Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Pharmacol Exp Ther ; 384(3): 372-381, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36507847

RESUMEN

Although there are no Food and Drug Administration-approved treatments for cocaine use disorder, several modafinil analogs have demonstrated promise in reducing cocaine self-administration and reinstatement in rats. Furthermore, the range of dopamine transporter (DAT) compounds provides an opportunity to develop pharmacotherapeutics without abuse liability. This study extended the comparison of JJC8-088 and JJC8-091, the former compound having higher DAT affinity and predicted abuse liability, to rhesus monkeys using a concurrent cocaine versus food schedule of reinforcement. First, binding to striatal DAT was examined in cocaine-naïve monkey tissue. Next, intravenous pharmacokinetics of both JJC compounds were evaluated in cocaine-experienced male monkeys (n = 3/drug). In behavioral studies, acute and chronic administration of both compounds were evaluated in these same monkeys responding under a concurrent food versus cocaine (0 and 0.003-0.1 mg/kg per injection) schedule of reinforcement. In nonhuman primate striatum, JJC8-088 had higher DAT affinity compared with JJC8-091 (14.4 ± 9 versus 2730 ± 1270 nM, respectively). Both JJC compounds had favorable plasma pharmacokinetics for behavioral assessments, with half-lives of 1.1 hours and 3.5 hours for JJC8-088 (0.7 mg/kg, i.v.) and JJC8-091 (1.9 mg/kg, i.v.), respectively. Acute treatment with both compounds shifted the cocaine dose-response curve to the left. Chronic treatment with JJC8-088 decreased cocaine choice in two of the three monkeys, whereas JJC8-091 only modestly reduced cocaine allocation in one monkey. Differences in affinities of JJC8-091 DAT binding in monkeys compared with rats may account for the poor rodent-to-monkey translation. Future studies should evaluate atypical DAT blockers in combination with behavioral interventions that may further decrease cocaine choice. SIGNIFICANCE STATEMENT: Cocaine use disorder (CUD) remains a significant public health problem with no Food and Drug Administration-approved treatments. The ability of drugs that act in the brain in a similar manner to cocaine, but with lower abuse liability, has clinical implications for a treatment of CUD.


Asunto(s)
Cocaína , Masculino , Ratas , Animales , Cocaína/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Macaca mulatta/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Autoadministración , Relación Dosis-Respuesta a Droga
2.
Synapse ; 75(4): e22190, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33025628

RESUMEN

Metabotropic glutamate (mGlu) receptors are regulators of glutamate release and targets for development of therapies for hyperactive glutamatergic signaling. However, the effects of long-term stimulation of mGlu receptors on cellular signaling in the brain have not been described. This study investigated the effects of 2-day and 14-day osmotic mini-pump administration of the mGlu2,3 agonist LY379268 (3.0 mg kg-1  day-1 ) to rats on receptor-mediated G-protein activation and signaling in mesocorticolimbic regions in rat brain sections. A significant reduction in LY379268-stimulated [35 S]GTPγS binding was observed in the 14-day group in some cortical regions, prefrontal cortex, nucleus accumbens, and ventral pallidum. The 14-day LY379268 treatment group exhibited mGlu2 mRNA levels significantly lower in hippocampus, nucleus accumbens, caudate, and ventral pallidum. In both 2-day and 14-day treatment groups immunodetectable phosphorylated cAMP Response Element-Binding protein (CREB) was significantly reduced across all brain regions. In the 2-day group, we observed significantly lower immunodetectable CREB protein across all brain regions, which was subsequently increased in the 14-day group but failed to achieve control values. Neither immunodetectable extracellular signal-regulated kinase (ERK) protein nor phosphorylated ERK from 2-day or 14-day treatment groups differed significantly from control across all brain regions. However, the ratio of phosphorylated ERK to total ERK protein was significantly greater in the 14-day treatment group compared with the control. These results identify compensatory changes to mGlu2,3 signal transduction in rat brains after chronic systemic administration of agonist, which could be predictive of the mechanism of action in human pharmacotherapies.


Asunto(s)
Ácido Glutámico , Receptores de Glutamato Metabotrópico , Animales , Encéfalo/metabolismo , Proteínas de Unión al GTP/metabolismo , Ratas , Receptores de Glutamato Metabotrópico/agonistas , Transducción de Señal
3.
Handb Exp Pharmacol ; 258: 89-125, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31598835

RESUMEN

Opioid analgesics, most of which act through mu opioid receptors, have long represented valuable therapeutic agents to treat severe pain. Concerted drug development efforts for over a 100 years have resulted in a large variety of opioid analgesics used in the clinic, but all of them continue to exhibit the side effects, especially respiratory depression, that have long plagued the use of morphine. The recent explosion in fatalities resulting from overdose of prescription and synthetic opioids has dramatically increased the need for safer analgesics, but recent developments in mu receptor research have provided new strategies to develop such drugs. This chapter reviews recent advances in developing novel opioid analgesics from an understanding of mu receptor structure and function. This includes a summary of the mechanism of agonist binding deduced from the crystal structure of mu receptors. It will also highlight the development of novel agonist mechanisms, including biased agonists, bivalent ligands, and allosteric modulators of mu receptor function, and describe how receptor phosphorylation modulates these pathways. Finally, it will summarize research on the alternative pre-mRNA splicing mechanisms that produces a multiplicity of mu receptor isoforms. Many of these isoforms exhibit different pharmacological specificities and brain circuitry localization, thus providing an opportunity to develop novel drugs with increased therapeutic windows.


Asunto(s)
Analgésicos Opioides/farmacología , Dolor/tratamiento farmacológico , Receptores Opioides mu , Humanos , Ligandos , Trastornos Relacionados con Opioides
4.
Synapse ; 68(10): 437-44, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24916769

RESUMEN

Concurrent use of cocaine and heroin (speedball) has been shown to exert synergistic effects on dopamine neurotransmission in the nucleus accumbens (NAc), as observed by significant increases in extracellular dopamine levels and compensatory elevations in the maximal reuptake rate of dopamine. The present studies were undertaken to determine whether chronic self-administration of cocaine, heroin or a combination of cocaine:heroin led to compensatory changes in the abundance and/or affinity of high- and low-affinity DAT binding sites. Saturation binding of the cocaine analog [(125) I] 3ß-(4-iodophenyl)tropan-2ß-carboxylic acid methyl ester ([(125) I]RTI-55) in rat NAc membranes resulted in binding curves that were best fit to two-site binding models, allowing calculation of dissociation constant (Kd ) and binding density (Bmax ) values corresponding to high- and low-affinity DAT binding sites. Scatchard analysis of the saturation binding curves clearly demonstrate the presence of high- and low- affinity binding sites in the NAc, with low-affinity sites comprising 85 to 94% of the binding sites. DAT binding analyses revealed that self-administration of cocaine and a cocaine:heroin combination increased the affinity of the low-affinity site for the cocaine congener RTI-55 compared to saline. These results indicate that the alterations observed following chronic speedball self-administration are likely due to the cocaine component alone; thus further studies are necessary to elaborate upon the synergistic effect of cocaine:heroin combinations on the dopamine system in the NAc.


Asunto(s)
Cocaína/administración & dosificación , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Inhibidores de Captación de Dopamina/administración & dosificación , Heroína/administración & dosificación , Narcóticos/administración & dosificación , Núcleo Accumbens/efectos de los fármacos , Administración Intravenosa , Animales , Membrana Celular/metabolismo , Cocaína/análogos & derivados , Cocaína/farmacocinética , Combinación de Medicamentos , Drogas Ilícitas , Radioisótopos de Yodo/farmacocinética , Masculino , Núcleo Accumbens/metabolismo , Radiofármacos , Ratas Endogámicas F344 , Autoadministración
5.
J Neurochem ; 124(6): 808-20, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23286559

RESUMEN

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


Asunto(s)
Proteínas Portadoras/biosíntesis , Cuerpo Estriado/metabolismo , Receptor Cannabinoide CB1/fisiología , Receptores de Dopamina D2/fisiología , Receptores Opioides delta/biosíntesis , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Antagonistas de los Receptores de Dopamina D2 , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptores Opioides delta/genética
6.
Anesthesiology ; 114(3): 633-42, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21293255

RESUMEN

BACKGROUND: Neuropathic pain alters opioid self-administration in rats. The brain regions altered in the presence of neuropathic pain mediating these differences have not been identified, but likely involve ascending pain pathways interacting with the limbic system. The amygdala is a brain region that integrates noxious stimulation with limbic activity. METHODS: µ-Opioid receptors were blocked in the amygdala using the irreversible antagonist, ß-funaltrexamine, and the antiallodynic and reinforcing effects of heroin were determined in spinal nerve-ligated rats. In addition, the effect of ß-funaltrexamine was determined on heroin self-administration in sham-operated rats. RESULTS: ß-Funaltrexamine decreased functional activity of µ-opioid receptors by 60 ± 5% (mean ± SD). Irreversible inhibition of µ-opioid receptors in the amygdala significantly attenuated the ability of doses of heroin up to 100 µg/kg to reverse hypersensitivity after spinal nerve ligation. Heroin intake by self-administration in spinal nerve-ligated rats was increased from 5.0 ± 0.3 to 9.9 ± 2.1 infusions/h after administration of 2.5 nmol of ß-funaltrexamine in the lateral amygdala, while having no effect in sham-operated animals (5.8 ± 1.6 before, 6.7 ± 0.9 after). The antiallodynic effects of 60 µg/kg heroin were decreased up to 4 days, but self-administration was affected for up to 14 days. CONCLUSIONS: µ-Opioid receptors in the lateral amygdala partially meditate heroin's antiallodynic effects and self-administration after peripheral nerve injury. The lack of effect of ß-funaltrexamine on heroin self-administration in sham-operated subjects suggests that opioids maintain self-administration through a distinct mechanism in the presence of pain.


Asunto(s)
Amígdala del Cerebelo/fisiología , Analgésicos Opioides/farmacología , Condicionamiento Operante/efectos de los fármacos , Heroína/farmacología , Hiperalgesia/tratamiento farmacológico , Traumatismos de los Nervios Periféricos , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hiperalgesia/psicología , Infusiones Intravenosas , Masculino , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Ratas , Ratas Endogámicas F344 , Refuerzo en Psicología , Autoadministración , Nervios Espinales/lesiones
7.
Brain Res ; 1761: 147387, 2021 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-33631209

RESUMEN

Phenmetrazine (PHEN) is a putative treatment for cocaine and psychostimulant recidivism; however, neurochemical changes underlying its activity have not been fully elucidated. We sought to characterize brain homeostatic adaptations to chronic PHEN, specifically on functional brain activity (local cerebral glucose utilization), G-Protein Coupled Receptor-stimulated G-protein activation, and phosphorylation of ERK1/2Thr202/Tyr204, GSK3ßTyr216, and DARPP-32Thr34. Male Sprague-Dawley rats were implanted with sub-cutaneous minipumps delivering either saline (vehicle), acute (2-day) or chronic (14-day) low dose (25 mg/kg/day) or high dose (50 mg/kg/day) PHEN. Acute administration of high dose PHEN increased local cerebral glucose utilization measured by 2-[14C]-deoxyglucose uptake in basal ganglia and motor-related regions of the rat brain. However, chronically treated animals developed tolerance to these effects. To identify the neurochemical changes associated with PHEN's activity, we performed [35S]GTPγS binding assays on unfixed and immunohistochemistry on fixed coronal brain sections. Chronic PHEN treatment dose-dependently attenuated D2 dopamine and α2-adrenergic, but not 5-HT1A, receptor-mediated G-protein activation. Two distinct patterns of effects on pERK1/2 and pDARPP-32 were observed: 1) chronic low dose PHEN decreased pERK1/2, and also significantly increased pDARPP-32 levels in some regions; 2) acute and chronic PHEN increased pERK1/2, but chronic high dose PHEN treatment tended to decrease pDARPP-32. Chronic low dose, but not high dose, PHEN significantly reduced pGSK3ß levels in several regions. Our study provides definitive evidence that extended length PHEN dosage schedules elicit distinct modes of neuronal acclimatization in cellular signaling. These pharmacodynamic modifications should be considered in drug development for chronic use.

8.
Bioorg Med Chem Lett ; 19(1): 58-61, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19038547

RESUMEN

A series of enantiomerically pure 1-naphthyl and 4-indolyl arylalkylamines were prepared and evaluated for their binding affinities to the monoamine transporters. The two series of enantiomers displayed considerable differences in binding selectivity between the monoamine transporters, leading to the design of (S)-4-(3,4-dichlorophenyl)-4-(1H-indol-4-yl)-N-methylbutan-1-amine as a potent inhibitor for the dopamine and serotonin transporters.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Inhibidores de la Captación de Neurotransmisores/síntesis química , Inhibidores de la Captación de Neurotransmisores/farmacología , Aminas , Animales , Antidepresivos de Segunda Generación , Antagonistas de Dopamina/síntesis química , Inhibidores de Captación de Dopamina/síntesis química , Diseño de Fármacos , Humanos , Unión Proteica , Antagonistas de la Serotonina/síntesis química , Inhibidores Selectivos de la Recaptación de Serotonina/síntesis química , Estereoisomerismo
9.
J Pharmacol Exp Ther ; 326(2): 587-95, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18492949

RESUMEN

Previous studies have shown that the phenylisothiocyanate tropane analog 2-beta-propanoyl-3-beta-(2-naphthyl)-8-[4-isothiocyanato)benzyl]nortropane (HD-205) binds covalently to dopamine and serotonin transporters (DAT and SERT, respectively) in rat brain membranes (Biochem Pharmacol 74:336-344, 2007). The present study evaluated the irreversible effects of HD-205 in vivo in rats after intracranial injection. Rats were implanted with unilateral cannulae in rat striatum, and HD-205 (0.001-3 nmol) was administered by intrastriatal injection. In vitro autoradiography of DAT binding with [125I]2-carbomethoxy-3-(4-iodophenyl)tropane (RTI-55) on brain sections obtained 24 h after injection showed a highly localized blockade of binding in striatum, with maximal blockade of binding by 1 to 3 nmol HD-205. Similar blockade of SERT binding (using [3H]-citalopram) was observed in the same area. No blockade of DAT or SERT binding was observed after intrastriatal injections of the reversible analog 2-beta-propanoyl-3-beta-(2-naphthyl)-8-benzyl nortropane (HD-206), and HD-205 treatment had no effect on D(2)- and mu-opioid-stimulated guanosine 5'-O-(3-[35S]thio)-triphosphate binding in sections from the same animals. In a time course study, rats administered with 1 nmol HD-205 showed recovery of 50% DAT binding after 3 to 4 days postinjection, and full recovery after 6 weeks. Rats implanted with bilateral cannulae were tested for cocaine-induced locomotor activity. Two days after intrastriatal injection of 1 nmol of HD-205, systemic (20 mg/kg i.p.) cocaine-induced locomotor activity was not affected; however, locomotor activity induced by intrastriatal administration of cocaine (6 nmol) was eliminated. This strategy of site-specific chemical blockade of transporters could serve as a valuable tool to evaluate the neuroanatomical basis of DAT-mediated cocaine effects.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Isotiocianatos/farmacología , Nortropanos/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Tropanos/farmacología , Animales , Cocaína/farmacología , Cuerpo Estriado/metabolismo , Relación Dosis-Respuesta a Droga , Isotiocianatos/química , Masculino , Estructura Molecular , Actividad Motora/efectos de los fármacos , Nortropanos/química , Unión Proteica , Ratas , Ratas Endogámicas F344 , Tropanos/química
10.
Biochem Pharmacol ; 74(2): 336-44, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17540345

RESUMEN

Irreversible tropane analogs have been useful in identifying binding sites of cocaine on biogenic amine transporters, including transporters for dopamine (DAT), serotonin (SERT) and norepinephrine (NET). The present study characterizes the properties of the novel phenylisothiocyanate tropane HD-205, synthesized from the highly potent 2-napthyl tropane analog WF-23. In radioligand binding studies in brain membranes, direct IC(50) values of HD-205 were 4.1, 14 and 280nM at DAT, SERT and NET, respectively. Wash-resistant binding was characterized by preincubation of HD-205 with brain membranes, followed by extensive washing before performing transporter radioligand binding. Results for HD-205 showed wash-resistant IC(50) values of 191, 230 and 840nM at DAT, SERT and NET, respectively. Saturation binding studies with [(125)I]RTI-55 in membranes pretreated with 100nM HD-205 showed that HD-205 significantly decreased the B(max) but not K(D) of DAT and SERT binding. To further characterize its irreversible binding, an iodinated analog of HD-205, HD-244, was prepared from a trimethylsilyl precursor. The direct IC(50) of HD-244 at DAT was 20nM. [(125)I]HD-244 was synthesized with chloramine-T, purified on HPLC, reacted with rat striatal membranes, and proteins were separated by SDS-PAGE. Results showed several non-specific labeled bands, but only a single specific band of radioactivity co-migrating with an immunoreactive DAT band at approx. 80 kilodaltons was detected, suggesting that [(125)I]HD-244 covalently labeled DAT protein in striatal membranes. These results demonstrate that phenylisothiocyanate analogs of WF-23 can be used as potential ligands to map distinct binding sites of cocaine analogs at DAT.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Isotiocianatos/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Tropanos/metabolismo , Animales , Cocaína/análogos & derivados , Cocaína/metabolismo , Masculino , Ratas
11.
Neuroreport ; 17(15): 1619-22, 2006 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-17001280

RESUMEN

Spinally administered adenosine reduces hypersensitivity in animals and humans with nerve injury, but also causes transient pain in humans and reduces tonic inhibition in spinal neurons. Nerve injury results in increased tonic spinal cord adenosine A1 receptor activation, consistent with a role for adenosine to generate hypersensitivity. Here, we demonstrate that chronic intrathecal adenosine induces hypersensitivity in normal animals and that chronic blockade of spinal adenosine A1 receptors by the A1 antagonist 8-cyclopentyl-1,3-dipropylxanthine partially prevents nerve injury-induced hypersensitivity. In contrast, chronic blockade of spinal adenosine A1 receptors failed to reduce increased tonic G-protein signaling in the spinal cord after nerve injury. These data support a role for chronic adenosine A1 receptor stimulation after nerve injury to result in hypersensitivity.


Asunto(s)
Receptor de Adenosina A1/metabolismo , Enfermedades de la Médula Espinal/fisiopatología , Médula Espinal/metabolismo , Adenosina/efectos adversos , Antagonistas del Receptor de Adenosina A1 , Animales , Esquema de Medicación , Lateralidad Funcional/efectos de los fármacos , Lateralidad Funcional/fisiología , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Estimulación Física/métodos , Unión Proteica/efectos de los fármacos , Ratas , Umbral Sensorial/efectos de los fármacos , Umbral Sensorial/fisiología , Médula Espinal/efectos de los fármacos , Enfermedades de la Médula Espinal/inducido químicamente , Enfermedades de la Médula Espinal/tratamiento farmacológico , Isótopos de Azufre/farmacocinética , Teofilina/análogos & derivados , Teofilina/uso terapéutico
12.
Neuropharmacology ; 48(1): 62-71, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15617728

RESUMEN

This study examines the effect of long-term elevation of brain monoamine levels on receptor/G-protein coupling by chronic administration of a highly potent tropane analog, WF-23 (2beta-propanoyl-3beta-(2-naphthyl) tropane). WF-23 blocks dopamine, serotonin and norepinephrine transporters with high affinity in vitro, and blocks transporters for at least two days following a single in vivo administration. Rats were chronically treated for 15 days with 1mg/kg WF-23, injected i.p. every two days. Receptor activation of G-proteins was determined by [35S]GTPgammaS autoradiography in brain sections for D2, 5-HT1A and alpha2-adrenergic receptors, as well as mu opioid receptors as a non-monoamine receptor control. Chronic treatment with WF-23 produced significant reductions in D2, 5-HT1A, and alpha2-adrenergic receptor-stimulated [35S]GTPgammaS binding in caudate/putamen, hippocampus and amygdala, respectively. There were no effects of WF-23 treatment on mu opioid-stimulated [35S]GTPgammaS binding. Additionally, there was no effect of WF-23 treatment on D2 receptor binding, as determined by [3H]spiperone autoradiography. These data show that chronic blockade of monoamine transporters produces specific uncoupling of receptors and G-proteins in specific brain regions in the absence of receptor downregulation.


Asunto(s)
Apomorfina/análogos & derivados , Monoaminas Biogénicas/metabolismo , Encéfalo/efectos de los fármacos , Cocaína/análogos & derivados , Cocaína/farmacología , Moduladores del Transporte de Membrana , Proteínas de Transporte de Membrana/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Animales , Apomorfina/farmacología , Monoaminas Biogénicas/agonistas , Monoaminas Biogénicas/antagonistas & inhibidores , Encéfalo/anatomía & histología , Cocaína/química , Antagonistas de Dopamina/farmacocinética , Esquema de Medicación , Interacciones Farmacológicas , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Isótopos/farmacocinética , Masculino , Unión Proteica/efectos de los fármacos , Ensayo de Unión Radioligante/métodos , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Espiperona/farmacocinética , Tiempo
13.
Brain Res ; 1038(1): 76-82, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15748875

RESUMEN

Intrathecal and epidural administration of the alpha2-adrenergic receptor agonist clonidine in humans results in analgesia to both acute nociceptive and chronic neuropathic pain. The potency of clonidine increases with hypersensitivity to mechanical stimuli after nerve injury, although the reasons for this change are unknown. In the present study, we tested the hypothesis that peripheral nerve injury alters either spinal alpha2-adrenergic receptor-mediated G-protein activity or alpha2-adrenergic receptor number. Rats were randomized to left spinal nerve ligation (SNL) or sham surgery. Tactile hypersensitivity in the hindpaw was confirmed and lumbar spinal cords were removed for binding assays. To examine agonist-induced G-protein coupling, [35S]GTP gamma S binding experiments were performed in spinal cord membranes and sections using norepinephrine as an alpha2-adrenergic agonist. SNL was associated with an increase in maximal efficacy, but not potency, of norepinephrine-stimulated [35S]GTP gamma S binding in dorsal horn. SNL had no effect on basal [35S]GTP gamma S binding or on muscarinic cholinergic-stimulated [35S]GTP gamma S binding. [35S]GTP gamma S autoradiography showed that this increase in alpha2-adrenergic-activated G-proteins occurred both ipsilateral and contralateral to SNL surgery. SNL did not alter total alpha2-adrenergic receptor number or affinity to [3H]-rauwolscine binding, and displacement studies with the alpha2A-adrenergic antagonist BRL44408 revealed that most of the binding was associated with the alpha2A-adrenergic subtype. These data suggest that the increased potency of clonidine in neuropathic pain could reflect increased efficiency of G-protein coupling from spinal alpha2-adrenergic receptors.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Umbral del Dolor/fisiología , Receptores Adrenérgicos alfa 2/metabolismo , Transducción de Señal/fisiología , Médula Espinal/metabolismo , Nervios Espinales/metabolismo , Agonistas alfa-Adrenérgicos/farmacología , Animales , Modelos Animales de Enfermedad , Guanosina Trifosfato/metabolismo , Ligadura , Masculino , Mecanorreceptores/efectos de los fármacos , Mecanorreceptores/fisiología , Umbral del Dolor/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Nervios Espinales/lesiones , Yohimbina/farmacología
14.
Life Sci ; 77(10): 1140-54, 2005 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-15890372

RESUMEN

Previous studies have shown that chronic opiate treatment decreases mu opioid-stimulated [35S]GTPgammaS binding in specific brain regions. To extend these findings, the present study investigated DAMGO-stimulated [35S]GTPgammaS binding in membrane homogenates and coronal sections from rats non-contingently administered heroin. Rats were administered saline or increasing doses of heroin i.v. hourly up to 288 mg/kg/day over 40 days. In brain sections, chronic heroin administration decreased DAMGO-stimulated [35S]GTPgammaS binding in medial thalamus and amygdala, with no effect in cingulate cortex or nucleus accumbens. Chronic heroin administration also reduced [35S]GTPgammaS binding stimulated by the principal metabolite of heroin, 6-monoacetylmorphine. In contrast, no significant changes in mu opioid receptor binding were observed in amygdala or thalamus using [3H]DAMGO autoradiography. In membranes from amygdala and thalamus, chronic heroin treatment decreased the maximal effect of DAMGO in stimulating [35S]GTPgammaS binding, with no effect on DAMGO potency. GTPgammaS saturation analysis showed that chronic heroin treatment decreased the Bmax, and increased the K(D), of DAMGO-stimulated [35S]GTPgammaS binding. These data suggest potential mechanisms by which chronic agonist treatment produces opioid receptor/G-protein desensitization in brain.


Asunto(s)
Química Encefálica/efectos de los fármacos , Proteínas de Unión al GTP/efectos de los fármacos , Heroína/farmacología , Narcóticos/farmacología , Receptores Opioides mu/efectos de los fármacos , Analgésicos Opioides/metabolismo , Animales , Autorradiografía , Encefalina Ala(2)-MeFe(4)-Gli(5)/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Heroína/administración & dosificación , Masculino , Membranas/efectos de los fármacos , Membranas/metabolismo , Derivados de la Morfina/farmacología , Narcóticos/administración & dosificación , Prosencéfalo/efectos de los fármacos , Prosencéfalo/metabolismo , Ratas , Ratas Endogámicas F344 , Autoadministración
15.
J Neuroimmune Pharmacol ; 10(3): 493-505, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25749646

RESUMEN

Cocaine abuse in HIV patients accelerates the progression and severity of neuropathology, motor impairment and cognitive dysfunction compared to non-drug using HIV patients. Cocaine and HIV interact with the dopamine transporter (DAT); however, the effect of their interaction on DAT binding remains understudied. The present study compared the dose-response functions for intravenous self-administration of cocaine and heroin between male HIV-1 transgenic (HIV-1 Tg) and Fischer 344 rats. The cocaine and heroin dose-response functions exhibit an inverted U-shape for both HIV-1 Tg and F344 rats. For cocaine, the number of infusions for each dose on the ascending limb was greater for HIV-1 Tg versus F344 rats. No significant changes in the heroin dose-response function were observed in HIV-1 Tg animals. Following the conclusion of self-administration experiments, DAT binding was assessed in striatal membranes. Saturation binding of the cocaine analog [(125)I] 3ß-(4-iodophenyl)tropan-2ß-carboxylic acid methyl ester ([(125)I]RTI-55) in rat striatal membranes resulted in binding curves that were best fit to a two-site binding model, allowing for calculation of dissociation constant (Kd) and binding density (Bmax) values that correspond to high- and low-affinity DAT binding sites. Control HIV-1 Tg rats exhibited a significantly greater affinity (i.e., decrease in Kd value) in the low-affinity DAT binding site compared to control F344 rats. Furthermore, cocaine self-administration in HIV-1 Tg rats increased low-affinity Kd (i.e., decreased affinity) compared to levels observed in control F344 rats. Cocaine also increased low-affinity Bmax in HIV-1 Tg rats as compared to controls, indicating an increase in the number of low-affinity DAT binding sites. F344 rats did not exhibit any change in high- or low-affinity Kd or Bmax values following cocaine or heroin self-administration. The increase in DAT affinity in cocaine HIV-1 Tg rats is consistent with the leftward shift of the ascending limb of the cocaine dose-response curve observed in HIV-1 Tg vs. F344 rats, and has major implications for the function of cocaine binding to DAT in HIV patients. The absence of HIV-related changes in heroin intake are likely due to less dopaminergic involvement in the mediation of heroin reward, further emphasizing the preferential influence of HIV on dopamine-related behaviors.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cocaína/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Infecciones por VIH/metabolismo , Neostriado/metabolismo , Animales , Cocaína/administración & dosificación , Cocaína/análogos & derivados , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/efectos de los fármacos , Inhibidores de Captación de Dopamina/administración & dosificación , Relación Dosis-Respuesta a Droga , Heroína/administración & dosificación , Heroína/farmacología , Masculino , Narcóticos/administración & dosificación , Narcóticos/farmacología , Neostriado/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Ratas Transgénicas , Autoadministración
16.
Neuropharmacology ; 95: 492-502, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25724082

RESUMEN

The GABAB receptor is a therapeutic target for CNS and neuropathic disorders; however, few preclinical studies have explored effects of chronic stimulation. This study evaluated acute and chronic baclofen treatments on GABAB-activated G-proteins and signaling protein phosphorylation as indicators of GABAB signaling capacity. Brain sections from rats acutely administered baclofen (5 mg/kg, i.p.) showed no significant differences from controls in GABAB-stimulated GTPγS binding in any brain region, but displayed significantly greater phosphorylation/activation of focal adhesion kinase (pFAK(Tyr397)) in mesocorticolimbic regions (caudate putamen, cortex, hippocampus, thalamus) and elevated phosphorylated/activated glycogen synthase kinase 3-ß (pGSK3ß(Tyr216)) in the prefrontal cortex, cerebral cortex, caudate putamen, nucleus accumbens, thalamus, septum, and globus pallidus. In rats administered chronic baclofen (5 mg/kg, t.i.d. for five days), GABAB-stimulated GTPγS binding was significantly diminished in the prefrontal cortex, septum, amygdala, and parabrachial nucleus compared to controls. This effect was specific to GABAB receptors: there was no effect of chronic baclofen treatment on adenosine A1-stimulated GTPγS binding in any region. Chronically-treated rats also exhibited increases in pFAK(Tyr397) and pGSK3ß(Tyr216) compared to controls, and displayed wide-spread elevations in phosphorylated dopamine- and cAMP-regulated phosphoprotein-32 (pDARPP-32(Thr34)) compared to acutely-treated or control rats. We postulate that those neuroadaptive effects of GABAB stimulation mediated by G-proteins and their sequelae correlate with tolerance to several of baclofen's effects, whereas sustained signaling via kinase cascades points to cross-talk between GABAB receptors and alternative mechanisms that are resistant to desensitization. Both desensitized and sustained signaling pathways should be considered in the development of pharmacotherapies targeting the GABA system.


Asunto(s)
Baclofeno/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Agonistas de Receptores GABA-B/farmacología , Proteínas de Unión al GTP/metabolismo , Receptores de GABA-B/metabolismo , Animales , Autorradiografía , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Masculino , Fosforilación/efectos de los fármacos , Distribución Aleatoria , Ratas Sprague-Dawley , Factores de Tiempo , Tirosina/metabolismo
17.
Neuropharmacology ; 47(1): 81-91, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15165836

RESUMEN

Previous studies have indicated that cannabinoids inhibit presynaptic neurotransmitter release in brain through CB1 receptors. To examine this issue in a primary neuronal culture system, rat cerebellar granule cells (CGCs) were prepared. [35S]GTPgammaS binding assays in saponin-permeabilized CGCs showed that G-protein activation by the CB1 agonist, WIN55212-2, and adenosine A1 agonist, phenylisopropyladenosine, was maximal during the second week in culture. Delta9-tetrahydrocannabinol stimulated [35S]GTPgammaS binding to a lesser degree than WIN55212-2, and the antagonists SR141716A and AM281 acted as inverse agonists in intact CGCs, but not in CGC membrane preparations. Ten micromolar WIN55212-2 and Delta9-tetrahydrocannabinol decreased depolarization-evoked efflux of [3H]-D-aspartate from CGCs by 32% and 13%, respectively. SR141716A and AM281 increased [3H]-D-aspartate release by 28%. The fatty acid amide hydrolase (FAAH) inhibitor phenylmethylsulfonyl fluoride (PMSF) and the anandamide uptake inhibitor AM404 inhibited transmitter release, implying that the antagonist effects were mediated by blockade of endocannabinoid activity. Levels of endocannabinoids (both anandamide and 2-arachidonyl glycerol [2-AG]) in extracts of the cells and cell incubation buffer were increased by PMSF pre-treatment. Depolarization with KCl significantly decreased the amount of anandamide and 2-AG in PMSF-treated CGCs. These results suggest that endogenous cannabinoids inhibit neurotransmitter release in CGCs, which may also release endocannabioids upon neural stimulation.


Asunto(s)
Cannabinoides/farmacología , Cerebelo/fisiología , Ácido Glutámico/metabolismo , Neuronas/fisiología , Transducción de Señal/fisiología , Animales , Ácidos Araquidónicos/farmacología , Ácido Aspártico/metabolismo , Células Cultivadas , Cerebelo/efectos de los fármacos , Endocannabinoides , Femenino , Proteínas de Unión al GTP/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Cinética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Neuronas/efectos de los fármacos , Alcamidas Poliinsaturadas , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
18.
Neuropharmacology ; 45(2): 220-30, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12842128

RESUMEN

Opioid receptor-like (ORL-1) receptors and ORL-1-activated G-proteins are found in high levels in the forebrain, particularly cingulate cortex, an area involved in processing of nociceptive stimuli. [(3)H]nociceptin/orphanin FQ (N/OFQ) and N/OFQ-stimulated [(35)S]GTPgammaS autoradiography in rat brain were used to localize ORL-1 receptors and activated G-proteins, respectively. N/OFQ binding and activated G-proteins were highest in anterior cingulate, agranular insula, piriform, perirhinal and entorhinal cortices; midline and intralaminar thalamic nuclei; and subnuclei of the amygdala and hippocampus. In anterior cingulate area 24, [(3)H]N/OFQ and N/OFQ-stimulated [(35)S]GTPgammaS binding were highest in layers V and VI. The cellular localization of ORL-1 receptors and activated G-proteins in area 24 was examined using two strategies: ibotenic acid injection into the cortex or undercut lesions to remove afferent axons, followed by autoradiography. Ibotenic acid lesions that destroyed neurons in the anterior cingulate cortex decreased [(3)H]N/OFQ binding by 75-80% and reduced N/OFQ-stimulated [(35)S]GTPgammaS binding to basal levels seen in the absence of agonist. Deafferentation lesions increased [(3)H]N/OFQ binding by 40-50%, with no significant change in N/OFQ-stimulated [(35)S]GTPgammaS binding. These data demonstrate that ORL-1 receptors in layer V of anterior cingulate cortex are located on somatodendritic elements and that deafferentation increases ORL-1 receptor binding.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Giro del Cíngulo/metabolismo , Prosencéfalo/metabolismo , Receptores Opioides/metabolismo , Animales , Proteínas de Unión al GTP/análisis , Giro del Cíngulo/química , Masculino , Prosencéfalo/química , Unión Proteica/fisiología , Ratas , Ratas Long-Evans , Receptores Opioides/análisis , Receptor de Nociceptina
19.
Neuropharmacology ; 47 Suppl 1: 345-58, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15464149

RESUMEN

Delta9-Tetrahydrocannabinol from Cannabis sativa is mimicked by cannabimimetic analogs such as CP55940 and WIN55212-2, and antagonized by rimonabant and SR144528, through G-protein-coupled receptors, CB1 in the brain, and CB2 in the immune system. Eicosanoids anandamide and 2-arachidonoylglycerol are the "endocannabinoid" agonists for these receptors. CB1 receptors are abundant in basal ganglia, hippocampus and cerebellum, and their functional activity can be mapped during behaviors using cerebral metabolism as the neuroimaging tool. CB1 receptors couple to G(i/o) to inhibit cAMP production, decrease Ca2+ conductance, increase K+ conductance, and increase mitogen-activated protein kinase activity. Functional activation of G-proteins can be imaged by [35S]GTPgammaS autoradiography. Post-synaptically generated endocannabinoids form the basis of a retrograde signaling mechanism referred to as depolarization-induced suppression of inhibition (DSI) or excitation (DSE). Under circumstances of sufficient intracellular Ca2+ (e.g., burst activity in seizures), synthesis of endocannabinoids releases a diffusible retrograde messenger to stimulate presynaptic CB1 receptors. This results in suppression of gamma-aminobutyric acid (GABA) release, thereby relieving the post-synaptic inhibition. Tolerance develops as neurons adjust both receptor number and cellular signal transduction to the chronic administration of cannabinoid drugs. Future therapeutic drug design can progress based upon our current understanding of the physiology and pharmacology of CB1, CB2 and related receptors. One very important role for CB1 antagonists will be in the treatment of craving in the disease of substance abuse.


Asunto(s)
Cannabinoides/farmacología , Receptores de Cannabinoides/fisiología , Animales , Agonistas de Receptores de Cannabinoides , Moduladores de Receptores de Cannabinoides/fisiología , Cannabis/química , Humanos , Receptores de Cannabinoides/efectos de los fármacos , Investigación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
20.
Life Sci ; 71(19): 2217-26, 2002 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-12215369

RESUMEN

Cannabinoid receptors are found in moderate density throughout the cerebral cortex. The anterior cingulate cortex (ACC) is of particular interest due its high level of cannabinoid receptors and role in behaviors known to be modulated by cannabinoids. These studies were conducted to determine the cellular localization of cannabinoid receptors and to compare the level of cannabinoid receptor binding with receptor-mediated G-protein activity in the rat ACC. Either ibotenic acid or undercut lesions were made in ACC, and brains were processed for [3H]WIN 55,212-2 and WIN 55,212-2-stimulated [35S]GTPgammaS autoradiography. Both cannabinoid receptors and receptor-activated G-proteins were highest in laminae I and VI of ACC in control tissue. Although similar levels of receptor binding were found in these laminae, significantly higher levels of receptor-activated G-proteins were found in lamina VI. Ibotenic acid lesions that destroyed ACC neurons decreased [3H]WIN 55,212-2 binding by 60-70% and eliminated WIN 55,212-2-stimulated [35S]GTPgammaS binding. In contrast, deafferentation of the ACC with undercut lesions had no significant effect on cannabinoid receptor binding or G-protein activation. These results indicate that cannabinoid receptors in laminae I and VI of the ACC are located on somatodendritic elements or axons intrinsic to the ACC. In addition, differences in the relative levels of cannabinoid binding sites and activated G-proteins between cortical laminae indicate that the efficiency of cannabinoid receptors for G-protein activation may vary within a specific brain region.


Asunto(s)
Proteínas de Unión al GTP/biosíntesis , Giro del Cíngulo/metabolismo , Neuronas/metabolismo , Receptores de Droga/metabolismo , Animales , Autorradiografía , Benzoxazinas , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Giro del Cíngulo/citología , Giro del Cíngulo/patología , Ácido Iboténico/farmacología , Procesamiento de Imagen Asistido por Computador , Masculino , Morfolinas/metabolismo , Morfolinas/farmacología , Naftalenos/metabolismo , Naftalenos/farmacología , Neuronas/citología , Neuronas/patología , Ratas , Ratas Long-Evans , Receptores de Cannabinoides , Radioisótopos de Azufre , Tritio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA