Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
1.
J Pharmacol Exp Ther ; 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38670800

RESUMEN

Recent studies suggest that amongst the GABAA receptor subtype heterogeneity, α2/α3 subunits of GABAA receptors mediate pain processing. Therefore, α2/α3-subtype selective GABAA receptor positive allosteric modulators (PAMs) may be candidate analgesics. Antinociceptive effects of α2/α3-subtype selective GABAA receptor PAMs have been reported, but the behavioral effects of these compounds have not been systematically evaluated. This study examined the behavioral effects of two α2/α3 subtype-selective GABAA receptor PAMs, KRM-II-81 and NS16085, in male rats. The antinociceptive effects of KRM-II-81 and NS16085 were examined using rat models of inflammatory (complete Freund's adjuvant) and neuropathic pain (chronic constriction injury). The effect of KRM-II-81 on affective pain was measured using the place escape/avoidance paradigm (PEAP). Rate-response of food-maintained operant responding, horizontal wire test, and the spontaneous alternation T-maze, were assessed to study the side-effect profiles of KRM-II-81 and NS16085. The benzodiazepine midazolam was used as a comparator in these studies. KRM-II-81 and NS16085 attenuated mechanical allodynia but not thermal hyperalgesia in both pain states, and their effects were attenuated by the benzodiazepine receptor antagonist flumazenil. KRM-II-81 attenuated affective pain-related behavior in the PEAP test. In the operant responding procedure and horizontal wire test, only midazolam produced significant effects at the dose that produced maximal antinociception. In the T-maze assay, only midazolam significantly decreased the percentage of alternation at an antinociceptive dose. Thus, KRM-II-81 and NS16085 but not midazolam selectively produced antinociceptive effects. Collectively, these data suggest that α2/α3-subtype selective GABAA PAMs could be a novel class of analgesics and warrant further investigation. Significance Statement This study demonstrates that α2/α3-subtype selective GABAA PAMs KRM-II-81 and NS16085 produce selective antinociceptive effects devoid of sedation, myorelaxation, cognitive impairment in two rat models of persistent pain. Unlikely classical benzodiazepines, this study supports the development of α2/α3-subtype selective GABAA PAMs as safe and novel analgesics for pain management.

2.
Mol Psychiatry ; 27(4): 2136-2145, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35079125

RESUMEN

Relapse remains a major challenge to the treatment of cocaine addiction. Recent studies suggested that the trace amine-associated receptor 1 (TAAR1) could be a promising target to treat cocaine addiction and relapse; however, the underlying mechanism remains unclear. Here, we aimed to investigate the neural mechanism underlying the role of TAAR1 in the drug priming-induced reinstatement of cocaine-seeking behavior in rats, an animal model of cocaine relapse. We focused on the shell subregion of nucleus accumbens (NAc), a key brain region of the brain reward system. We found that activation of TAAR1 by systemic and intra-NAc shell administration of the selective TAAR1 agonist RO5166017 attenuated drug-induced reinstatement of cocaine-seeking and prevented drug priming-induced CaMKIIα activity in the NAc shell. Activation of TAAR1 dampened the CaMKIIα/GluR1 signaling pathway in the NAc shell and reduced AMPAR-EPSCs on the NAc slice. Microinjection of the selective TAAR1 antagonist EPPTB into the NAc shell enhanced drug-induced reinstatement as well as potentiated CaMKIIα activity in the NAc shell. Furthermore, viral-mediated expression of CaMKIIα in the NAc shell prevented the behavioral effects of TAAR1 activation. Taken together, our findings indicate that TAAR1 regulates drug-induced reinstatement of cocaine-seeking by negatively regulating CaMKIIα activity in the NAc. Our findings elucidate a novel mechanism of TAAR1 in regulating drug-induced reinstatement of cocaine-seeking and further suggests that TAAR1 is a promising target for the treatment of cocaine relapse.


Asunto(s)
Trastornos Relacionados con Cocaína , Cocaína , Animales , Cocaína/farmacología , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Trastornos Relacionados con Cocaína/metabolismo , Comportamiento de Búsqueda de Drogas , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G , Recurrencia , Autoadministración
3.
Pharmacol Rev ; 72(1): 50-79, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31819014

RESUMEN

Imidazoline receptors historically referred to a family of nonadrenergic binding sites that recognize compounds with an imidazoline moiety, although this has proven to be an oversimplification. For example, none of the proposed endogenous ligands for imidazoline receptors contain an imidazoline moiety but they are diverse in their chemical structure. Three receptor subtypes (I1, I2, and I3) have been proposed and the understanding of each has seen differing progress over the decades. I1 receptors partially mediate the central hypotensive effects of clonidine-like drugs. Moxonidine and rilmenidine have better therapeutic profiles (fewer side effects) than clonidine as antihypertensive drugs, thought to be due to their higher I1/α 2-adrenoceptor selectivity. Newer I1 receptor agonists such as LNP599 [3-chloro-2-methyl-phenyl)-(4-methyl-4,5-dihydro-3H-pyrrol-2-yl)-amine hydrochloride] have little to no activity on α 2-adrenoceptors and demonstrate promising therapeutic potential for hypertension and metabolic syndrome. I2 receptors associate with several distinct proteins, but the identities of these proteins remain elusive. I2 receptor agonists have demonstrated various centrally mediated effects including antinociception and neuroprotection. A new I2 receptor agonist, CR4056 [2-phenyl-6-(1H-imidazol-1yl) quinazoline], demonstrated clear analgesic activity in a recently completed phase II clinical trial and holds great promise as a novel I2 receptor-based first-in-class nonopioid analgesic. The understanding of I3 receptors is relatively limited. Existing data suggest that I3 receptors may represent a binding site at the Kir6.2-subtype ATP-sensitive potassium channels in pancreatic ß-cells and may be involved in insulin secretion. Despite the elusive nature of their molecular identities, recent progress on drug discovery targeting imidazoline receptors (I1 and I2) demonstrates the exciting potential of these compounds to elicit neuroprotection and to treat various disorders such as hypertension, metabolic syndrome, and chronic pain.


Asunto(s)
Receptores de Imidazolina/metabolismo , Imidazolinas/metabolismo , Imidazolinas/farmacología , Animales , Antihipertensivos/farmacología , Antihipertensivos/uso terapéutico , Clonidina/farmacología , Clonidina/uso terapéutico , Humanos , Imidazoles/farmacología , Imidazoles/uso terapéutico , Ligandos , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto
4.
Brain Behav Immun ; 101: 37-48, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34958862

RESUMEN

Opioid addiction remains a severe health problem. While substantial insights underlying opioid addiction have been yielded from neuron-centric studies, the contribution of non-neuronal mechanisms to opioid-related behavioral adaptations has begun to be recognized. Toll-like receptor 4 (TLR4), a pattern recognition receptor, has been widely suggested in opioid-related behaviors. Interleukin-1 receptor-associated kinase 4 (IRAK4) is a kinase essential for TLR4 responses, However, the potential role of IRAK4 in opioid-related responses has not been examined. Here, we explored the role of IRAK4 in cue-induced opioid-seeking behavior in male rats. We found that morphine self-administration increased the phosphorylation level of IRAK4 in the nucleus accumbens (NAc) in rats; the IRAK4 signaling remained activated after morphine extinction and cue-induced reinstatement test. Both systemic and local inhibition of IRAK4 in the NAc core attenuated cue-induced morphine-seeking behavior without affecting the locomotor activity and cue-induced sucrose-seeking. In addition, inhibition of IRAK4 also reduced the cue-induced reinstatement of fentanyl-seeking. Our findings suggest an important role of IRAK4 in opioid relapse-like behaviors and provide novel evidence in the association between innate immunity and drug addiction.


Asunto(s)
Comportamiento de Búsqueda de Drogas , Quinasas Asociadas a Receptores de Interleucina-1 , Núcleo Accumbens , Trastornos Relacionados con Opioides , Analgésicos Opioides/farmacología , Animales , Señales (Psicología) , Extinción Psicológica , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Masculino , Morfina/farmacología , Proteínas Serina-Treonina Quinasas , Ratas , Ratas Sprague-Dawley , Receptor Toll-Like 4
5.
Addict Biol ; 27(1): e13075, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34170054

RESUMEN

Nicotine addiction is a leading avoidable brain disorder globally. Although nicotine induces a modest reinforcing effect, which is important for the initial drug use, the transition from nicotine use to nicotine addiction involves the mechanisms responsible for the negative consequences of drug abstinence. Recent study suggested that trace amine-associated receptor 1 (TAAR1) is a promising pharmacological target for the modulation of positive reinforcing effects of nicotine. However, whether TAAR1 plays a part in the negative reinforcement of nicotine withdrawal remains to be determined. Here, using a long-access (LA) self-administration model, we investigated whether LA rats show increased nicotine intake and withdrawal symptoms in comparison with saline and ShA rats and then tested the effect of TAAR1 partial agonist RO5263397 on nicotine withdrawal effects. We found that rats from long-access group showed significant abstinence-induced anxiety-like behaviour, mechanic hypersensitivity, increased number of precipitated withdrawal signs and higher motivation for the drug, while rats from short-access did not differ from saline group. TAAR1 partial agonist RO5263397 significantly reduced the physical and motivational withdrawal effects of nicotine in LA rats, as reflected by increased time spent on the open arm in the elevated plus maze (EPM) test, normalized paw withdrawal threshold, decreased withdrawal signs and motivation to self-administer nicotine. This study indicates that activation of TAAR1 attenuates the negative-reinforcing effects of nicotine withdrawal and further suggests TAAR1 as a promising target to treat nicotine addiction.


Asunto(s)
Nicotina/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Tabaquismo/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Masculino , Oxazoles , Ratas , Refuerzo en Psicología , Autoadministración
6.
Handb Exp Pharmacol ; 276: 275-290, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35434747

RESUMEN

Drug addiction is a chronic brain disease characterized by compulsive drug-seeking and drug-taking behaviors despite the major negative consequences. Current well-established neuronal underpinnings of drug addiction have promoted the substantial progress in understanding this disorder. However, non-neuronal mechanisms of drug addiction have long been underestimated. Fortunately, increased evidence indicates that neuroimmune system, especially Toll-like receptor 4 (TLR4) signaling, plays an important role in the different stages of drug addiction. Drugs like opioids, psychostimulants, and alcohol activate TLR4 signaling and enhance the proinflammatory response, which is associated with drug reward-related behaviors. While extensive studies have shown that inhibition of TLR4 attenuated drug-related responses, there are conflicting findings implicating that TLR4 signaling may not be essential to drug addiction. In this chapter, preclinical and clinical studies will be discussed to further evaluate whether TLR4-based neuroimmune pharmacotherapy can be used to treat drug addiction. Furthermore, the possible mechanisms underlying the effects of TLR4 inhibition in modulating drug-related behaviors will also be discussed.


Asunto(s)
Comportamiento de Búsqueda de Drogas , Trastornos Relacionados con Sustancias , Receptor Toll-Like 4 , Analgésicos Opioides/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Etanol/farmacología , Humanos , Transducción de Señal , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Trastornos Relacionados con Sustancias/genética , Receptor Toll-Like 4/antagonistas & inhibidores
7.
Molecules ; 27(11)2022 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-35684506

RESUMEN

Oxypeucedanin, a furanocoumarin extracted from many traditional Chinese herbal medicines, has a variety of pharmacological effects. However, the independent pharmacokinetic characteristics and bioavailability of this compound remains elusive. In this study, a rapid, sensitive, and selective method using ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC/MS/MS) was developed for evaluating the intravenous and oral pharmacokinetics of oxypeucedanin. After intravenous administration of oxypeucedanin (2.5, 5, and 10 mg/kg), and intragastric administration of oxypeucedanin (20 mg/kg), blood samples were collected periodically from the tail vein. The plasma concentration-time curves were plotted, and the pharmacokinetic parameters were calculated using a non-compartmental model analysis. After intravenous administration of oxypeucedanin (single dosing at 2.5, 5, and 10 mg/kg) to rats, the pharmacokinetics fit the linear kinetics characteristics, which showed that some parameters including average elimination half-life (T1/2Z of 0.61~0.66 h), mean residence time (MRT of 0.62~0.80 h), apparent volume of distribution (VZ of 4.98~7.50 L/kg), and systemic clearance (CLZ of 5.64~8.55 L/kg/h) are dose-independent and the area under concentration-time curve (AUC) increased in a dose-proportional manner. Single oral administration of oxypeucedanin (20 mg/kg) showed poor and slow absorption with the mean time to reach the peak concentration (Tmax) of 3.38 h, MRT of 5.86 h, T1/2Z of 2.94 h, and a mean absolute bioavailability of 10.26% in rats. These results provide critical information for a better understanding of the pharmacological effect of oxypeucedanin, which will facilitate its research and development.


Asunto(s)
Furocumarinas , Espectrometría de Masas en Tándem , Administración Intravenosa , Administración Oral , Animales , Disponibilidad Biológica , Cromatografía Líquida de Alta Presión , Ratas , Espectrometría de Masas en Tándem/métodos
8.
Neurochem Res ; 46(5): 1068-1080, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33683629

RESUMEN

Alzheimer's disease (AD) process is characterized classically by two hallmark pathologies: ß-amyloid (Aß) plaque deposition and neurofibrillary tangles of hyperphosphorylated tau. Aß peptides play an important role in AD, but despite much effort the molecular mechanisms of how Aß contributes to AD remain unclear. The present study evaluated the effects of the active components of Epimedium, Astragalus and Radix Puerariae induced HAMP on key enzymes in the hydrolysis of APP in HT22 cells. The active components of Epimedium, Astragalus and Radix Puerariae could effectively up-regulate the expression of HAMP, alleviate the iron overload in the brain tissues of mice, significantly improve the learning and memory ability of AD, down-regulate the expression of Aß and reduce the deposition of SP in an APPswe/PS1ΔE9 transgenic mouse model of AD. HAMP and Aß25-35 induced HT22 cells are used as AD cell models in this study to investigate the effect of the compound consisting of the effective components of Epimedium, Astragalus and Pueraria on the key enzymes in the hydrolysis of APP. After the administration of traditional Chinese medicine (TCM), the expression levels of ADAM10 and ADAM17 were increased while the expression level of BACE1 decreased. This indicates that TCM can promote the expression level of ADAM10 and ADAM17, inhibit the expression level of BACE1, thus further inhibiting the production of amyloid protein and reducing the production of Aß and SP. Compared with RNAi group, the expression level of ADAM10 and ADAM17 in Aß + RNAi group was decreased while the expression level of BACE1 increased. Compared with the Aß + RNAi group the expression level of ADAM10 and ADAM17 in the Aß + RNAi + TCM group was increased while the expression level of BACE1 was decreased. The present study indicated the effects of the active components of Epimedium, Astragalus and Radix Puerariae may alleviate AD by up-regulating the expression of HAMP, thus reducing brain iron overload, promoting the expression of ADAM10 and ADAM17, inhibiting the expression of BACE1, and reducing the deposition of Aß.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Precursor de Proteína beta-Amiloide/metabolismo , Medicamentos Herbarios Chinos/farmacología , Hepcidinas/metabolismo , Fármacos Neuroprotectores/farmacología , Proteolisis/efectos de los fármacos , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Fragmentos de Péptidos/farmacología , Regulación hacia Arriba/efectos de los fármacos
9.
Behav Pharmacol ; 32(4): 345-350, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33290344

RESUMEN

Benzodiazepines bind to and act on α1-3 and α5-containing GABAA receptors. Previous studies suggest that different GABAA receptor α-subtypes mediate the various behavioral effects of benzodiazepines, which raises the possibility of combining benzodiazepines with subtype-selective GABAA receptor antagonists to improve the therapeutic profiles of benzodiazepines. This study examined the GABAA receptor subtype mediation of the tolerance to midazolam-induced antinociception in rats. Midazolam (3.2 mg/kg) significantly reduced the locomotion in rats which was prevented by the selective α1-preferring GABAA receptor antagonist ß-carboline-3-carboxylate-t-butyl ester (ßCCt) (3.2 mg/kg). Midazolam increased the paw withdrawal threshold as tested by the von Frey filament assay in the complete Freund's adjuvant-induced inflammatory pain model in rats, and this effect was not altered by ßCCt or another α1-preferring GABAA receptor antagonist 3-propoxy-ß-carboline hydrochloride (3PBC). Repeated treatment with midazolam in combination with vehicle, ßCCt or 3PBC (twice daily) for 7 days led to a progressive increase of the ED50 values in the midazolam- and vehicle-treated rats, but not in other rats, suggesting the development of tolerance to midazolam but not to the combination of midazolam with α1-preferring GABAA receptor antagonists. These results suggest the essential role of the α1-subtype of GABAA receptors in mediating the development of tolerance to midazolam-induced antinociceptive effects and raise the possibility of increasing therapeutic profiles of benzodiazepines by selectively blocking specific α-subtypes of GABAA receptors.


Asunto(s)
Analgésicos/farmacología , Benzodiazepinas/farmacología , Carbolinas/farmacología , Tolerancia a Medicamentos/fisiología , Midazolam/farmacología , Animales , Quimioterapia Combinada/métodos , Moduladores del GABA , Antagonistas de Receptores de GABA-A/farmacología , Umbral del Dolor/efectos de los fármacos , Vehículos Farmacéuticos/farmacología , Ratas , Receptores de GABA-A/metabolismo
10.
J Neurosci ; 39(50): 10071-10080, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31704786

RESUMEN

The claustrum connects with a broad range of cortical areas including the prefrontal cortex (PFC). However, the function of the claustrum (CLA) and its neural projections remains largely unknown. Here, we elucidated the role of the neural projections from the CLA to the PFC in regulating impulsivity in male rats. We first identified the CLA-PFC pathway by retrograde tracer and virus expression. By using immunofluorescent staining of the c-Fos-positive neurons, we showed that chemogenetic activation and inhibition of the CLA-PFC pathway reduced and increased overall activity of the PFC, respectively. In the 5-choice serial reaction time task (5-CSRTT), we found that chemogenetic activation and inhibition of the CLA-PFC pathway increased and reduced the impulsive-like behavior (i.e., premature responses), respectively. Furthermore, chemogenetic inhibition of the CLA-PFC pathway prevented methamphetamine-induced impulsivity, without affecting methamphetamine-induced hyperactivity. In contrast to the role of CLA-PFC pathway in selectively regulating impulsivity, activation of the claustrum disrupted attention in the 5-CSRTT. These results indicate that the CLA-PFC pathway is essential for impulsivity. This study may shed light on the understanding of impulsivity-related disorders such as drug addiction.SIGNIFICANCE STATEMENT The claustrum is one of the most mysterious brain regions. Although extensive anatomical studies demonstrated that the claustrum connects with many cortical areas, the function of the neural projections between the claustrum and cortical areas remain largely unknown. Here, we showed that the neural projections from the claustrum to the prefrontal cortex regulates impulsivity by using the designer drugs (DREADDs)-based chemogenetic tools. Interestingly, the claustrum-prefrontal cortex pathway also regulates methamphetamine-induced impulsivity, suggesting a critical role of this neural pathway in regulating impulsivity-related disorders such as drug addiction. Our results provided preclinical evidence that the claustrum-prefrontal cortex regulates impulsivity. The claustrum-prefrontal cortex pathway may be a novel target for the treatment of impulsivity-related brain disorders.


Asunto(s)
Conducta de Elección/fisiología , Claustro/fisiología , Conducta Impulsiva/fisiología , Corteza Prefrontal/fisiología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Estimulantes del Sistema Nervioso Central/farmacología , Conducta de Elección/efectos de los fármacos , Claustro/efectos de los fármacos , Conducta Impulsiva/efectos de los fármacos , Masculino , Metanfetamina/farmacología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología
11.
Cell Mol Neurobiol ; 40(2): 229-238, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31974906

RESUMEN

Trace amine-associated receptor 1 is one of the best-characterized receptors of trace amines. Growing evidence shows that TAAR1 negatively regulates the monoaminergic activity, including dopamine transmission in the mesocorticolimbic system. Neurochemical assays demonstrated that selective TAAR1 full and partial agonists were effective to prevent psychostimulants-induced dopamine transmission in vitro and in vivo. In the last decade, many preclinical models of psychostimulant addiction such as drug-induced behavioral sensitization, drug-induced conditioned place preference, drug self-administration, drug discrimination, and relapse models were used to assess the effects of TAAR1 agonists on psychostimulants' behavioral effects. In general, activation of TAAR1 attenuated while knockout of TAAR1 potentiated psychostimulant abuse-related behaviors. Here, we review the advances in TAAR1 and its agonists in modulating psychostimulant addiction. We discuss the similarities and differences between the neurochemical and behavioral effects of TAAR1 full and partial agonists. We also discuss several concerns including the abuse liability, sleep reduction, and species-dependent effects that might affect the successful translation of TAAR1 agonists from preclinical studies to clinical application. In conclusion, although further investigations are in need to address certain concerns and the underlying neural mechanisms, TAAR1 agonists appear to be a promising pharmacotherapy to treat psychostimulant addiction and prevent relapse.


Asunto(s)
Estimulantes del Sistema Nervioso Central/metabolismo , Psicotrópicos/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Animales , Conducta Adictiva/metabolismo , Conducta Adictiva/prevención & control , Estimulantes del Sistema Nervioso Central/farmacología , Dopamina/metabolismo , Humanos , Psicotrópicos/farmacología
12.
Eur J Neurosci ; 50(3): 2503-2512, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30113098

RESUMEN

Drug addiction is an aberrant memory that shares the same memory processes as other memories. Brief exposure to drug-associated cues could result in reconsolidation, a hypothetical process during which original memory could be updated. In contrast, longer exposure times to drug-associated cues could trigger extinction, a process that decreases the conditioned responding. In this review, we discuss the pharmacological and non-pharmacological manipulations on the reconsolidation and extinction that could be used to interfere with drug reward memories. Pharmacological agents such as ß-adrenergic receptor antagonist propranolol can interfere with reconsolidation to disrupt drug reward memory. Pharmacological agents such as the NMDA receptor glycine site agonists d-cycloserine and d-serine can facilitate extinction and then attenuate the expression of drug reward memory. Besides pharmacological interventions, drug-free behavioral approaches by utilizing the reconsolidation and extinction, such as 'post-retrieval extinction' and 'UCS-retrieval extinction', are also effective to erase or inhibit the recall of drug reward memory. Taken together, pharmacological modulation and non-pharmacological modulation of reconsolidation and extinction are promising approaches to regulate drug reward memory and prevent relapse.


Asunto(s)
Extinción Psicológica/fisiología , Memoria/fisiología , Recompensa , Trastornos Relacionados con Sustancias/psicología , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/uso terapéutico , Animales , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Extinción Psicológica/efectos de los fármacos , Humanos , Memoria/efectos de los fármacos , Recuerdo Mental/efectos de los fármacos , Recuerdo Mental/fisiología , Trastornos Relacionados con Sustancias/tratamiento farmacológico
13.
Behav Pharmacol ; 30(5): 429-434, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30383551

RESUMEN

Pharmacotherapies for fibromyalgia treatment are lacking. This study examined the antinociceptive and antidepressant-like effects of imidazoline I2 receptor (I2R) agonists in a reserpine-induced model of fibromyalgia in rats. Rats were treated for 3 days with vehicle or reserpine. The von Frey filament test was used to assess the antinociceptive effects of I2 receptor agonists, and the forced swim test was used to assess the antidepressant-like effects of these drugs. 2-BFI (3.2-10 mg/kg, intraperitoneally), phenyzoline (17.8-56 mg/kg, intraperitoneally), and CR4056 (3.2-10 mg/kg, intraperitoneally) all dose-dependently produced significant antinociceptive effects, which were attenuated by the I2R antagonist idazoxan. Only CR4056 significantly reduced the immobility time in the forced swim test in both vehicle-treated and reserpine-treated rats. These data suggest that I2R agonists may be useful to treat fibromyalgia-related pain and comorbid depression.


Asunto(s)
Hiperalgesia/tratamiento farmacológico , Idazoxan/farmacología , Receptores de Imidazolina/metabolismo , Analgésicos/farmacología , Animales , Benzofuranos/farmacología , Depresión/tratamiento farmacológico , Modelos Animales de Enfermedad , Fibromialgia/tratamiento farmacológico , Hiperalgesia/inducido químicamente , Idazoxan/metabolismo , Imidazoles/farmacología , Receptores de Imidazolina/agonistas , Imidazolinas/metabolismo , Imidazolinas/farmacología , Masculino , Dolor/tratamiento farmacológico , Dimensión del Dolor/efectos de los fármacos , Quinazolinas/farmacología , Ratas , Ratas Sprague-Dawley , Reserpina/farmacología
14.
Behav Pharmacol ; 30(5): 422-428, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30480551

RESUMEN

Previous preclinical research suggests that L-methamphetamine (L -MA) has potential therapeutic utility to treat psychostimulant abuse. This study examined potential abuse-related and adverse physiological effects of D -MA and L -MA alone and in combination in rats, as these effects had not been previously characterized. Potential abuse-related effects were examined in locomotor sensitization and conditioned place preference paradigms. Body temperature was monitored to assess the physiological effects of these drugs or drug combinations. In the locomotor study, D-MA induced locomotor sensitization to both D-MA and L -MA. L -MA induced locomotor sensitization only to D-MA. Responses to a combination of L-MA and D -MA were not differentially affected by L-MA or D-MA conditioning. In the conditioned place preference study, D-MA and L -MA each induced significant place preference. L -MA did not attenuate D-MA-induced place preference. In the body temperature study, D-MA induced hyperthermia and L -MA induced hypothermia. In combination, L -MA did not affect D-MA-induced hyperthermia. These data suggest that L -MA alone produces less abuse-related and adverse physiological effects than D-MA, but modulates and is modulated by concurrent and subsequent D-MA exposure, which may enhance the abuse liability of both drugs. These findings should be considered when L -MA is proposed for replacement therapy.


Asunto(s)
Metanfetamina/análogos & derivados , Metanfetamina/farmacología , Estereoisomerismo , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Clásico/efectos de los fármacos , Fiebre , Hipotermia , Locomoción/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
15.
J Neurosci ; 37(4): 882-892, 2017 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-28123023

RESUMEN

A novel G-protein coupled receptor, trace amine-associated receptor 1 (TAAR1), has been shown to be a promising target to prevent stimulant relapse. Our recent studies showed that systemic administration of TAAR1 agonists decreased abuse-related behaviors of cocaine. However, the role of TAAR1 in specific subregions of the reward system in drug addiction is unknown. Here, using a local pharmacological activation method, we assessed the role of TAAR1 within the subregions of the mesocorticolimbic system: that is, the VTA, the prelimbic cortex (PrL), and infralimbic cortex of medial prefrontal cortex, the core and shell of NAc, BLA, and CeA, on cue- and drug-induced cocaine-seeking in the rat cocaine reinstatement model. We first showed that TAAR1 mRNA was expressed throughout these brain regions. Rats underwent cocaine self-administration, followed by extinction training. RO5166017 (1.5 or 5.0 µg/side) or vehicle was microinjected into each brain region immediately before cue- and drug-induced reinstatement of cocaine-seeking. The results showed that microinjection of RO5166017 into the VTA and PrL decreased both cue- and drug priming-induced cocaine-seeking. Microinjection of RO5166017 into the NAc core and shell inhibited cue- and drug-induced cocaine-seeking, respectively. Locomotor activity or food reinforced operant responding was unaffected by microinjection of RO5166017 into these brain regions. Cocaine-seeking behaviors were not affected by RO5166017 when microinjected into the substantia nigra, infralimbic cortex, BLA, and CeA. Together, these results indicate that TAAR1 in different subregions of the mesocorticolimbic system distinctly contributes to cue- and drug-induced reinstatement of cocaine-seeking behavior. SIGNIFICANCE STATEMENT: TAAR1 has been indicated as a modulator of the dopaminergic system. Previous research showed that systemic administration of TAAR1 agonists could attenuate cocaine-related behaviors, suggesting that TAAR1 may be a promising drug target for the treatment of cocaine addiction. However, the specific role of TAAR1 in subregions of the mesocorticolimbic system in drug addiction is unknown. Here, we first showed that TAAR1 mRNA is expressed throughout the subregions of the mesocorticolimbic system. Then, by using a local pharmacological activation method, we demonstrated that TAAR1 in different subregions of the mesocorticolimbic system distinctly contributes to cue- and drug-induced reinstatement of cocaine-seeking behavior.


Asunto(s)
Amígdala del Cerebelo/fisiología , Cocaína/administración & dosificación , Neuronas Dopaminérgicas/fisiología , Comportamiento de Búsqueda de Drogas/fisiología , Corteza Prefrontal/fisiología , Receptores Acoplados a Proteínas G/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Dopamina/fisiología , Neuronas Dopaminérgicas/efectos de los fármacos , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Sistema Límbico/efectos de los fármacos , Sistema Límbico/fisiología , Masculino , Microinyecciones , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiología , Oxazoles/administración & dosificación , Fenetilaminas/administración & dosificación , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Autoadministración , Área Tegmental Ventral/fisiología
16.
Eur J Neurosci ; 47(9): 1087-1095, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29514408

RESUMEN

Chronic pain is a significant public health problem with a lack of safe and effective analgesics. The imidazoline I2 receptor (I2 R) is a promising analgesic target, but the neuroanatomical structures involved in mediating I2 R-associated behaviors are unknown. I2 Rs are enriched in the arcuate nucleus, dorsal raphe (DR), interpeduncular nucleus, lateral mammillary body, medial habenula, nucleus accumbens (NAc) and paraventricular nucleus; thus, this study investigated the antinociceptive and hypothermic effects of microinjections of the I2 R agonist 2-(2-benzofuranyl)-2-imidazoline hydrochloride (2-BFI). In rats, intra-DR microinjections produced antinociception in complete Freund's adjuvant- and chronic constriction injury-induced pain models. Intra-NAc microinjections produced antinociception and increased noxious stimulus-associated side time in a place escape/avoidance paradigm. Intra-NAc pretreatment with the I2 R antagonist idazoxan but not the D1 receptor antagonist SCH23390 or the D2 receptor antagonist raclopride attenuated intra-NAc 2-BFI-induced antinociception. Intra-NAc idazoxan did not attenuate systemically administered 2-BFI-induced antinociception. Microinjections into the other regions did not produce antinociception, and in none of the regions produced hypothermia. These data suggest that I2 R activation in some but not all I2 R-enriched brain regions is sufficient to produce antinociception and supports the theory that different I2 R-associated effects are mediated via distinct receptor populations, which may in turn be distributed differentially throughout the CNS.


Asunto(s)
Encéfalo/efectos de los fármacos , Imidazoles/farmacología , Núcleo Accumbens/efectos de los fármacos , Receptores de Dopamina D1/efectos de los fármacos , Analgésicos/farmacología , Animales , Benzofuranos/farmacología , Femenino , Imidazolinas/farmacología , Masculino , Dimensión del Dolor/métodos , Ratas Sprague-Dawley
17.
Acta Pharmacol Sin ; 39(12): 1823-1829, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30382181

RESUMEN

Drug addiction is a chronic, relapsing brain disorder. Multiple neural networks in the brain including the reward system (e.g., the mesocorticolimbic system), the anti-reward/stress system (e.g., the extended amygdala), and the central immune system, are involved in the development of drug addiction and relapse after withdrawal from drugs of abuse. Preclinical and clinical studies have demonstrated that it is promising to control drug addiction by pharmacologically targeting the addiction-related systems in the brain. Here we review the pharmacological targets within the dopamine system, glutamate system, trace amine system, anti-reward system, and central immune system, which are of clinical interests. Furthermore, we discuss other potential therapies, e.g., brain stimulation, behavioral treatments, and therapeutic gene modulation, which could be effective to treat drug addiction. We conclude that, although drug addiction is a complex disorder that involves complicated neural mechanisms and psychological processes, this mental disorder is treatable and may be curable by therapies such as gene modulation in the future.


Asunto(s)
Trastornos Relacionados con Sustancias/tratamiento farmacológico , Trastornos Relacionados con Sustancias/terapia , Animales , Terapia Conductista/métodos , Encéfalo/efectos de los fármacos , Antagonistas de Dopamina/uso terapéutico , Terapia por Estimulación Eléctrica/métodos , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Terapia Genética/métodos , Humanos , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Trastornos Relacionados con Sustancias/genética
18.
Med Res Rev ; 37(3): 441-474, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27879006

RESUMEN

The cannabinoid CB1 receptor is a G protein coupled receptor and plays an important role in many biological processes and physiological functions. A variety of CB1 receptor agonists and antagonists, including endocannabinoids, phytocannabinoids, and synthetic cannabinoids, have been discovered or developed over the past 20 years. In 2005, it was discovered that the CB1 receptor contains allosteric site(s) that can be recognized by small molecules or allosteric modulators. A number of CB1 receptor allosteric modulators, both positive and negative, have since been reported and importantly, they display pharmacological characteristics that are distinct from those of orthosteric agonists and antagonists. Given the psychoactive effects commonly associated with CB1 receptor agonists and antagonists/inverse agonists, allosteric modulation may offer an alternate approach to attain potential therapeutic benefits while avoiding inherent side effects of orthosteric ligands. This review details the complex pharmacological profiles of these allosteric modulators, their structure-activity relationships, and efforts in elucidating binding modes and mechanisms of actions of reported CB1 allosteric modulators. The ultimate development of CB1 receptor allosteric ligands could potentially lead to improved therapies for CB1-mediated neurological disorders.


Asunto(s)
Receptor Cannabinoide CB1/metabolismo , Regulación Alostérica , Animales , Humanos , Relación Estructura-Actividad
19.
Behav Pharmacol ; 28(7): 590-593, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28704278

RESUMEN

Increasing evidence suggests that trace amine-associated receptor 1 (TAAR1) is an important modulator of the dopaminergic system. Existing molecular evidence indicates that TAAR1 regulates dopamine levels through interactions with dopamine transporters and D2 receptors. However, investigations to date have not been exhaustive and other pathways may be involved. In this study, we used a well-described set of behaviors, quinpirole-induced yawning and hypothermia, to explore the potential interaction of TAAR1 and D3 receptors, which are members of the 'D2-like' dopamine receptor subfamily. Previous studies have shown that for D2/D3 receptor agonists, the induction of yawning is a D3 receptor-mediated effect, whereas the inhibition of yawning and induction of hypothermia are D2 receptor-mediated effects. Quinpirole produced an inverted U-shaped dose-effect curve for yawning, which was shifted downward dose-dependently by each of the TAAR1 agonists RO5263397 and RO5166017. Quinpirole also produced dose-dependent hypothermia, which was not affected by either TAAR1 agonist. These results suggest that TAAR1 agonists may interact with D3 receptors and/or its downstream pathways, as opposed to D2 receptors. These findings may shed light on a previously unexplored possibility for the mechanism of TAAR1-mediated effects.


Asunto(s)
Oxazoles/farmacología , Fenetilaminas/farmacología , Bostezo/efectos de los fármacos , Animales , Dopamina/metabolismo , Agonistas de Dopamina/farmacología , Hipotermia/inducido químicamente , Masculino , Quinpirol/metabolismo , Quinpirol/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/agonistas , Receptores de Dopamina D3/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo
20.
J Neurosci ; 35(8): 3676-88, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716865

RESUMEN

Therapeutic repair of myelin disorders may be limited by the relatively slow rate of human oligodendrocyte differentiation. To identify appropriate pharmacological targets with which to accelerate differentiation of human oligodendrocyte progenitors (hOPCs) directly, we used CD140a/O4-based FACS of human forebrain and microarray to hOPC-specific receptors. Among these, we identified CHRM3, a M3R muscarinic acetylcholine receptor, as being restricted to oligodendrocyte-biased CD140a(+)O4(+) cells. Muscarinic agonist treatment of hOPCs resulted in a specific and dose-dependent blockade of oligodendrocyte commitment. Conversely, when hOPCs were cocultured with human neurons, M3R antagonist treatment stimulated oligodendrocytic differentiation. Systemic treatment with solifenacin, an FDA-approved muscarinic receptor antagonist, increased oligodendrocyte differentiation of transplanted hOPCs in hypomyelinated shiverer/rag2 brain. Importantly, solifenacin treatment of engrafted animals reduced auditory brainstem response interpeak latency, indicative of increased conduction velocity and thereby enhanced functional repair. Therefore, solifenacin and other selective muscarinic antagonists represent new adjunct approaches to accelerate repair by engrafted human progenitors.


Asunto(s)
Células Madre Fetales/citología , Antagonistas Muscarínicos/farmacología , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Quinuclidinas/farmacología , Regeneración , Tetrahidroisoquinolinas/farmacología , Animales , Tronco Encefálico/citología , Tronco Encefálico/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Células Madre Fetales/efectos de los fármacos , Células Madre Fetales/metabolismo , Células Madre Fetales/trasplante , Humanos , Masculino , Ratones , Agonistas Muscarínicos/farmacología , Vaina de Mielina/genética , Neurogénesis , Antígenos O/genética , Antígenos O/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/trasplante , Prosencéfalo/citología , Prosencéfalo/embriología , Receptor Muscarínico M3 , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Succinato de Solifenacina
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda