Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Nat Neurosci ; 27(9): 1844-1857, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39009835

RESUMO

Neuropeptides are ubiquitous in the nervous system. Research into neuropeptides has been limited by a lack of experimental tools that allow for the precise dissection of their complex and diverse dynamics in a circuit-specific manner. Opioid peptides modulate pain, reward and aversion and as such have high clinical relevance. To illuminate the spatiotemporal dynamics of endogenous opioid signaling in the brain, we developed a class of genetically encoded fluorescence sensors based on kappa, delta and mu opioid receptors: κLight, δLight and µLight, respectively. We characterized the pharmacological profiles of these sensors in mammalian cells and in dissociated neurons. We used κLight to identify electrical stimulation parameters that trigger endogenous opioid release and the spatiotemporal scale of dynorphin volume transmission in brain slices. Using in vivo fiber photometry in mice, we demonstrated the utility of these sensors in detecting optogenetically driven opioid release and observed differential opioid release dynamics in response to fearful and rewarding conditions.


Assuntos
Técnicas Biossensoriais , Optogenética , Animais , Técnicas Biossensoriais/métodos , Camundongos , Optogenética/métodos , Neurônios/metabolismo , Humanos , Dinorfinas/metabolismo , Dinorfinas/genética , Masculino , Peptídeos Opioides/metabolismo , Peptídeos Opioides/genética , Células HEK293 , Camundongos Endogâmicos C57BL , Encéfalo/metabolismo , Neuropeptídeos/metabolismo , Neuropeptídeos/genética , Receptores Opioides/metabolismo , Receptores Opioides/genética , Estimulação Elétrica , Recompensa
2.
bioRxiv ; 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-39005415

RESUMO

It is well established that dopamine neurons of the ventral tegmental area (VTA) play a critical role in reward and aversion as well as pathologies including drug dependence and addiction. The distinct effects of acute and chronic opioid exposure have been previously characterized at VTA synapses. Recent work suggests that distinct VTA projections that target the medial and lateral shell of the nucleus accumbens (NAc), may play opposing roles in modulating behavior. It is possible that these two anatomically and functionally distinct pathways also have disparate roles in opioid reward, tolerance, and withdrawal in the brain. In this study we monitored dopamine release in the medial or lateral shell of the NAc of male mice during a week-long morphine treatment paradigm. We measured dopamine release in response to an intravenous morphine injection both acutely and following a week of repeated morphine. We also measured dopamine in response to a naloxone injection both prior to and following repeated morphine treatment. Morphine induced a transient increase in dopamine in the medial NAc shell that was much larger than the slower rise observed in the lateral shell. Surprisingly, chronic morphine treatment induced a sensitization of the medial dopamine response to morphine that opposed a diminished response observed in the saline-treated control group. This study expands on our current understanding of the medial NAc shell as hub of opioid-induced dopamine fluctuation. It also highlights the need for future opioid studies to appreciate the heterogeneity of dopamine neurons. Significance Statement: The social and economic consequences of the opioid epidemic are tragic and far-reaching. Yet, opioids are indisputably necessary in clinical settings where they remain the most useful treatment for severe pain. To combat this crisis, we must improve our understanding of opioid function in the brain, particularly the neural mechanisms that underlie opioid dependence and addictive behaviors. This study uses fiber photometry to examine dopamine changes that occur in response to repeated morphine, and morphine withdrawal, at multiple stages of a longitudinal opioid-dependence paradigm. We reveal key differences in how dopamine levels respond to opioid administration in distinct sub-regions of the ventral striatum and lay a foundation for future opioid research that appreciates our contemporary understanding of the dopamine system.

3.
bioRxiv ; 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38562752

RESUMO

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.

4.
bioRxiv ; 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38659831

RESUMO

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.

5.
Annu Rev Physiol ; 86: 1-25, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38029388

RESUMO

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.


Assuntos
Analgésicos Opioides , Transdução de Sinais , Humanos , Analgésicos Opioides/efeitos adversos , Tolerância a Medicamentos
6.
Biol Psychiatry ; 94(7): 531-542, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36931452

RESUMO

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.


Assuntos
Antipsicóticos , Dopamina , Camundongos , Animais , beta-Arrestina 2/metabolismo , Antipsicóticos/farmacologia , Receptores de Dopamina D3/metabolismo , Agonistas de Dopamina/farmacologia , Tolerância a Medicamentos , Receptores de Dopamina D1/metabolismo
7.
Neuropsychopharmacology ; 46(13): 2241-2249, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34257415

RESUMO

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.


Assuntos
Receptores Opioides , Insuficiência Respiratória , Analgésicos , Analgésicos Opioides/farmacologia , Animais , Tolerância a Medicamentos , Camundongos , Morfina/farmacologia , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/tratamento farmacológico , beta-Arrestina 2/metabolismo
8.
ACS Chem Neurosci ; 8(3): 473-485, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-27744679

RESUMO

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.


Assuntos
Analgésicos Opioides , Endorfinas/farmacologia , Naftiridinas , Receptores Opioides delta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Analgésicos Opioides/química , Analgésicos Opioides/isolamento & purificação , Analgésicos Opioides/farmacologia , Animais , Simulação por Computador , AMP Cíclico/metabolismo , Endocitose/efeitos dos fármacos , Endorfinas/química , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Indóis/química , Indóis/isolamento & purificação , Indóis/farmacologia , Locomoção/efeitos dos fármacos , Locomoção/genética , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Naftiridinas/química , Naftiridinas/isolamento & purificação , Naftiridinas/farmacologia , Poríferos/química , Receptores Opioides delta/genética , Transdução de Sinais/genética , Espectrometria de Massas por Ionização por Electrospray , Natação/psicologia , beta-Arrestinas/metabolismo
9.
Cell Rep ; 17(12): 3233-3245, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-28009292

RESUMO

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.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Reversão de Aprendizagem/fisiologia , Fator de Crescimento Transformador beta1/genética , Animais , Dendritos/genética , Dendritos/fisiologia , Neurônios Dopaminérgicos/fisiologia , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/fisiologia , Regulação da Expressão Gênica , Humanos , Mesencéfalo/crescimento & desenvolvimento , Mesencéfalo/metabolismo , Camundongos , Receptor do Fator de Crescimento Transformador beta Tipo II , Transdução de Sinais/genética , Sinapses/genética , Sinapses/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
10.
Cell Rep ; 16(6): 1518-1526, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27452469

RESUMO

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.


Assuntos
Segmento Inicial do Axônio/metabolismo , Canais de Cálcio/metabolismo , Dopamina/metabolismo , beta-Arrestinas/metabolismo , Animais , Cálcio/metabolismo , Células HEK293 , Humanos , Fosforilação , Receptores Acoplados a Proteínas G/metabolismo
11.
Cell Rep ; 14(12): 2774-83, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26997280

RESUMO

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.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Ansiedade , Dinorfinas/farmacologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Tonsila do Cerebelo/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Channelrhodopsins , Potenciais Evocados/efeitos dos fármacos , Ácido Glutâmico/farmacologia , Imidazóis/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Microscopia de Fluorescência , Técnicas de Patch-Clamp , Piridinas/farmacologia , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Núcleos Septais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Mol Cell Endocrinol ; 382(2): 938-49, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24275181

RESUMO

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.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/farmacologia , Ilhotas Pancreáticas/metabolismo , Receptores de Glucagon/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , AMP Cíclico/metabolismo , Exenatida , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Células HEK293 , Humanos , Incretinas/metabolismo , Incretinas/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/ultraestrutura , Liraglutida , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/metabolismo , Peptídeos/farmacologia , Estabilidade Proteica , Transporte Proteico , Proteólise , Imagem com Lapso de Tempo , Peçonhas/metabolismo , Peçonhas/farmacologia
13.
Cell ; 154(5): 1085-1099, 2013 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-23954414

RESUMO

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.


Assuntos
Autofagia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lisossomos/metabolismo , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Obesidade/metabolismo , Alinhamento de Sequência
14.
J Neurosci ; 33(30): 12329-36, 2013 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-23884939

RESUMO

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.


Assuntos
Cocaína/farmacologia , Receptores de AMPA/fisiologia , Receptores de Dopamina D2/metabolismo , Receptores de N-Metil-D-Aspartato/fisiologia , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia , Animais , Antipsicóticos/farmacologia , Aripiprazol , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Ácido Glutâmico/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Técnicas de Cultura de Órgãos , Piperazinas/farmacologia , Quinolonas/farmacologia , Receptores de Dopamina D2/agonistas , Potenciais Sinápticos/efeitos dos fármacos , Potenciais Sinápticos/fisiologia
15.
Psychopharmacology (Berl) ; 228(1): 1-18, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23649885

RESUMO

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.


Assuntos
Analgésicos Opioides/farmacologia , Desenho de Fármacos , Receptores Opioides delta/efeitos dos fármacos , Animais , Ensaios Clínicos como Assunto , Humanos , Ligantes , Terapia de Alvo Molecular , Receptores Opioides delta/metabolismo
16.
PLoS One ; 8(3): e58362, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23554887

RESUMO

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.


Assuntos
Dor/metabolismo , Multimerização Proteica , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Dor/tratamento farmacológico , Dor/genética , Dor/patologia , Manejo da Dor , Estabilidade Proteica/efeitos dos fármacos , Receptores Opioides delta/genética , Receptores Opioides mu/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
17.
Methods Mol Biol ; 995: 43-54, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23494371

RESUMO

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.


Assuntos
Compostos de Benzilideno/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , D-Penicilina (2,5)-Encefalina/farmacologia , Ensaios de Triagem em Larga Escala/métodos , Naltrexona/análogos & derivados , Receptores Opioides delta/agonistas , Sinalização do Cálcio , Técnicas de Cultura de Células , Células HEK293 , Humanos , Naltrexona/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/biossíntese , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/biossíntese , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/biossíntese , Espectrometria de Fluorescência , Transfecção
18.
J Pharmacol Exp Ther ; 344(1): 179-88, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23097213

RESUMO

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.


Assuntos
Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/efeitos dos fármacos , Sequência de Aminoácidos , Benzamidas/farmacologia , Cálcio/metabolismo , Sinalização do Cálcio , Clonagem Molecular , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Encefalina Leucina/farmacologia , Ensaio de Imunoadsorção Enzimática , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/efeitos dos fármacos , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/efeitos dos fármacos , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas Mutantes Quiméricas/química , Proteínas Mutantes Quiméricas/genética , Piperazinas/farmacologia , Reação em Cadeia da Polimerase , Receptores Acoplados a Proteínas G/genética , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides delta/genética , Proteínas Recombinantes de Fusão
19.
Neuropsychopharmacology ; 37(11): 2436-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22739468

RESUMO

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.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Ansiolíticos/uso terapêutico , Antidepressivos/uso terapêutico , Ansiedade/tratamento farmacológico , Depressão/tratamento farmacológico , Etanol/administração & dosagem , Neuropeptídeos/uso terapêutico , Adaptação Ocular/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Análise de Variância , Animais , Condicionamento Operante/efeitos dos fármacos , Modelos Animais de Doenças , Vias de Administração de Medicamentos , Sinergismo Farmacológico , Etanol/metabolismo , Elevação dos Membros Posteriores/métodos , Técnicas In Vitro , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Natação/psicologia , Fatores de Tempo
20.
J Am Chem Soc ; 134(25): 10321-4, 2012 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-22671299

RESUMO

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).


Assuntos
Modelos Moleculares , Compostos Organometálicos/química , Peptídeos/química , Receptores Acoplados a Proteínas G/química , Ródio/química , Tirosina/química , Ligação Competitiva , Neoplasias da Mama/tratamento farmacológico , Feminino , Células HT29 , Humanos , Concentração Inibidora 50 , Espectroscopia de Ressonância Magnética , Compostos Organometálicos/farmacologia , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Receptores Acoplados a Proteínas G/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA