Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
bioRxiv ; 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38659831

RESUMEN

The therapeutic benefits of opioids are compromised by the development of analgesic tolerance, which necessitates higher dosing for pain management thereby increasing the liability for dependence and addiction. Rodent models indicate opposing roles of the gut microbiota in tolerance: morphine-induced gut dysbiosis exacerbates tolerance, whereas probiotics ameliorate tolerance. Not all individuals develop tolerance which could be influenced by differences in microbiota, and yet no study has capitalized upon this natural variation to identify specific features linked to tolerance. We leveraged this natural variation in a murine model of voluntary oral morphine self-administration to elucidate the mechanisms by which microbiota influences tolerance. Although all mice shared similar and predictive morphine-driven microbiota changes that largely masked informative associations with variability in tolerance, our high-resolution temporal analyses revealed a divergence in the progression of dysbiosis that best explained differences in the development in tolerance. Mice that did not develop tolerance also maintained a higher abundance of taxa capable of producing the short-chain fatty acid (SCFA) butyrate, known to bolster intestinal barriers, suppress inflammation, and promote neuronal homeostasis. Furthermore, dietary butyrate supplementation significantly reduced the development of tolerance. These findings could inform immediate therapies to extend the analgesic efficacy of opioids.

2.
bioRxiv ; 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38562752

RESUMEN

Opioid drugs are potent analgesics that mimic the endogenous opioid peptides, endorphins and enkephalins, by activating the µ-opioid receptor. Opioid use is limited by side effects, including significant risk of opioid use disorder. Improvement of the effect/side effect profile of opioid medications is a key pursuit of opioid research, yet there is no consensus on how to achieve this goal. One hypothesis is that the degree of arrestin-3 recruitment to the µ-opioid receptor impacts therapeutic utility. However, it is not clear whether increased or decreased interaction of the µ-opioid receptor with arrestin-3 would reduce compulsive drug-seeking. To examine this question, we utilized three genotypes of mice with varying abilities to recruit arrestin-3 to the µ-opioid receptor in response to morphine in a novel longitudinal operant self-administration model. We demonstrate that arrestin-3 knockout and wild type mice have highly variable drug-seeking behavior with few genotype differences. In contrast, in mice where the µ-opioid receptor strongly recruits arrestin-3, drug-seeking behavior is much less varied. We created a quantitative method to define compulsivity in drug-seeking and found that mice lacking arrestin-3 were more likely to meet the criteria for compulsivity whereas mice with enhanced arrestin-3 recruitment did not develop a compulsive phenotype. Our data suggest that opioids that engage both G protein and arrestin-3, recapitulating the endogenous signaling pattern, will reduce abuse liability.

3.
Annu Rev Physiol ; 86: 1-25, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38029388

RESUMEN

The harmful side effects of opioid drugs such as respiratory depression, tolerance, dependence, and abuse potential have limited the therapeutic utility of opioids for their entire clinical history. However, no previous attempt to develop effective pain drugs that substantially ameliorate these effects has succeeded, and the current opioid epidemic affirms that they are a greater hindrance to the field of pain management than ever. Recent attempts at new opioid development have sought to reduce these side effects by minimizing engagement of the regulatory protein arrestin-3 at the mu-opioid receptor, but there is significant controversy around this approach. Here, we discuss the ongoing effort to develop safer opioids and its relevant historical context. We propose a new model that reconciles results previously assumed to be in direct conflict to explain how different signaling profiles at the mu-opioid receptor contribute to opioid tolerance and dependence. Our goal is for this framework to inform the search for a new generation of lower liability opioid analgesics.


Asunto(s)
Analgésicos Opioides , Transducción de Señal , Humanos , Analgésicos Opioides/efectos adversos , Tolerancia a Medicamentos
4.
Biol Psychiatry ; 94(7): 531-542, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36931452

RESUMEN

BACKGROUND: Second-generation antipsychotics (SGAs) are frontline treatments for serious mental illness. Often, individual patients benefit only from some SGAs and not others. The mechanisms underlying this unpredictability in treatment efficacy remain unclear. All SGAs bind the dopamine D3 receptor (D3R) and are traditionally considered antagonists for dopamine receptor signaling. METHODS: Here, we used a combination of two-photon calcium imaging, in vitro signaling assays, and mouse behavior to assess signaling by SGAs at D3R. RESULTS: We report that some clinically important SGAs function as arrestin-3 agonists at D3R, resulting in modulation of calcium channels localized to the site of action potential initiation in prefrontal cortex pyramidal neurons. We further show that chronic treatment with an arrestin-3 agonist SGA, but not an antagonist SGA, abolishes D3R function through postendocytic receptor degradation by GASP1 (G protein-coupled receptor-associated sorting protein-1). CONCLUSIONS: These results implicate D3R-arrestin-3 signaling as a source of SGA variability, highlighting the importance of including arrestin-3 signaling in characterizations of drug action. Furthermore, they suggest that postendocytic receptor trafficking that occurs during chronic SGA treatment may contribute to treatment efficacy.


Asunto(s)
Antipsicóticos , Dopamina , Ratones , Animales , Arrestina beta 2/metabolismo , Antipsicóticos/farmacología , Receptores de Dopamina D3/metabolismo , Agonistas de Dopamina/farmacología , Tolerancia a Medicamentos , Receptores de Dopamina D1/metabolismo
5.
Neuropsychopharmacology ; 46(13): 2241-2249, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34257415

RESUMEN

Opioid drugs are widely used analgesics that activate the G protein-coupled µ-opioid receptor, whose endogenous neuropeptide agonists, endorphins and enkephalins, are potent pain relievers. The therapeutic utility of opioid drugs is hindered by development of tolerance to the analgesic effects, requiring dose escalation for persistent pain control and leading to overdose and fatal respiratory distress. The prevailing hypothesis is that the intended analgesic effects of opioid drugs are mediated by µ-opioid receptor signaling to G protein, while the side-effects of respiratory depression and analgesic tolerance are caused by engagement of the receptor with the arrestin-3 protein. Consequently, opioid drug development has focused exclusively on identifying agonists devoid of arrestin-3 engagement. Here, we challenge the prevailing hypothesis with a panel of six clinically relevant opioid drugs and mice of three distinct genotypes with varying abilities to promote morphine-mediated arrestin-3 engagement. With this genetic and pharmacological approach, we demonstrate that arrestin-3 recruitment does not impact respiratory depression, and effective arrestin-3 engagement reduces, rather than exacerbates, the development of analgesic tolerance. These studies suggest that future development of safer opioids should focus on identifying opioid ligands that recruit both G protein and arrestin-3, thereby mimicking the signaling profile of most endogenous µ-opioid receptor agonists.


Asunto(s)
Receptores Opioides , Insuficiencia Respiratoria , Analgésicos , Analgésicos Opioides/farmacología , Animales , Tolerancia a Medicamentos , Ratones , Morfina/farmacología , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico , Arrestina beta 2/metabolismo
6.
ACS Chem Neurosci ; 8(3): 473-485, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-27744679

RESUMEN

Opioid therapeutics are excellent analgesics, whose utility is compromised by dependence. Morphine (1) and its clinically relevant derivatives such as OxyContin (2), Vicodin (3), and Dilaudid (4) are "biased" agonists at the µ opioid receptor (OR), wherein they engage G protein signaling but poorly engage ß-arrestin and the endocytic machinery. In contrast, endorphins, the endogenous peptide agonists for ORs, are potent analgesics, show reduced liability for tolerance and dependence, and engage both G protein and ß-arrestin pathways as "balanced" agonists. We set out to determine if marine-derived alkaloids could serve as novel OR agonist chemotypes with a signaling profile distinct from morphine and more similar to the endorphins. Screening of 96 sponge-derived extracts followed by LC-MS-based purification to pinpoint the active compounds and subsequent evaluation of a mini library of related alkaloids identified two structural classes that modulate the ORs. These included the following: aaptamine (10), 9-demethyl aaptamine (11), demethyl (oxy)-aaptamine (12) with activity at the δ-OR (EC50: 5.1, 4.1, 2.3 µM, respectively) and fascaplysin (17), and 10-bromo fascaplysin (18) with activity at the µ-OR (EC50: 6.3, 4.2 µM respectively). An in vivo evaluation of 10 using δ-KO mice indicated its previously reported antidepressant-like effects are dependent on the δ-OR. Importantly, 17 functioned as a balanced agonist promoting both G protein signaling and ß-arrestin recruitment along with receptor endocytosis similar to the endorphins. Collectively these results demonstrate the burgeoning potential for marine natural products to serve as novel lead compounds for therapeutic targets in neuroscience research.


Asunto(s)
Analgésicos Opioides , Endorfinas/farmacología , Naftiridinas , Receptores Opioides delta/metabolismo , Transducción de Señal/efectos de los fármacos , Analgésicos Opioides/química , Analgésicos Opioides/aislamiento & purificación , Analgésicos Opioides/farmacología , Animales , Simulación por Computador , AMP Cíclico/metabolismo , Endocitosis/efectos de los fármacos , Endorfinas/química , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Indoles/química , Indoles/aislamiento & purificación , Indoles/farmacología , Locomoción/efectos de los fármacos , Locomoción/genética , Masculino , Ratones , Ratones Transgénicos , Modelos Moleculares , Naftiridinas/química , Naftiridinas/aislamiento & purificación , Naftiridinas/farmacología , Poríferos/química , Receptores Opioides delta/genética , Transducción de Señal/genética , Espectrometría de Masa por Ionización de Electrospray , Natación/psicología , beta-Arrestinas/metabolismo
7.
Cell Rep ; 17(12): 3233-3245, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28009292

RESUMEN

Neural circuits involving midbrain dopaminergic (DA) neurons regulate reward and goal-directed behaviors. Although local GABAergic input is known to modulate DA circuits, the mechanism that controls excitatory/inhibitory synaptic balance in DA neurons remains unclear. Here, we show that DA neurons use autocrine transforming growth factor ß (TGF-ß) signaling to promote the growth of axons and dendrites. Surprisingly, removing TGF-ß type II receptor in DA neurons also disrupts the balance in TGF-ß1 expression in DA neurons and neighboring GABAergic neurons, which increases inhibitory input, reduces excitatory synaptic input, and alters phasic firing patterns in DA neurons. Mice lacking TGF-ß signaling in DA neurons are hyperactive and exhibit inflexibility in relinquishing learned behaviors and re-establishing new stimulus-reward associations. These results support a role for TGF-ß in regulating the delicate balance of excitatory/inhibitory synaptic input in local microcircuits involving DA and GABAergic neurons and its potential contributions to neuropsychiatric disorders.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Aprendizaje Inverso/fisiología , Factor de Crecimiento Transformador beta1/genética , Animales , Dendritas/genética , Dendritas/fisiología , Neuronas Dopaminérgicas/fisiología , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Regulación de la Expresión Génica , Humanos , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/metabolismo , Ratones , Receptor Tipo II de Factor de Crecimiento Transformador beta , Transducción de Señal/genética , Sinapsis/genética , Sinapsis/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
8.
Cell Rep ; 16(6): 1518-1526, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27452469

RESUMEN

G-protein-coupled receptors (GPCRs) initiate a variety of signaling cascades, depending on effector coupling. ß-arrestins, which were initially characterized by their ability to "arrest" GPCR signaling by uncoupling receptor and G protein, have recently emerged as important signaling effectors for GPCRs. ß-arrestins engage signaling pathways that are distinct from those mediated by G protein. As such, arrestin-dependent signaling can play a unique role in regulating cell function, but whether neuromodulatory GPCRs utilize ß-arrestin-dependent signaling to regulate neuronal excitability remains unclear. Here, we find that D3 dopamine receptors (D3R) regulate axon initial segment (AIS) excitability through ß-arrestin-dependent signaling, modifying CaV3 voltage dependence to suppress high-frequency action potential generation. This non-canonical D3R signaling thereby gates AIS excitability via pathways distinct from classical GPCR signaling pathways.


Asunto(s)
Segmento Inicial del Axón/metabolismo , Canales de Calcio/metabolismo , Dopamina/metabolismo , beta-Arrestinas/metabolismo , Animales , Calcio/metabolismo , Células HEK293 , Humanos , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo
9.
Cell Rep ; 14(12): 2774-83, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26997280

RESUMEN

Kappa opioid receptors (KORs) are involved in a variety of aversive behavioral states, including anxiety. To date, a circuit-based mechanism for KOR-driven anxiety has not been described. Here, we show that activation of KORs inhibits glutamate release from basolateral amygdala (BLA) inputs to the bed nucleus of the stria terminalis (BNST) and occludes the anxiolytic phenotype seen with optogenetic activation of BLA-BNST projections. In addition, deletion of KORs from amygdala neurons results in an anxiolytic phenotype. Furthermore, we identify a frequency-dependent, optically evoked local dynorphin-induced heterosynaptic plasticity of glutamate inputs in the BNST. We also find that there is cell type specificity to the KOR modulation of the BLA-BNST input with greater KOR-mediated inhibition of BLA dynorphin-expressing neurons. Collectively, these results provide support for a model in which local dynorphin release can inhibit an anxiolytic pathway, providing a discrete therapeutic target for the treatment of anxiety disorders.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Ansiedad , Dinorfinas/farmacología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Amígdala del Cerebelo/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Channelrhodopsins , Potenciales Evocados/efectos de los fármacos , Ácido Glutámico/farmacología , Imidazoles/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Microscopía Fluorescente , Técnicas de Placa-Clamp , Piridinas/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Núcleos Septales/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Mol Cell Endocrinol ; 382(2): 938-49, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24275181

RESUMEN

The glucagon-like peptide-1 incretin receptor (GLP-1R) of family B G protein-coupled receptors (GPCRs) is a major drug target in type-2-diabetes due to its regulatory effect on post-prandial blood-glucose levels. The mechanism(s) controlling GLP-1R mediated signaling are far from fully understood. A fundamental mechanism controlling the signaling capacity of GPCRs is the post-endocytic trafficking of receptors between recycling and degradative fates. Here, we combined microscopy with novel real-time assays to monitor both receptor trafficking and signaling in living cells. We find that the human GLP-1R internalizes rapidly and with similar kinetics in response to equipotent concentrations of GLP-1 and the stable GLP-1 analogues exendin-4 and liraglutide. Receptor internalization was confirmed in mouse pancreatic islets. GLP-1R is shown to be a recycling receptor with faster recycling rates mediated by GLP-1 as compared to exendin-4 and liraglutide. Furthermore, a prolonged cycling of ligand-activated GLP-1Rs was observed and is suggested to be correlated with a prolonged cAMP signal.


Asunto(s)
Péptido 1 Similar al Glucagón/farmacología , Islotes Pancreáticos/metabolismo , Receptores de Glucagón/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , AMP Cíclico/metabolismo , Exenatida , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón , Células HEK293 , Humanos , Incretinas/metabolismo , Incretinas/farmacología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/ultraestructura , Liraglutida , Ratones , Ratones Endogámicos C57BL , Péptidos/metabolismo , Péptidos/farmacología , Estabilidad Proteica , Transporte de Proteínas , Proteolisis , Imagen de Lapso de Tiempo , Ponzoñas/metabolismo , Ponzoñas/farmacología
11.
Cell ; 154(5): 1085-1099, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23954414

RESUMEN

The molecular mechanism of autophagy and its relationship to other lysosomal degradation pathways remain incompletely understood. Here, we identified a previously uncharacterized mammalian-specific protein, Beclin 2, which, like Beclin 1, functions in autophagy and interacts with class III PI3K complex components and Bcl-2. However, Beclin 2, but not Beclin 1, functions in an additional lysosomal degradation pathway. Beclin 2 is required for ligand-induced endolysosomal degradation of several G protein-coupled receptors (GPCRs) through its interaction with GASP1. Beclin 2 homozygous knockout mice have decreased embryonic viability, and heterozygous knockout mice have defective autophagy, increased levels of brain cannabinoid 1 receptor, elevated food intake, and obesity and insulin resistance. Our findings identify Beclin 2 as a converging regulator of autophagy and GPCR turnover and highlight the functional and mechanistic diversity of Beclin family members in autophagy, endolysosomal trafficking, and metabolism.


Asunto(s)
Autofagia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Lisosomas/metabolismo , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Obesidad/metabolismo , Alineación de Secuencia
12.
J Neurosci ; 33(30): 12329-36, 2013 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-23884939

RESUMEN

Potentiation of glutamate responses is a critical synaptic response to cocaine exposure in ventral tegmental area (VTA) neurons. However, the mechanism by which cocaine exposure promotes potentiation of NMDA receptors (NMDARs) and subsequently AMPA receptors (AMPARs) is not fully understood. In this study we demonstrate that repeated cocaine treatment causes loss of D2 dopamine receptor functional responses via interaction with lysosome-targeting G-protein-associated sorting protein1 (GASP1). We also show that the absence of D2 downregulation in GASP1-KO mice prevents cocaine-induced potentiation of NMDAR currents, elevation of the AMPA/NMDA ratio, and redistribution of NMDAR and AMPAR subunits to the membrane. As a pharmacological parallel, coadministration of the high-affinity D2 agonist, aripiprazole, reduces not only functional downregulation of D2s in response to cocaine but also potentiation of NMDAR and AMPAR responses in wild-type mice. Together these data suggest that functional loss of D2 receptors is a critical mechanism mediating cocaine-induced glutamate plasticity in VTA neurons.


Asunto(s)
Cocaína/farmacología , Receptores AMPA/fisiología , Receptores de Dopamina D2/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/fisiología , Animales , Antipsicóticos/farmacología , Aripiprazol , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Ácido Glutámico/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Técnicas de Cultivo de Órganos , Piperazinas/farmacología , Quinolonas/farmacología , Receptores de Dopamina D2/agonistas , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/fisiología
13.
Psychopharmacology (Berl) ; 228(1): 1-18, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23649885

RESUMEN

RATIONALE: Delta opioid receptors (DORs) have been considered as a potential target to relieve pain as well as treat depression and anxiety disorders and are known to modulate other physiological responses, including ethanol and food consumption. A small number of DOR-selective drugs are in clinical trials, but no DOR-selective drugs have been approved by the Federal Drug Administration and some candidates have failed in phase II clinical trials, highlighting current difficulties producing effective delta opioid-based therapies. Recent studies have provided new insights into the pharmacology of the DOR, which is often complex and at times paradoxical. OBJECTIVE: This review will discuss the existing literature focusing on four aspects: (1) Two DOR subtypes have been postulated based on differences in pharmacological effects of existing DOR-selective ligands. (2) DORs are expressed ubiquitously throughout the body and central nervous system and are, thus, positioned to play a role in a multitude of diseases. (3) DOR expression is often dynamic, with many reports of increased expression during exposure to chronic stimuli, such as stress, inflammation, neuropathy, morphine, or changes in endogenous opioid tone. (4) A large structural variety in DOR ligands implies potential different mechanisms of activating the receptor. CONCLUSION: The reviewed features of DOR pharmacology illustrate the potential benefit of designing tailored or biased DOR ligands.


Asunto(s)
Analgésicos Opioides/farmacología , Diseño de Fármacos , Receptores Opioides delta/efectos de los fármacos , Animales , Ensayos Clínicos como Asunto , Humanos , Ligandos , Terapia Molecular Dirigida , Receptores Opioides delta/metabolismo
14.
PLoS One ; 8(3): e58362, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23554887

RESUMEN

Delta (DOR) and mu opioid receptors (MOR) can complex as heteromers, conferring functional properties in agonist binding, signaling and trafficking that can differ markedly from their homomeric counterparts. Because of these differences, DOR/MOR heteromers may be a novel therapeutic target in the treatment of pain. However, there are currently no ligands selective for DOR/MOR heteromers, and, consequently, their role in nociception remains unknown. In this study, we used a pharmacological opioid cocktail that selectively activates and stabilizes the DOR/MOR heteromer at the cell surface by blocking its endocytosis to assess its role in antinociception. We found that mice treated chronically with this drug cocktail showed a significant right shift in the ED50 for opioid-mediated analgesia, while mice treated with a drug that promotes degradation of the heteromer did not. Furthermore, promoting degradation of the DOR/MOR heteromer after the right shift in the ED50 had occurred, or blocking signal transduction from the stabilized DOR/MOR heteromer, shifted the ED50 for analgesia back to the left. Taken together, these data suggest an anti-analgesic role for the DOR/MOR heteromer in pain. In conclusion, antagonists selective for DOR/MOR heteromer could provide an avenue for alleviating reduced analgesic response during chronic pain treatment.


Asunto(s)
Dolor/metabolismo , Multimerización de Proteína , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacología , Animales , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Dolor/tratamiento farmacológico , Dolor/genética , Dolor/patología , Manejo del Dolor , Estabilidad Proteica/efectos de los fármacos , Receptores Opioides delta/genética , Receptores Opioides mu/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
15.
Methods Mol Biol ; 995: 43-54, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23494371

RESUMEN

Calcium signaling plays a major role in the function of cells. Measurement of intracellular calcium mobilization is a robust assay that can be performed in a high-throughput manner to study the effect of compounds on potential drug targets. Pharmaceutical companies frequently use calcium signaling assays to screen compound libraries on G-protein-coupled receptors (GPCRs). In this chapter we describe the application of FLIPR technology to the evaluation of GPCR-induced calcium mobilization. We also include the implications of GPCR hetero-oligomerization and the identification of heteromeric receptors as novel drug targets on high-throughput calcium screening.


Asunto(s)
Compuestos de Bencilideno/farmacología , Evaluación Preclínica de Medicamentos/métodos , Encefalina D-Penicilamina (2,5)/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Naltrexona/análogos & derivados , Receptores Opioides delta/agonistas , Señalización del Calcio , Técnicas de Cultivo de Célula , Células HEK293 , Humanos , Naltrexona/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides delta/biosíntesis , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/biosíntesis , Espectrometría de Fluorescencia , Transfección
16.
J Pharmacol Exp Ther ; 344(1): 179-88, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097213

RESUMEN

Drugs targeting G-protein-coupled receptors (GPCRs) make up more than 25% of all prescribed medicines. The ability of GPCRs to form heteromers with unique signaling properties suggests an entirely new and unexplored pool of drug targets. However, current in vitro assays are ill equipped to detect heteromer-selective compounds. We have successfully adapted an approach, using fusion proteins of GPCRs and chimeric G proteins, to create an in vitro screening assay (in human embryonic kidney cells) in which only activated heteromers are detectable. Here we show that this assay can demonstrate heteromer-selective G-protein bias as well as measure transinhibition. Using this assay, we reveal that the δ-opioid receptor agonist ADL5859, which is currently in clinical trials, has a 10-fold higher potency against δ-opioid receptor homomers than δ/µ-opioid receptor heteromers (pEC(50) = 6.7 ± 0.1 versus 5.8 ± 0.2). The assay enables the screening of large compound libraries to identify heteromer-selective compounds that could then be used in vivo to determine the functional role of heteromers and develop potential therapeutic agents.


Asunto(s)
Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/efectos de los fármacos , Secuencia de Aminoácidos , Benzamidas/farmacología , Calcio/metabolismo , Señalización del Calcio , Clonación Molecular , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Encefalina Leucina/farmacología , Ensayo de Inmunoadsorción Enzimática , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Células HEK293 , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/química , Proteínas Mutantes Quiméricas/genética , Piperazinas/farmacología , Reacción en Cadena de la Polimerasa , Receptores Acoplados a Proteínas G/genética , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides delta/genética , Proteínas Recombinantes de Fusión
17.
Neuropsychopharmacology ; 37(11): 2436-45, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22739468

RESUMEN

Alleviating anxiety and depression is pivotal for reducing the risk of relapse in alcoholics. Currently available anxiolytic treatments are limited by side effects, including reduced efficacy in alcoholics, addiction, and sedation. We examined whether the neuropeptide S receptor (NPSR) was effective at controlling ethanol consumption and the anxiety and depression produced by forced abstinence from ethanol. We found that the anxiolytic and anti-depressant effects of NPS are enhanced in acute ethanol abstinent mice. In addition, we found that NPS reduced ethanol consumption and is not in and of itself rewarding. We also provide evidence that ethanol consumption increases the ability of NPS to modulate neuronal activity in the basolateral amygdala. Finally, we found that local injection of NPS in the basolateral amygdala promotes anxiolysis after chronic ethanol consumption, thereby providing insight into the molecular mechanism underlying the changes in behavioral response to NPS. In light of the improved anxiolytic efficacy and benign side effects of NPS in ethanol-withdrawn animals, the NPSR may prove a suitable target for reducing relapse in alcoholism.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Ansiolíticos/uso terapéutico , Antidepresivos/uso terapéutico , Ansiedad/tratamiento farmacológico , Depresión/tratamiento farmacológico , Etanol/administración & dosificación , Neuropéptidos/uso terapéutico , Adaptación Ocular/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Análisis de Varianza , Animales , Condicionamiento Operante/efectos de los fármacos , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Sinergismo Farmacológico , Etanol/metabolismo , Suspensión Trasera/métodos , Técnicas In Vitro , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Natación/psicología , Factores de Tiempo
18.
J Am Chem Soc ; 134(25): 10321-4, 2012 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-22671299

RESUMEN

The bioconjugation of organometallic complexes with peptides has proven to be a novel approach for drug discovery. We report the facile and chemoselective reaction of tyrosine-containing G-protein-coupled receptor (GPCR) peptides with [Cp*Rh(H(2)O)(3)](OTf)(2), in water, at room temperature, and at pH 5-6. We have focused on three important GPCR peptides; namely, [Tyr(1)]-leu-enkephalin, [Tyr(4)]-neurotensin(8-13), and [Tyr(3)]-octreotide, each of which has a different position for the tyrosine residue, together with competing functionalities. Importantly, all other functional groups present, i.e., amino, carboxyl, disulfide, phenyl, and indole, were not prominent sites of reactivity by the Cp*Rh tris aqua complex. Furthermore, the influence of the Cp*Rh moiety on the structure of [Tyr(3)]-octreotide was characterized by 2D NMR, resulting in the first representative structure of an organometallic-peptide complex. The biological consequences of these Cp*Rh-peptide complexes, with respect to GPCR binding and growth inhibition of MCF7 and HT29 cancer cells, will be presented for [(η(6)-Cp*Rh-Tyr(1))-leu-enkephalin](OTf)(2) and [(η(6)-Cp*Rh-Tyr(3))-octreotide](OTf)(2).


Asunto(s)
Modelos Moleculares , Compuestos Organometálicos/química , Péptidos/química , Receptores Acoplados a Proteínas G/química , Rodio/química , Tirosina/química , Unión Competitiva , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Células HT29 , Humanos , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética , Compuestos Organometálicos/farmacología , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/metabolismo
19.
PLoS One ; 7(3): e33329, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22442685

RESUMEN

Prostaglandin H(1) (PGH(1)) is the cyclo-oxygenase metabolite of dihomo-γ-linolenic acid (DGLA) and the precursor for the 1-series of prostaglandins which are often viewed as "anti-inflammatory". Herein we present evidence that PGH(1) is a potent activator of the pro-inflammatory PGD(2) receptor CRTH2, an attractive therapeutic target to treat allergic diseases such as asthma and atopic dermatitis. Non-invasive, real time dynamic mass redistribution analysis of living human CRTH2 transfectants and Ca(2+) flux studies reveal that PGH(1) activates CRTH2 as PGH(2), PGD(2) or PGD(1) do. The PGH(1) precursor DGLA and the other PGH(1) metabolites did not display such effect. PGH(1) specifically internalizes CRTH2 in stable CRTH2 transfectants as assessed by antibody feeding assays. Physiological relevance of CRTH2 ligation by PGH(1) is demonstrated in several primary human hematopoietic lineages, which endogenously express CRTH2: PGH(1) mediates migration of and Ca(2+) flux in Th2 lymphocytes, shape change of eosinophils, and their adhesion to human pulmonary microvascular endothelial cells under physiological flow conditions. All these effects are abrogated in the presence of the CRTH2 specific antagonist TM30089. Together, our results identify PGH(1) as an important lipid intermediate and novel CRTH2 agonist which may trigger CRTH2 activation in vivo in the absence of functional prostaglandin D synthase.


Asunto(s)
Células Endoteliales/metabolismo , Prostaglandinas H/metabolismo , Receptores Inmunológicos/agonistas , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/metabolismo , Células Th2/metabolismo , Señalización del Calcio/genética , Femenino , Células HEK293 , Humanos , Hipersensibilidad/tratamiento farmacológico , Hipersensibilidad/genética , Hipersensibilidad/metabolismo , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/genética , Lipocalinas/metabolismo , Masculino , Prostaglandinas H/genética , Receptores Inmunológicos/genética , Receptores de Prostaglandina/genética
20.
Drug Alcohol Depend ; 121(3): 189-204, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22226706

RESUMEN

Opioid drugs remain the gold standard for the treatment of severe pain, both acute/post-surgical and chronic. However, the utility of opioid drugs for the treatment of chronic pain is compromised by the development of analgesic tolerance which, in turn, leads to dose-escalation and increased likelihood of dangerous side effects, including dependence. Consequently, there remains resistance among clinicians and the general population to using opiates for pain management because of risk of "addiction." These fears are not unwarranted. More than 2.5 million people begin abusing opioid painkillers each year, and prescription opioid abuse is now the second most common type of illegal drug use after marijuana. Some abusers become dependent due to recreational use of prescription painkillers. However, many abusers are among the 40 million people suffering from chronic pain, and developed dependence while using the drugs for legitimate purposes. Both of these trends highlight the need to develop opioid therapeutics with a reduced liability to cause tolerance, dependence and addiction. Identifying the ideal properties of opioid drugs that would retain analgesia but reduce these side-effects has been a goal of my laboratory for more than a decade. During this time, we have proposed the novel hypothesis that opioid drugs that promote desensitization, endocytosis and recycling of the mu-opioid-receptor (MOR) will retain analgesic efficacy, but will have a reduced liability to cause tolerance, dependence and addiction. We have generated substantial data, both pharmacological and genetic to suggest that our hypothesis is a valid one. These data are summarized in this review.


Asunto(s)
Analgésicos Opioides/farmacología , Endocitosis/fisiología , Trastornos Relacionados con Opioides/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/uso terapéutico , Animales , Tolerancia a Medicamentos , Endocitosis/efectos de los fármacos , Humanos , Ratones , Dolor/tratamiento farmacológico , Dolor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...