Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Psychiatry ; 28(7): 2946-2963, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37131076

RESUMO

While persistence of fear memories is essential for survival, a failure to inhibit fear in response to harmless stimuli is a feature of anxiety disorders. Extinction training only temporarily suppresses fear memory recovery in adults, but it is highly effective in juvenile rodents. Maturation of GABAergic circuits, in particular of parvalbumin-positive (PV+) cells, restricts plasticity in the adult brain, thus reducing PV+ cell maturation could promote the suppression of fear memories following extinction training in adults. Epigenetic modifications such as histone acetylation control gene accessibility for transcription and help couple synaptic activity to changes in gene expression. Histone deacetylase 2 (Hdac2), in particular, restrains both structural and functional synaptic plasticity. However, whether and how Hdac2 controls the maturation of postnatal PV+ cells is not well understood. Here, we show that PV+- cell specific Hdac2 deletion limits spontaneous fear memory recovery in adult mice, while enhancing PV+ cell bouton remodeling and reducing perineuronal net aggregation around PV+ cells in prefrontal cortex and basolateral amygdala. Prefrontal cortex PV+ cells lacking Hdac2, show reduced expression of Acan, a critical perineuronal net component, which is rescued by Hdac2 re-expression. Pharmacological inhibition of Hdac2 before extinction training is sufficient to reduce both spontaneous fear memory recovery and Acan expression in wild-type adult mice, while these effects are occluded in PV+-cell specific Hdac2 conditional knockout mice. Finally, a brief knock-down of Acan expression mediated by intravenous siRNA delivery before extinction training but after fear memory acquisition is sufficient to reduce spontaneous fear recovery in wild-type mice. Altogether, these data suggest that controlled manipulation of PV+ cells by targeting Hdac2 activity, or the expression of its downstream effector Acan, promotes the long-term efficacy of extinction training in adults.


Assuntos
Condicionamento Psicológico , Parvalbuminas , Camundongos , Animais , Parvalbuminas/metabolismo , Regulação para Baixo , Condicionamento Psicológico/fisiologia , Memória/fisiologia , Medo/fisiologia , Camundongos Knockout , Extinção Psicológica/fisiologia
2.
J Cell Mol Med ; 24(10): 5718-5730, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32279433

RESUMO

δ-opioid receptor (DOPr) agonists have analgesic efficacy in chronic pain models but development of tolerance limits their use for long-term pain management. Although agonist potential for inducing acute analgesic tolerance has been associated with distinct patterns of DOPr internalization, the association between trafficking and chronic tolerance remains ill-defined. In a rat model of streptozotocin (STZ)-induced diabetic neuropathy, deltorphin II and TIPP produced sustained analgesia  following daily (intrathecal) i.t. injections over six days, whereas similar treatment with SNC-80 or SB235863 led to progressive tolerance and loss of the analgesic response. Trafficking assays in murine neuron cultures showed no association between the magnitude of ligand-induced sequestration and development of chronic tolerance. Instead, ligands that supported DOPr recycling were also the ones producing sustained analgesia over 6-day treatment. Moreover, endosomal endothelin-converting enzyme 2 (ECE2) blocker 663444 prevented DOPr recycling by deltorphin II and TIPP and precipitated tolerance by these ligands. In conclusion, agonists, which support DOPr recycling, avoid development of analgesic tolerance over repeated administration.


Assuntos
Analgésicos/farmacologia , Tolerância a Medicamentos , Receptores Opioides delta/metabolismo , Animais , Células Cultivadas , Dor Crônica/diagnóstico , Dor Crônica/tratamento farmacológico , Dor Crônica/etiologia , Dor Crônica/metabolismo , Nefropatias Diabéticas/complicações , Modelos Animais de Doenças , Ligantes , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Medição da Dor , Ligação Proteica , Ratos
3.
Cell Mol Life Sci ; 75(12): 2257-2271, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29288293

RESUMO

Soon after internalization delta opioid receptors (DOPrs) are committed to the degradation path by G protein-coupled receptor (GPCR)-associated binding protein. Here we provide evidence that this classical post-endocytic itinerary may be rectified by downstream sorting decisions which allow DOPrs to regain to the membrane after having reached late endosomes (LE). The LE sorting mechanism involved ESCRT accessory protein Alix and the TIP47/Rab9 retrieval complex which supported translocation of the receptor to the TGN, from where it subsequently regained the cell membrane. Preventing DOPrs from completing this itinerary precipitated acute analgesic tolerance to the agonist DPDPE, supporting the relevance of this recycling path in maintaining the analgesic response by this receptor. Taken together, these findings reveal a post-endocytic itinerary where GPCRs that have been sorted for degradation can still recycle to the membrane.


Assuntos
Membrana Celular/metabolismo , Receptores Opioides delta/metabolismo , Animais , Células Cultivadas , Células HEK293 , Humanos , Masculino , Camundongos , Neurônios/metabolismo , Transporte Proteico , Proteólise , Ratos , Ratos Sprague-Dawley
4.
Pharmacol Rev ; 68(3): 631-700, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27343248

RESUMO

Opioids are among the most effective analgesics available and are the first choice in the treatment of acute severe pain. However, partial efficacy, a tendency to produce tolerance, and a host of ill-tolerated side effects make clinically available opioids less effective in the management of chronic pain syndromes. Given that most therapeutic opioids produce their actions via µ-opioid receptors (MOPrs), other targets are constantly being explored, among which δ-opioid receptors (DOPrs) are being increasingly considered as promising alternatives. This review addresses DOPrs from the perspective of cellular and molecular determinants of their pharmacological diversity. Thus, DOPr ligands are examined in terms of structural and functional variety, DOPrs' capacity to engage a multiplicity of canonical and noncanonical G protein-dependent responses is surveyed, and evidence supporting ligand-specific signaling and regulation is analyzed. Pharmacological DOPr subtypes are examined in light of the ability of DOPr to organize into multimeric arrays and to adopt multiple active conformations as well as differences in ligand kinetics. Current knowledge on DOPr targeting to the membrane is examined as a means of understanding how these receptors are especially active in chronic pain management. Insight into cellular and molecular mechanisms of pharmacological diversity should guide the rational design of more effective, longer-lasting, and better-tolerated opioid analgesics for chronic pain management.


Assuntos
Analgésicos Opioides/farmacologia , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inibidores , Animais , Humanos , Ligantes , Modelos Moleculares , Estrutura Molecular , Terapia de Alvo Molecular/métodos , Medição da Dor , Receptores Opioides delta/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Methods ; 92: 78-86, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26364590

RESUMO

Signaling bias makes reference to the capacity of G-protein coupled receptor (GPCR) ligands to direct pharmacological stimuli to a subset of effectors among all of those controlled by the receptor. This new signaling modality has added texture to the classical notion of efficacy. In doing so, it has opened new avenues for the development of therapeutic GPCR ligands that specifically modulate signals underlying desired effects while sparing those that support undesired drug actions. Essential to taking advantage of this texture is the ability to identify, quantify and represent bias in a reliable and intuitive manner that ensures comparison among ligands. Here, we present a practical guide on how the operational model may be used to evaluate ligand efficiency to induce different responses, how differences in response may be used to estimate bias and how quantitative information derived from this analysis may be graphically represented to recreate a drug's unique signaling footprint. The approach used is discussed in terms of data interpretation and limitations that may influence the conclusions drawn from the analysis.


Assuntos
Biologia Computacional/métodos , Descoberta de Drogas/métodos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Relação Dose-Resposta a Droga , Humanos , Ligantes , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/metabolismo , Ligação Proteica/fisiologia , Transdução de Sinais/efeitos dos fármacos
6.
Methods ; 92: 19-35, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26210401

RESUMO

Ion channels play a vital role in numerous physiological functions and drugs that target them are actively pursued for development of novel therapeutic agents. Here we report a means for monitoring in real time the conformational changes undergone by channel proteins upon exposure to pharmacological stimuli. The approach relies on tracking structural rearrangements by monitoring changes in bioluminescence energy transfer (BRET). To provide proof of principle we have worked with Kir3 neuronal channels producing 10 different constructs which were combined into 17 donor-acceptor BRET pairs. Among these combinations, pairs bearing the donor Nano-Luc (NLuc) at the C-terminal end of Kir3.2 subunits and the FlAsH acceptor at the N-terminal end (NT) or the interfacial helix (N70) of Kir3.1 subunits were identified as potential tools. These pairs displayed significant changes in energy transfer upon activation with direct channel ligands or via stimulation of G protein-coupled receptors. Conformational changes associated with channel activation followed similar kinetics as channel currents. Dose response curves generated by different agonists in FlAsH-BRET assays displayed similar rank order of potency as those obtained with conventional BRET readouts of G protein activation and ion flux assays. Conformational biosensors as the ones reported herein should prove a valuable complement to other methodologies currently used in channel drug discovery.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Técnicas Biossensoriais/métodos , Desenho de Fármacos , Fluoresceína/síntese química , Fluoresceína/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Analgésicos Opioides/síntese química , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/química , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Camundongos , Conformação Proteica
7.
Cell Mol Life Sci ; 72(18): 3543-57, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25900661

RESUMO

Kir3 channels control excitability in the nervous system and the heart. Their surface expression is strictly regulated, but mechanisms responsible for channel removal from the membrane remain incompletely understood. Using transfected cells, we show that Kir3.1/3.2 channels and delta opioid receptors (DORs) associate in a complex which persists during receptor activation, behaving as a scaffold that allows beta-arrestin (ßarr) to interact with both signaling partners. This organization favored co-internalization of DORs and Kir3 channels in a ßarr-dependent manner via a clathrin/dynamin-mediated endocytic path. Taken together, these findings identify a new way of modulating Kir3 channel availability at the membrane and assign a putatively novel role for ßarrs in regulating canonical effectors for G protein-coupled receptors.


Assuntos
Arrestinas/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Receptores Opioides delta/metabolismo , Animais , Membrana Celular/metabolismo , Células Cultivadas , Clatrina/metabolismo , Dinaminas/metabolismo , Endocitose/fisiologia , Células HEK293 , Humanos , Ratos , Transdução de Sinais/fisiologia , beta-Arrestinas
8.
Cell Mol Life Sci ; 71(8): 1529-46, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24022593

RESUMO

Signaling bias refers to G protein-coupled receptor ligand ability to preferentially activate one type of signal over another. Bias to evoke signaling as opposed to sequestration has been proposed as a predictor of opioid ligand potential for generating tolerance. Here we measured whether delta opioid receptor agonists preferentially inhibited cyclase activity over internalization in HEK cells. Efficacy (τ) and affinity (KA) values were estimated from functional data and bias was calculated from efficiency coefficients (log τ/KA). This approach better represented the data as compared to alternative methods that estimate bias exclusively from τ values. Log (τ/KA) coefficients indicated that SNC-80 and UFP-512 promoted cyclase inhibition more efficiently than DOR internalization as compared to DPDPE (bias factor for SNC-80: 50 and for UFP-512: 132). Molecular determinants of internalization were different in HEK293 cells and neurons with ßarrs contributing to internalization in both cell types, while PKC and GRK2 activities were only involved in neurons. Rank orders of ligand ability to engage different internalization mechanisms in neurons were compared to rank order of E max values for cyclase assays in HEK cells. Comparison revealed a significant reversal in rank order for cyclase E max values and ßarr-dependent internalization in neurons, indicating that these responses were ligand-specific. Despite this evidence, and because kinases involved in internalization were not the same across cellular backgrounds, it is not possible to assert if the magnitude and nature of bias revealed by rank orders of maximal responses is the same as the one measured in HEK cells.


Assuntos
Dor Crônica/tratamento farmacológico , AMP Cíclico/metabolismo , Tolerância a Medicamentos/fisiologia , Neurônios/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Análise de Variância , Benzamidas/farmacologia , Benzimidazóis/farmacologia , Western Blotting , Relação Dose-Resposta a Droga , D-Penicilina (2,5)-Encefalina , Imunofluorescência , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Proteínas de Fluorescência Verde , Células HEK293 , Humanos , Ligantes , Oligopeptídeos/farmacologia , Piperazinas/farmacologia
9.
Mol Pharmacol ; 86(2): 138-49, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24874699

RESUMO

In this study, we used a combination of traditional signaling investigation approaches, bioluminescence resonance energy transfer (BRET) biosensors, and the label-free approach surface plasmon resonance (SPR) spectroscopy to monitor the signaling cascades of the µ-opioid receptor (MOP). In human embryonic kidney cells stably expressing a Flag-tagged version of human MOP, we compared the signals triggered by the noninternalizing and internalizing MOP agonists morphine and DAMGO (Tyr-D-Ala-Gly-N-methyl-Phe-Gly-ol), respectively. We studied three major and well described components of MOP signaling: receptor internalization, G protein coupling, and activation of extracellular signal-regulated kinase ERK1/ERK2. Our results show that morphine and DAMGO display different profiles of receptor internalization and a similar ability to trigger the phosphorylation of ERK1/ERK2. Our SPR analyses revealed that morphine and DAMGO evoke similar SPR signatures and that Gαi, cAMP-dependent pathways, and ERK1/ERK2 have key roles in morphine- and DAMGO-mediated signaling. Most interestingly, we found that the so-called MOP neutral antagonists CTOP (D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2)), naloxone, and naltrexone behave like partial agonists. Even more intriguing, BRET experiments indicate that CTAP (D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH(2)) induces similar conformational changes as naltrexone at the Gαi-ßγ interface, whereas it appears as an inverse agonist based on its SPR response thus indicating distinct signaling mechanisms for the two ligands. Taken together, our results support the usefulness of label-free methods such as SPR to study whole-cell responses and signaling cascades triggered by G protein-coupled receptors and complement the conventional approaches by revealing cellular responses that would have been otherwise undetectable.


Assuntos
Receptores Opioides mu/metabolismo , Transdução de Sinais/fisiologia , Linhagem Celular , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/genética , Fosforilação/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores
10.
Mol Pharmacol ; 85(1): 148-61, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24174495

RESUMO

Traditional assays that monitor cAMP inhibition by opioid receptor ligands require second-messenger accumulation over periods of 10-20 minutes. Since receptor regulation occurs within a similar time frame, such assays do not discriminate the actual signal from its modulation. Here we used bioluminescence resonance energy transfer to monitor inhibition of cAMP production by δ-opioid receptor (DOR) agonists in real time. cAMP inhibition elicited by different agonists over a period of 15 minutes was biphasic, with response buildup during the first 6 to 7 minutes, followed by a second phase of response decay or of no further increment. The rate at which the cAMP response disappeared was correlated with operational parameters describing ligand efficiency [log(τ/KA)] to promote Gαi activation, as well as with ligand ability to promote internalization during the time course of the assay. Thus, ligands that displayed low signaling efficiency and poor sequestration(SB235863 ([8R-(4bS*,8aα,8aß,12bß)]7,10-dimethyl-1-methoxy-11-(2-ethylpropyl)oxycarbonyl 5,6,7,8,12,12b-hexahydro-(9H)-4,8-methanobenzofuro[3,2-e]pyrrolo[2,3-g]isoquinoline hydrochloride), morphine) had minimal or no response decay. On the other hand, the decay rate was pronounced for deltorphin II, [d-Pen(2), d-pen(5)]-enkephalin, met-enkephalin, and SNC-80 ((+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide), which displayed high signaling efficiency and internalization. Moreover, inhibition of internalization by dynasore reduced or abolished response decay by internalizing ligands. Unlike acute responses, endocytic profiles were not predictive of whether an agonist would induce prolonged cAMP inhibition over sustained (30-120 minutes) DOR stimulation. Taken together, the data indicate that ligand ability to evoke G-protein activation or promote endocytosis was predictive of response duration over short, but not over sustained periods of cAMP inhibition.


Assuntos
AMP Cíclico/biossíntese , Endocitose , Receptores Opioides delta/metabolismo , Técnicas de Transferência de Energia por Ressonância de Bioluminescência , AMP Cíclico/antagonistas & inibidores , Endocitose/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Ligantes , Receptores Opioides delta/agonistas , Fatores de Tempo
11.
J Neurosci ; 32(14): 4827-40, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22492038

RESUMO

Opioid tendency to generate analgesic tolerance has been previously linked to biased internalization. Here, we assessed an alternative possibility; whether tolerance of delta opioid receptor agonists (DORs) could be related to agonist-specific recycling. A first series of experiments revealed that DOR internalization by DPDPE and SNC-80 was similar, but only DPDPE induced recycling. We then established that the non-recycling agonist SNC-80 generated acute analgesic tolerance that was absent in mice treated with DPDPE. Furthermore, both agonists stabilized different conformations, whose distinct interaction with Gßγ subunits led to different modalities of ß-arrestin2 (ßarr2) recruitment. In particular, bioluminescence resonance energy transfer (BRET) assays revealed that sustained activation by SNC-80 drew the receptor C terminus in close proximity of the N-terminal domain of Gγ2, causing ßarr2 to interact with receptors and Gßγ subunits. DPDPE moved the receptor C-tail away from the Gßγ dimer, resulting in ßarr2 recruitment to the receptor but not in the vicinity of Gγ2. These differences were associated with stable DOR-ßarr2 association, poor recycling, and marked desensitization following exposure to SNC-80, while DPDPE promoted transient receptor interaction with ßarr2 and effective recycling, which conferred protection from desensitization. Together, these data indicate that DORs may adopt ligand-specific conformations whose distinct recycling properties determine the extent of desensitization and are predictive of analgesic tolerance. Based on these findings, we propose that the development of functionally selective DOR ligands that favor recycling could constitute a valid strategy for the production of longer acting opioid analgesics.


Assuntos
Analgésicos Opioides/metabolismo , Arrestinas/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Analgésicos Opioides/farmacologia , Animais , Animais Recém-Nascidos , Arrestinas/fisiologia , Linhagem Celular Transformada , Células Cultivadas , Tolerância a Medicamentos/fisiologia , Subunidades beta da Proteína de Ligação ao GTP/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/fisiologia , Células HEK293 , Humanos , Masculino , Camundongos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , beta-Arrestinas
12.
Mol Pharmacol ; 83(2): 416-28, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23175530

RESUMO

This study assessed how conformational information encoded by ligand binding to δ-opioid receptors (DORs) is transmitted to Kir3.1/Kir3.2 channels. Human embryonic kidney 293 cells were transfected with bioluminescence resonance energy transfer (BRET) donor/acceptor pairs that allowed us to evaluate independently reciprocal interactions among signaling partners. These and coimmunoprecipitation studies indicated that DORs, Gßγ, and Kir3 subunits constitutively interacted with one another. GαoA associated with DORs and Gßγ, but despite being part of the complex, no evidence of its direct association with the channel was obtained. DOR activation by different ligands left DOR-Kir3 interactions unmodified but modulated BRET between DOR-GαoA, DOR-Gßγ, GαoA-Gßγ, and Gßγ-Kir3 interfaces. Ligand-induced BRET changes assessing Gßγ-Kir3.1 subunit interaction 1) followed similar kinetics to those monitoring the GαoA-Gßγ interface, 2) displayed the same order of efficacy as those observed at the DOR-Gßγ interface, 3) were sensitive to pertussis toxin, and 4) were predictive of whether a ligand could evoke channel currents. Conformational changes at the Gßγ/Kir3 interface were lost when Kir3.1 subunits were replaced by a mutant lacking essential sites for Gßγ-mediated activation. Thus, conformational information encoded by agonist binding to the receptor is relayed to the channel via structural rearrangements that involve repositioning of Gßγ with respect to DORs, GαoA, and channel subunits. Further, the fact that BRET changes at the Gßγ-Kir3 interface are predictive of a ligand's ability to induce channel currents points to these conformational biosensors as screening tools for identifying GPCR ligands that induce Kir3 channel activation.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Receptores Opioides delta/metabolismo , Sítios de Ligação , Linhagem Celular , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/química , Células HEK293 , Humanos , Cinética , Ligantes , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas , Receptores Opioides delta/química
13.
Front Pharmacol ; 14: 1235255, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37745077

RESUMO

Introduction: Cannabis extracts are being increasingly used to mitigate chronic pain. Current guidelines for their prescription rely on Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) content as well as the ratio of these major cannabinoids present in the blend. Here we assessed whether these descriptors were representative of product effectiveness to produce a desired outcome such as analgesia. Methods: In this study, we used a rat model of diabetic neuropathy and assessed the reduction in mechanical allodynia following intrathecal injection of pure THC, pure CBD, a 1:1 mix of these compounds and a "balanced" chemotype II cannabis extract. Engagement of endocannabinoid targets by different treatments was investigated using CB1 (AM251) and CB2 (AM630) receptor antagonists as well as a TRPV1 channel blocker (capsazepine). Results: Antinociceptive responses induced by an equivalent amount of THC administered in its pure form, as a THC:CBD mix or as a "balanced" extract were distinct. Furthermore, the 1:1 THC:CBD mix and the balanced extract had not only different response profiles but their relative engagement of CB1, CB2 receptors and TRPV1 channels was distinct. Discussion: These findings indicate that antinociceptive responses and targets engaged by blended cannabinoids are composition-specific, and cannot be simply inferred from THC and CBD contents. This information may have implications in relation to the way medicinal cannabis products are prescribed.

14.
J Biol Chem ; 286(11): 9174-84, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21233215

RESUMO

The peptide neurotensin (NT) is known to exert a potent excitatory effect on the dopaminergic system by inhibiting D2 dopamine (DA) receptor (D2R) function. This regulation is dependent on activation of PKC, a well known effector of the type 1 NT receptor (NTR1). Because PKC phosphorylation of the D2R has recently been shown to induce its internalization, we hypothesized that NT acts to reduce D2R function through heterologous desensitization of the D2R. In the present study, we first used HEK-293 cells to demonstrate that NT induces PKC-dependent D2R internalization. Furthermore, internalization displayed faster kinetics in cells expressing the D2R short isoform, known to act as an autoreceptor in DA neurons, than in cells expressing the long isoform, known to act as a postsynaptic D2R. In patch clamp experiments on cultured DA neurons, overexpression of a mutant D2S lacking three key PKC phosphorylation sites abrogated the ability of NT to reduce D2R-mediated cell firing inhibition. Short interfering RNA-mediated inhibition of ß-arrestin1 and dynamin2, proteins important for receptor desensitization, reduced agonist-induced desensitization of D2R function, but only the inhibition of ß-arrestin1 reduced the effect of NT on D2R function. Taken together, our data suggest that NT acutely regulates D2 autoreceptor function and DA neuron excitability through PKC-mediated phosphorylation of the D2R, leading to heterologous receptor desensitization.


Assuntos
Arrestinas/metabolismo , Neurônios/metabolismo , Neurotensina/farmacologia , Proteína Quinase C/metabolismo , Receptores de Dopamina D2/metabolismo , Animais , Arrestinas/genética , Dinamina II/genética , Dinamina II/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Células HEK293 , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Transgênicos , Neurotensina/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteína Quinase C/genética , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Receptores de Dopamina D2/genética , Membranas Sinápticas/genética , Membranas Sinápticas/metabolismo , beta-Arrestinas
15.
J Cereb Blood Flow Metab ; 42(7): 1294-1308, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35107038

RESUMO

Perinatal hypoxic/ischemic (HI) brain injury is a major clinical problem with devastating neurodevelopmental outcomes in neonates. During HI brain injury, dysregulated factor production contributes to microvascular impairment. Glycolysis-derived lactate accumulated during ischemia has been proposed to protect against ischemic injury, but its mechanism of action is poorly understood. Herein, we hypothesize that lactate via its G-protein coupled receptor (GPR81) controls postnatal brain angiogenesis and plays a protective role after HI injury. We show that GPR81 is predominantly expressed in neurons of the cerebral cortex and hippocampus. GPR81-null mice displayed a delay in cerebral microvascular development linked to reduced levels of various major angiogenic factors and augmented expression of anti-angiogenic Thrombospondin-1 (TSP-1) in comparison to their WT littermates. Coherently, lactate stimulation induced an increase in growth factors (VEGF, Ang1 and 2, PDGF) and reduced TSP-1 expression in neurons, which contributed to accelerating angiogenesis. HI injury in GPR81-null animals curtailed vascular density and consequently increased infarct size compared to changes seen in WT mice; conversely intracerebroventricular lactate injection increased vascular density and diminished infarct size in WT but not in GPR81-null mice. Collectively, we show that lactate acting via GPR81 participates in developmental brain angiogenesis, and attenuates HI injury by restoring compromised microvasculature.


Assuntos
Lesões Encefálicas , Hipóxia-Isquemia Encefálica , Neovascularização Fisiológica , Receptores Acoplados a Proteínas G , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Lesões Encefálicas/metabolismo , Feminino , Hipóxia-Isquemia Encefálica/metabolismo , Infarto , Isquemia/metabolismo , Ácido Láctico/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Gravidez , Receptores Acoplados a Proteínas G/genética , Trombospondina 1/metabolismo
16.
Neuron ; 55(5): 712-25, 2007 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-17785179

RESUMO

Current antidepressants are clinically effective only after several weeks of administration. Here, we show that serotonin(4) (5-HT(4)) agonists reduce immobility in the forced swimming test, displaying an antidepressant potential. Moreover, a 3 day regimen with such compounds modifies rat brain parameters considered to be key markers of antidepressant action, but that are observed only after 2-3 week treatments with classical molecules: desensitization of 5-HT(1A) autoreceptors, increased tonus on hippocampal postsynaptic 5-HT(1A) receptors, and enhanced phosphorylation of the CREB protein and neurogenesis in the hippocampus. In contrast, a 3 day treatment with the SSRI citalopram remains devoid of any effect on these parameters. Finally, a 3 day regimen with the 5-HT(4) agonist RS 67333 was sufficient to reduce both the hyperlocomotion induced by olfactory bulbectomy and the diminution of sucrose intake consecutive to a chronic mild stress. These findings point out 5-HT(4) receptor agonists as a putative class of antidepressants with a rapid onset of action.


Assuntos
Antidepressivos/farmacologia , Encéfalo/efeitos dos fármacos , Transtorno Depressivo/tratamento farmacológico , Agonistas do Receptor 5-HT4 de Serotonina , Agonistas do Receptor de Serotonina/farmacologia , Serotonina/metabolismo , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Animais , Antidepressivos/uso terapêutico , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Transtorno Depressivo/metabolismo , Transtorno Depressivo/fisiopatologia , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Masculino , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Fosforilação/efeitos dos fármacos , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Receptor 5-HT1A de Serotonina/metabolismo , Receptores 5-HT4 de Serotonina/metabolismo , Agonistas do Receptor 5-HT1 de Serotonina , Agonistas do Receptor de Serotonina/uso terapêutico , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Fatores de Tempo
17.
Cell Signal ; 80: 109906, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33383156

RESUMO

Opioid analgesics are elective for treating moderate to severe pain but their use is restricted by severe side effects. Signaling bias has been proposed as a viable means for improving this situation. To exploit this opportunity, continuous efforts are devoted to understand how ligand-specific modulations of receptor functions could mediate the different in vivo effects of opioids. Advances in the field have led to the development of biased agonists based on hypotheses that allocated desired and undesired effects to specific signaling pathways. However, the prevalent hypothesis associating ß-arrestin to opioid side effects was recently challenged and multiple of the newly developed biased drugs may not display the superior side effects profile that was sought. Moreover, biased agonism at opioid receptors is now known to be time- and cell-dependent, which adds a new layer of complexity for bias estimation. Here, we first review the signaling mechanisms underlying desired and undesired effects of opioids. We then describe biased agonism at opioid receptors and discuss the different perspectives that support the desired and undesired effects of opioids in view of exploiting biased signaling for therapeutic purposes. Finally, we explore how signaling kinetics and cellular background can influence the magnitude and directionality of bias at those receptors.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Transdução de Sinais , beta-Arrestinas/metabolismo , Analgésicos Opioides/farmacologia , Proteínas de Ligação ao GTP/agonistas , Humanos , Cinética , Ligantes , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Transdução de Sinais/efeitos dos fármacos , beta-Arrestinas/agonistas
18.
ACS Pharmacol Transl Sci ; 4(5): 1483-1498, 2021 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-34661070

RESUMO

Prolonged exposure to opioid receptor agonists triggers adaptations in the adenylyl cyclase (AC) pathway that lead to enhanced production of cyclic adenosine monophosphate (cAMP) upon withdrawal. This cellular phenomenon contributes to withdrawal symptoms, hyperalgesia and analgesic tolerance that interfere with clinical management of chronic pain syndromes. Since δ-opioid receptors (DOPrs) are a promising target for chronic pain management, we were interested in finding out if cell-based signaling profiles as generated for drug discovery purposes could inform us of the ligand potential to induce sensitization of the cyclase path. For this purpose, signaling of DOPr agonists was monitored at multiple effectors. The resulting signaling profiles revealed marked functional selectivity, particularly for Met-enkephalin (Met-ENK) whose signaling bias profile differed from those of synthetic ligands like SNC-80 and ARM390. Signaling diversity among ligands was systematized by clustering agonists according to similarities in E max and Log(τ) values for the different responses. The classification process revealed that the similarity in Gα/Gßγ, but not in ß-arrestin (ßarr), responses was correlated with the potential of Met-ENK, deltorphin II, (d-penicillamine2,5)-enkephalin (DPDPE), ARM390, and SNC-80 to enhance cAMP production, all of which required Ca2+ mobilization to produce this response. Moreover, superactivation by Met-ENK, which was the most-effective Ca2+ mobilizing agonist, required Gαi/o activation, availability of Gßγ subunits at the membrane, and activation of Ca2+ effectors such as calmodulin and protein kinase C (PKC). In contrast, superactivation by (N-(l-tyrosyl)-(3S)-1,2,3,4-tetrahydroisoquinoline-3-carbonyl)-l-phenylalanyl-l-phenylalanine (TIPP), which was set in a distinct category through clustering, required activation of Gαi/o subunits but was independent of the Gßγ dimer and Ca2+ mobilization, relying instead on Src and Raf-1 to induce this cellular adaptation.

20.
Sci Rep ; 10(1): 8779, 2020 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-32471984

RESUMO

A comprehensive understanding of signalling downstream of GPCRs requires a broad approach to capture novel signalling modalities in addition to established pathways. Here, using an array of sixteen validated BRET-based biosensors, we analyzed the ability of seven different ß-adrenergic ligands to engage five distinct signalling pathways downstream of the ß1-adrenergic receptor (ß1AR). In addition to generating signalling signatures and capturing functional selectivity for the different ligands toward these pathways, we also revealed coupling to signalling pathways that have not previously been ascribed to the ßAR. These include coupling to Gz and G12 pathways. The signalling cascade linking the ß1AR to calcium mobilization was also characterized using a combination of BRET-based biosensors and CRISPR-engineered HEK 293 cells lacking the Gαs subunit or with pharmacological or genetically engineered pathway inhibitors. We show that both Gs and G12 are required for the full calcium response. Our work highlights the power of combining signal profiling with genome editing approaches to capture the full complement of GPCR signalling activities in a given cell type and to probe their underlying mechanisms.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Técnicas Biossensoriais/métodos , Sistemas CRISPR-Cas , Cálcio/metabolismo , Edição de Genes , Células HEK293 , Humanos , Ligantes , Fenótipo , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA