. We investigated KGNOP1-related behavioral effects after intravenous administration in rats by assessing thermal nociception, cold hyperalgesia in a model of neuropathic pain induced by chronic constriction injury of the sciatic nerve, and plethysmography parameters including inspiratory time (TI) and minute ventilation (VM) in comparison to the well-known opioid analgesics, tramadol and morphine. Time-course and dose-dependent effects were investigated for all behavioral parameters to determine the effective doses 50% (ED50). Pain-related effects on cold hyperalgesia were markedly increased by KGNOP1 as compared to KGNOP3 and tramadol (ED50: 0.0004, 0.32, and 12.1 µmol/kg, respectively), whereas effects on thermal nociception were significantly higher with KGNOP1 as compared to morphine (ED50: 0.41 and 14.7 µmol/kg, respectively). KGNOP1 and KGOP1 produced a larger increase in TI and deleterious decrease in VM in comparison to morphine and tramadol (ED50(TI): 0.63, 0.52, 12.2, and 50.9 µmol/kg; ED50(VM): 0.57, 0.66, 10.6, and 50.0 µmol/kg, respectively). Interestingly, the calculated ratios of anti-neuropathic pain/antinociceptive to respiratory effects revealed that KGNOP1 was safer than tramadol (ED50 ratio: 5.44 × 10 vs 0.24) and morphine (ED50 ratio: 0.72 vs 1.39). We conclude that KGNOP1 is able to treat both experimental neuropathic and nociceptive pain, more efficiently and safely than tramadol and morphine, respectively, and thus should be a candidate for future clinical developments.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Neuralgia/tratamiento farmacológico , Dolor Nociceptivo/tratamiento farmacológico , Péptidos Opioides/antagonistas & inhibidores , Péptidos/uso terapéutico , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hiperalgesia/tratamiento farmacológico , Indoles/uso terapéutico , Ligandos , Masculino , Oligopéptidos/química , Oligopéptidos/uso terapéutico , Dimensión del Dolor , Fenalenos/uso terapéutico , Pletismografía , Ratas , Ratas Sprague-Dawley , Receptores Opioides , Respiración/efectos de los fármacos , Factores de Tiempo , Tramadol/uso terapéutico , Nociceptina
14.
Proc Natl Acad Sci U S A ; 113(37): E5511-8, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27573832

RESUMEN

Despite the critical need, no previous research has substantiated safe opioid analgesics without abuse liability in primates. Recent advances in medicinal chemistry have led to the development of ligands with mixed mu opioid peptide (MOP)/nociceptin-orphanin FQ peptide (NOP) receptor agonist activity to achieve this objective. BU08028 is a novel orvinol analog that displays a similar binding profile to buprenorphine with improved affinity and efficacy at NOP receptors. The aim of this preclinical study was to establish the functional profile of BU08028 in monkeys using clinically used MOP receptor agonists for side-by-side comparisons in various well-honed behavioral and physiological assays. Systemic BU08028 (0.001-0.01 mg/kg) produced potent long-lasting (i.e., >24 h) antinociceptive and antiallodynic effects, which were blocked by MOP or NOP receptor antagonists. More importantly, the reinforcing strength of BU08028 was significantly lower than that of cocaine, remifentanil, or buprenorphine in monkeys responding under a progressive-ratio schedule of drug self-administration. Unlike MOP receptor agonists, BU08028 at antinociceptive doses and ∼10- to 30-fold higher doses did not cause respiratory depression or cardiovascular adverse events as measured by telemetry devices. After repeated administration, the monkeys developed acute physical dependence on morphine, as manifested by precipitated withdrawal signs, such as increased respiratory rate, heart rate, and blood pressure. In contrast, monkeys did not show physical dependence on BU08028. These in vivo findings in primates not only document the efficacy and tolerability profile of bifunctional MOP/NOP receptor agonists, but also provide a means of translating such ligands into therapies as safe and potentially abuse-free opioid analgesics.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Buprenorfina/análogos & derivados , Cocaína/toxicidad , Dolor/tratamiento farmacológico , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/agonistas , Analgésicos Opioides/antagonistas & inhibidores , Animales , Buprenorfina/administración & dosificación , Buprenorfina/efectos adversos , Buprenorfina/química , Buprenorfina/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Humanos , Ligandos , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Dolor/patología , Primates , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/patología
15.
Psychopharmacology (Berl) ; 233(13): 2525-32, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27129865

RESUMEN

RATIONALE: Pharmacological and genetic evidence support antidepressant-like effects elicited by the blockade of the NOP receptor. The learned helplessness (LH) model employs uncontrollable and unpredictable electric footshocks as a stressor stimulus to induce a depressive-like phenotype that can be reversed by classical antidepressants. OBJECTIVES: The present study aimed to evaluate the action of NOP receptor antagonists in helpless mice. METHODS: Male Swiss mice were subjected to the three steps of the LH paradigm (i.e., (1) induction, (2) screening, and (3) test). Only helpless animals were subjected to the test session. During the test session, animals were placed in the electrified chamber and the latency to escape after the footshock and the frequency of escape failures were recorded. The effect of the following treatments administered before the test session were evaluated: nortriptyline (30 mg/kg, ip, 60 min), fluoxetine (30 mg/kg, ip, four consecutive days of treatment), and NOP antagonists SB-612111 (1-10 mg/kg, ip, 30 min) and UFP-101 (1-10 nmol, icv, 5 min). To rule out possible biases, the effects of treatments on controllable stressful and non stressful situations were assessed. RESULTS: In helpless mice, nortriptyline, fluoxetine, UFP-101 (3-10 nmol), and SB-612111 (3-10 mg/kg) significantly reduced escape latencies and escape failures. No effects of drug treatments were observed in mice subjected to the controllable electric footshocks and non stressful situations. CONCLUSIONS: Acute treatment with NOP antagonists reversed helplessness similarly to the classical antidepressants. These findings support the proposal that NOP receptor antagonists are worthy of development as innovative antidepressant drugs.


Asunto(s)
Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Cicloheptanos/farmacología , Fluoxetina/farmacología , Desamparo Adquirido , Antagonistas de Narcóticos/farmacología , Nortriptilina/farmacología , Péptidos Opioides/farmacología , Piperidinas/farmacología , Animales , Trastorno Depresivo/tratamiento farmacológico , Modelos Animales de Enfermedad , Masculino , Ratones , Péptidos Opioides/antagonistas & inhibidores , Receptores Opioides , Transducción de Señal/efectos de los fármacos , Receptor de Nociceptina , Nociceptina
16.
Emotion ; 15(4): 494-500, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26098729

RESUMEN

"Heartwarming" social experiences, when one feels interpersonally connected to others, have recently been linked with physical warmth. According to one theory (Panksepp, 1998), "social warmth" and physical warmth may be closely linked because both experiences are supported by similar neurobiological mechanisms; however, the neurochemical substrates underlying this overlap have not been explored. Here, an opioid antagonist, naltrexone, was administered in order to examine the role of opioids, previously shown to alter temperature and social bonding behavior, on perceived thermal intensity, general positive affect, and feelings of social connection from physical warmth. Thirty-one participants took both naltrexone and a placebo and completed a temperature manipulation task (held a warm pack, cold pack, and neutral object) while on each drug. Replicating previous research, holding a warm (vs. a cold or neutral) object increased feelings of social connection. Moreover, blocking opioids reduced this effect. Hence, naltrexone specifically reduced feelings of social connection to holding a warm (vs. neutral) object but not to holding a cold (vs. neutral) object. These results lend further support to the theory that social and physical warmth share neurobiological, opioid receptor dependent mechanisms.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Emociones/efectos de los fármacos , Calor , Apego a Objetos , Péptidos Opioides/antagonistas & inhibidores , Receptores Opioides/metabolismo , Conducta Social , Adolescente , Adulto , Femenino , Humanos , Masculino , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/metabolismo , Adulto Joven
17.
Vitam Horm ; 97: 147-65, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25677771

RESUMEN

Nociceptin/orphanin FQ (N/OFQ) and nocistatin (NST) are neuropeptides produced from the same precursor protein. N/OFQ is involved in a broad range of central functions including pain, learning, memory, anxiety, and feeding. However, NST has opposite effects on various central functions evoked by N/OFQ. The regulation of their receptors may be important for these opposite functions of NST and N/OFQ. Although N/OFQ binds to a specific N/OFQ receptor, the target molecule of NST remains unclear. Some biological effects of NST are mediated by a G protein-coupled receptor. Furthermore, using high-performance affinity nanobeads, we recently identified a 4-nitrophenylphosphatase domain and nonneuronal SNAP25-like protein homolog 1 (NIPSNAP1) as a protein that interacts with NST in the mouse spinal cord. The inhibition of N/OFQ-evoked tactile pain allodynia by NST is mediated by NIPSNAP1. This review focuses on the molecular mechanisms of pain regulation by the target molecules of NST including a G protein-coupled receptor and NIPSNAP1.


Asunto(s)
Drogas en Investigación/farmacología , Antagonistas de Narcóticos/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Péptidos Opioides/antagonistas & inhibidores , Proteínas/metabolismo , Receptores Opioides/metabolismo , Animales , Drogas en Investigación/química , Drogas en Investigación/metabolismo , Drogas en Investigación/uso terapéutico , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ligandos , Antagonistas de Narcóticos/química , Antagonistas de Narcóticos/metabolismo , Antagonistas de Narcóticos/uso terapéutico , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Neuronas/metabolismo , Péptidos Opioides/metabolismo , Dolor/tratamiento farmacológico , Dolor/metabolismo , Proteínas/agonistas , Proteínas/antagonistas & inhibidores , Receptores Opioides/agonistas , Receptores Opioides/química , Transmisión Sináptica/efectos de los fármacos
18.
Vitam Horm ; 97: 167-86, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25677772

RESUMEN

Phosphorylated Rec8, a key component of cohesin, mediates the association and disassociation, "dynamics," of chromosomes occurring in synaptonemal complex formation, crossover recombination, and sister chromatid cohesion during meiosis in germ cells. Yet, the extrinsic factors triggering meiotic chromosome dynamics remained unclear. In postnatal testes, follicle-stimulating hormone (FSH) acts directly on somatic Sertoli cells to activate gene expression via an intracellular signaling pathway composed of cAMP, cAMP-dependent protein kinase (PKA), and cAMP-response element-binding protein (CREB), and promotes germ cell development and spermatogenesis indirectly. Yet, the paracrine factors mediating the FSH effects to germ cells remained elusive. We have shown that nociceptin, known as a neuropeptide, is upregulated by FSH signaling through cAMP/PKA/CREB pathway in Sertoli cells of postnatal murine testes. Chromatin immunoprecipitation from Sertoli cells demonstrated that CREB phosphorylated at Ser133 associates with prepronociceptin gene encoding nociceptin. Analyses with Sertoli cells and testes revealed that both prepronociceptin mRNA and the nociceptin peptide are induced after FSH signaling is activated. In addition, the nociceptin peptide is induced in testes after 9 days post partum following FSH surge. Thus, our findings may identify nociceptin as a novel paracrine mediator of the FSH effects in the regulation of spermatogenesis; however, very little has known about the functional role of nociceptin in spermatogenesis. We have shown that nociceptin induces Rec8 phosphorylation, triggering chromosome dynamics, during meiosis in spermatocytes of postnatal murine testes. The nociceptin receptor Oprl-1 is exclusively expressed in the plasma membrane of testicular germ cells, mostly spermatocytes. Treatment of testes with nociceptin resulted in a rapid phosphorylation of Rec8. Injection of nociceptin into mice stimulated Rec8 phosphorylation and meiotic chromosome dynamics in testes, whereas injection of nocistatin, a specific inhibitor for nociceptin, abolished them. Therefore, our findings suggest that nociceptin is a novel extrinsic factor that plays a crucial role in the progress of meiosis during spermatogenesis.


Asunto(s)
Péptidos Opioides/metabolismo , Espermatogénesis , Testículo/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Humanos , Masculino , Meiosis , Péptidos Opioides/antagonistas & inhibidores , Fosforilación , Procesamiento Proteico-Postraduccional , Células de Sertoli/citología , Células de Sertoli/metabolismo , Testículo/citología , Testículo/crecimiento & desarrollo , Nociceptina
19.
Gen Comp Endocrinol ; 210: 81-6, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25307952

RESUMEN

Endocannabinoids and their receptors are found throughout the brain of all vertebrates. By virtue of their wide distribution, endocannabinoids have the potential to affect many behaviors. Prior research has shown that cannabinoids inhibit courtship-clasping and mediate behavioral responses to stress in male rough-skinned newts, Taricha granulosa, and cannabinoid signaling is initiated by rapid actions of the steroid corticosterone (CORT) at its specific membrane receptor (mCR). This same mCR also recognizes κ-opioid receptor agonists and antagonists. Prior behavioral studies show that κ-opioid agonists suppress clasping behavior in a dose dependent manner. Combined, these studies suggest that κ-opioid agonists might suppress clasping behavior via the same pathway initiated by CORT: up-regulation of endocannabinoid signaling. We examined whether pretreatment with a CB1 antagonist, AM281, would block κ-opioid-mediated suppression of clasping. We found that the CB1 antagonist did not reverse κ-opioid-induced suppression of clasping, revealing that while endocannabinoids mediate CORT-induced suppression of clasping, endocannabinoids do not mediate the κ-opioid-induced suppression of clasping.


Asunto(s)
Corticosterona/farmacología , Sistemas Neurosecretores/efectos de los fármacos , Péptidos Opioides/farmacología , Salamandridae , Conducta Sexual Animal/efectos de los fármacos , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos Opioides/metabolismo , Animales , Encéfalo/efectos de los fármacos , Cortejo , Regulación hacia Abajo/efectos de los fármacos , Endocannabinoides/farmacología , Femenino , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Sistemas Neurosecretores/fisiología , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Salamandridae/fisiología , Conducta Sexual Animal/fisiología
20.
Addict Biol ; 20(4): 643-51, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24930632

RESUMEN

Dysregulation of the nociceptin (N/OFQ) system has been implicated in alcohol abuse and alcoholism, and growing evidence suggests that targeting this system may be beneficial for treating alcoholism. To further explore the treatment target potential of the N/OFQ system, the novel non-peptide, small-molecule N/OFQ (NOP) agonist MT-7716, (R)-2-{3-[1-(Acenaphthen-1-yl)piperidin-4-yl]-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl}-N-methylacetamide hydrochloride hydrate, was examined for its effects on ethanol self-administration and stress-induced reinstatement of alcohol seeking in non-dependent and post-dependent rats. Male Wistar rats were trained to self-administer ethanol and then made ethanol dependent via repeated intragastric ethanol intubation. The effects of MT-7716 (0.3 and 1 mg/kg; PO) on alcohol self-administration were determined 2 weeks following dependence induction, when baseline self-administration was restored. Effects of MT-7716 on stress-induced reinstatement were tested in separate cohorts of rats, 1 and 3 weeks post-withdrawal. MT-7716 reduced alcohol self-administration and stress-induced reinstatement of alcohol seeking in post-dependent rats, but was ineffective in non-dependent animals. Moreover, the prevention of stress-induced reinstatement by MT-7716 was more pronounced at 3 weeks post-dependence. The results further confirm treatment target potential for the NOP receptor and identify non-peptide NOP agonists as promising potential treatment drugs for alcohol abuse and relapse prevention. The findings also support dysregulation of the N/OFQ system as a factor in alcohol seeking and reinforcement.


Asunto(s)
Acenaftenos/farmacología , Alcoholismo/prevención & control , Bencimidazoles/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/antagonistas & inhibidores , Receptores Opioides , Refuerzo en Psicología , Alcoholismo/sangre , Análisis de Varianza , Animales , Condicionamiento Operante , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Etanol/metabolismo , Masculino , Nocicepción/efectos de los fármacos , Ratas Wistar , Receptores Opioides/efectos de los fármacos , Autoadministración , Estrés Psicológico/psicología , Receptor de Nociceptina , Nociceptina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA