Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Pharmacol ; 185: 114440, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33539816

RESUMO

Antipsychotic drugs remain the current standard for schizophrenia treatment. Although they directly recognize the orthosteric binding site of numerous monoaminergic G protein-coupled receptors (GPCRs), these drugs, and particularly second-generation antipsychotics such as clozapine, all have in common a very high affinity for the serotonin 5-HT2A receptor (5-HT2AR). Using classical pharmacology and targeted signaling pathway assays, previous findings suggest that clozapine and other atypical antipsychotics behave principally as 5-HT2AR neutral antagonists and/or inverse agonists. However, more recent findings showed that antipsychotics may also behave as pathway-specific agonists. Reversible phosphorylation is a common element in multiple signaling networks. Combining a quantitative phosphoproteomic method with signaling network analysis, we tested the effect of clozapine treatment on the overall level of protein phosphorylation and signal transduction cascades in vitro in mammalian cell lines induced to express either the human 5-HT2AR or the H452Y variant of the gene encoding the 5-HT2AR receptor. This naturally occurring variation within the 5-HT2AR gene was selected because it has been repeatedly associated with schizophrenia patients who do not respond to clozapine treatment. Our data show that short time exposure (5 or 10 min) to clozapine (10-5 M) led to phosphorylation of numerous signaling components of pathways involved in processes such as endocytosis, ErbB signaling, insulin signaling or estrogen signaling. Cells induced to express the H452Y variant showed a different basal phosphoproteome, with increases in the phosphorylation of mTOR signaling components as a translationally relevant example. However, the effect of clozapine on the functional landscape of the phosphoproteome was significantly reduced in cells expressing the 5-HT2AR-H452Y construct. Together, these findings suggest that clozapine behaves as an agonist inducing phosphorylation of numerous pathways downstream of the 5-HT2AR, and that the single nucleotide polymorphism encoding 5-HT2AR-H452Y affects these clozapine-induced phosphorylation-dependent signaling networks.


Assuntos
Clozapina/metabolismo , Histamina/genética , Polimorfismo de Nucleotídeo Único/genética , Proteômica/métodos , Receptor 5-HT2A de Serotonina/genética , Tirosina/genética , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Clozapina/farmacologia , Relação Dose-Resposta a Droga , Células HEK293 , Histamina/metabolismo , Humanos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Receptor 5-HT2A de Serotonina/metabolismo , Antagonistas da Serotonina/metabolismo , Antagonistas da Serotonina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tirosina/metabolismo
2.
Sci Signal ; 13(654)2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33082287

RESUMO

Membrane trafficking processes regulate G protein-coupled receptor (GPCR) activity. Although class A GPCRs are capable of activating G proteins in a monomeric form, they can also potentially assemble into functional GPCR heteromers. Here, we showed that the class A serotonin 5-HT2A receptors (5-HT2ARs) affected the localization and trafficking of class C metabotropic glutamate receptor 2 (mGluR2) through a mechanism that required their assembly as heteromers in mammalian cells. In the absence of agonists, 5-HT2AR was primarily localized within intracellular compartments, and coexpression of 5-HT2AR with mGluR2 increased the intracellular distribution of the otherwise plasma membrane-localized mGluR2. Agonists for either 5-HT2AR or mGluR2 differentially affected trafficking through Rab5-positive endosomes in cells expressing each component of the 5-HT2AR-mGluR2 heterocomplex alone, or together. In addition, overnight pharmacological 5-HT2AR blockade with clozapine, but not with M100907, decreased mGluR2 density through a mechanism that involved heteromerization between 5-HT2AR and mGluR2. Using TAT-tagged peptides and chimeric constructs that are unable to form the interclass 5-HT2AR-mGluR2 complex, we demonstrated that heteromerization was necessary for the 5-HT2AR-dependent effects on mGluR2 subcellular distribution. The expression of 5-HT2AR also augmented intracellular localization of mGluR2 in mouse frontal cortex pyramidal neurons. Together, our data suggest that GPCR heteromerization may itself represent a mechanism of receptor trafficking and sorting.


Assuntos
Membrana Celular/metabolismo , Receptor 5-HT2A de Serotonina/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais , Aminoácidos/farmacologia , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Clozapina/farmacologia , Endossomos/metabolismo , Células HEK293 , Humanos , Camundongos da Linhagem 129 , Camundongos Knockout , Microscopia Confocal , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Multimerização Proteica , Transporte Proteico/efeitos dos fármacos , Receptor 5-HT2A de Serotonina/química , Receptor 5-HT2A de Serotonina/genética , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/genética , Antagonistas da Serotonina/farmacologia
3.
Exp Eye Res ; 188: 107790, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31494107

RESUMO

Oxidative stress (OxS) is involved in the development of cell injures occurring in retinal diseases while Poly(ADP-ribose) Polymerase-1 (PARP-1) is a key protein involved in the repair of the DNA damage caused by OxS. Inhibition of PARP-1 activity with the pharmacological inhibitor PJ34 in mouse retinal explants subjected to H2O2-induced oxidative damage resulted in an increase of apoptotic cells. Reduction of cell growth was also observed in the mouse cone like cell line 661 W in the presence of PJ34 under OxS conditions. Mass spectrometry-based phosphoproteomics analysis performed in 661 W cells determined that OxS induced significant changes in the phosphorylation in 1807 of the 8131 peptides initially detected. Blockade of PARP-1 activity after the oxidative treatment additionally increased the phosphorylation of multiple proteins, many of them at SQ motifs and related to the DNA-damage response (DDR). These motifs are substrates of the kinases ATM/ATR, which play a central role in DDR. Western blot analysis confirmed that the ATM/ATR activity measured and the phosphorylation at SQ motifs of ATM/ATR substrates was augmented when PARP-1 activity was inhibited under OxS conditions, in 661 W cells. Phosphorylation of ATM/ATR substrates, including the phosphorylation of the histone H2AX were also induced in organotypic cultures of retinal explants subjected to PARP-1 inhibition during exposure to OxS. In conclusion, inhibition of PARP-1 increased the phosphorylation and hence the activation of several proteins involved in the response to DNA damage, like the ATM protein kinase. This finally resulted in an augmented injury in mouse retinal cells suffering from OxS. Therefore, the inhibition of PARP-1 activity may have a negative outcome in the treatment of retinal diseases in which OxS is involved.


Assuntos
Dano ao DNA , Proteínas do Olho/metabolismo , Estresse Oxidativo , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Retina/patologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Western Blotting , Caspase 3/metabolismo , Morte Celular , Linhagem Celular , Proteínas de Ligação a DNA , Eletroforese em Gel de Poliacrilamida , Histonas/metabolismo , Peróxido de Hidrogênio/toxicidade , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Oxidantes/toxicidade , Fenantrenos/farmacologia , Fosfoproteínas/metabolismo , Fosforilação , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Retina/metabolismo
4.
PLoS One ; 14(2): e0211330, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30759168

RESUMO

Current drug discovery procedures require fast and effective quantification of the pharmacological response evoked in living cells by agonist compounds. In the case of G-protein coupled receptors (GPCRs), the efficacy of a particular drug to initiate the endocytosis process is related to the formation of endocytic vesicles or endosomes and their subsequent internalisation within intracellular compartments that can be observed with high spatial and temporal resolution by fluorescence microscopy techniques. Recently, an algorithm has been proposed to evaluate the pharmacological response by estimating the number of endosomes per cell on time series of images. However, the algorithm was limited by the dependence on some manually set parameters and in some cases the quality of the image does not allow a reliable detection of the endosomes. Here we propose a simple, fast and automated image analysis method-the Δm algorithm- to quantify a pharmacological response with data obtained from fluorescence microscopy experiments. This algorithm does not require individual object detection and computes the relative increment of the third order moment in fluorescence microscopy images after filtering with the Laplacian of Gaussian function. It was tested on simulations demonstrating its ability to discriminate different experimental situations according to the number and the fluorescence signal intensity of the simulated endosomes. Finally and in order to validate this methodology with real data, the algorithm was applied to several time-course experiments based on the endocytosis of the mu opioid receptor (MOP) initiated by different agonist compounds. Each drug displayed a different Δm sigmoid time-response curve and statistically significant differences were observed among drugs in terms of efficacy and kinetic parameters.


Assuntos
Endossomos/metabolismo , Interpretação de Imagem Assistida por Computador/métodos , Receptores Acoplados a Proteínas G/agonistas , Algoritmos , Simulação por Computador , Descoberta de Drogas , Endossomos/efeitos dos fármacos , Humanos , Microscopia de Fluorescência , Receptores Opioides mu/agonistas , Vesículas Transportadoras
5.
Curr Top Behav Neurosci ; 36: 45-73, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28677096

RESUMO

The neuropsychological effects of naturally occurring psychoactive chemicals have been recognized for millennia. Hallucinogens, which include naturally occurring chemicals such as mescaline and psilocybin, as well as synthetic compounds, such as lysergic acid diethylamide (LSD), induce profound alterations of human consciousness, emotion, and cognition. The discovery of the hallucinogenic effects of LSD and the observations that LSD and the endogenous ligand serotonin share chemical and pharmacological profiles led to the suggestion that biogenic amines like serotonin were involved in the psychosis of mental disorders such as schizophrenia. Although they bind other G protein-coupled receptor (GPCR) subtypes, studies indicate that several effects of hallucinogens involve agonist activity at the serotonin 5-HT2A receptor. In this chapter, we review recent advances in understanding hallucinogen drug action through characterization of structure, neuroanatomical location, and function of the 5-HT2A receptor.


Assuntos
Alucinógenos/farmacologia , Receptor 5-HT2A de Serotonina/efeitos dos fármacos , Agonistas do Receptor 5-HT2 de Serotonina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Humanos
6.
Pharmacol Biochem Behav ; 163: 83-89, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29024681

RESUMO

Morphine and related opioids are the mainstay of analgesic treatment, especially in patients suffering chronic pain. Besides their antinociceptive effects they may also exhibit anxiolytic-like properties that could contribute to pain relief. The pharmacological manipulation of the serotonergic system may not only modulate pain transmission and processing but also other behavioral effects of opioids. The present study aimed to analyze the effect of the concurrent treatment with citalopram, a selective serotonin reuptake inhibitor, on the antinociceptive, locomotor and anxiety-related effects induced by acute and subchronic administration of morphine in mice. Citalopram (15mg/kg) enhanced the acute antinociceptive effects of morphine when concurrently administered as evidenced by a two-fold increase in the ED50 for the antinociceptive effect of morphine in the hot-plate test. Chronic studies also revealed that concurrent citalopram treatment (15mg/kg) delayed the development of tolerance to the thermal antinociceptive effects of morphine. Additionally, morphine-induced hyperlocomotion was potentiated by citalopram as assessed in the open-field test and in the spontaneous activity recording in the home cage, a behavioral outcome to which tolerance or desensitization was not developed. Interestingly, chronic administration of both drugs promoted an anxiolytic effect as evidenced by the increased central activity in the open field test. Future investigations on this pharmacological interaction, such as the possible translational research in clinics, might have consequences in future strategies for the therapeutic management of pain.


Assuntos
Analgésicos Opioides/farmacologia , Ansiedade/induzido quimicamente , Citalopram/farmacologia , Locomoção/efeitos dos fármacos , Morfina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Analgésicos Opioides/efeitos adversos , Animais , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/efeitos adversos
7.
Nat Neurosci ; 20(9): 1247-1259, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28783139

RESUMO

Antipsychotic drugs remain the standard for schizophrenia treatment. Despite their effectiveness in treating hallucinations and delusions, prolonged exposure to antipsychotic medications leads to cognitive deficits in both schizophrenia patients and animal models. The molecular mechanisms underlying these negative effects on cognition remain to be elucidated. Here we demonstrate that chronic antipsychotic drug exposure increases nuclear translocation of NF-κB in both mouse and human frontal cortex, a trafficking event triggered via 5-HT2A-receptor-dependent downregulation of the NF-κB repressor IκBα. This upregulation of NF-κB activity led to its increased binding at the Hdac2 promoter, thereby augmenting Hdac2 transcription. Deletion of HDAC2 in forebrain pyramidal neurons prevented the negative effects of antipsychotic treatment on synaptic remodeling and cognition. Conversely, virally mediated activation of NF-κB signaling decreased cortical synaptic plasticity via HDAC2. Together, these observations may aid in developing therapeutic strategies to improve the outcome of schizophrenia treatment.


Assuntos
Antipsicóticos/efeitos adversos , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/metabolismo , Histona Desacetilase 2/metabolismo , NF-kappa B/metabolismo , Sinapses/metabolismo , Animais , Antipsicóticos/toxicidade , Transtornos Cognitivos/genética , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Células HEK293 , Histona Desacetilase 2/deficiência , Histona Desacetilase 2/genética , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/genética , Sinapses/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia
8.
Cell Signal ; 35: 208-222, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28411124

RESUMO

Morphine inefficiency to induce the internalization of mu opioid (MOP) receptors observed in numerous experimental models constitutes a paradigm of G-protein coupled receptor (GPCR) functional selectivity. We recently described that activation of Gαq/11 proteins through 5-HT2A serotonin receptors co-expressed in the same cells facilitates MOP receptor endocytosis promoted by morphine. In order to explore whether a different Gαq/11 coupled GPCR would emulate this effect, a double stable Flp-In T-REx HEK293 cell line permanently expressing MOP-YFP receptors along with FLAG-M3-Cerulean receptors expressed in an inducible manner was generated. Fluorescence microscopy examination of these cells revealed a co-distribution of both receptors mainly compartmentalized in plasma membrane. Concurrent stimulation with carbachol and morphine promoted MOP receptor internalization, desensitization and down-regulation and this facilitation was not dependent on PKC activation. Co-immunoprecipitation experiments demonstrated that FLAG-M3-Cerulean/MOP-YFP receptors interact forming heteromeric complexes in a time depending manner, i.e. the strongest interaction was detected after 96h of FLAG-M3-Cerulean induced expression. Under these experimental conditions, treatment of cells with carbachol plus morphine resulted in the internalization of both receptors within separated endocytic vesicles as visualized by confocal microscopy. This trafficking segregation observed for FLAG-M3-Cerulean and MOP-YFP receptors upon agonist stimulation suggests that this protein-protein interaction presents temporal and dynamic properties. Moreover, MOP-YFP receptor internalization facilitated by FLAG-M3-Cerulean receptors is independent of the constitution of heteromeric complexes.


Assuntos
Endocitose/genética , Morfina/metabolismo , Receptor Muscarínico M3/química , Receptores Opioides mu/química , Acetilcolina/metabolismo , Células HEK293 , Humanos , Morfina/química , Multimerização Proteica/genética , Transporte Proteico/genética , Receptor Muscarínico M3/genética , Receptores Opioides mu/genética , Transdução de Sinais/efeitos dos fármacos
10.
Sci Signal ; 9(410): ra5, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26758213

RESUMO

Heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) can form multiprotein complexes (heteromers), which can alter the pharmacology and functions of the constituent receptors. Previous findings demonstrated that the Gq/11-coupled serotonin 5-HT2A receptor and the Gi/o-coupled metabotropic glutamate 2 (mGlu2) receptor-GPCRs that are involved in signaling alterations associated with psychosis-assemble into a heteromeric complex in the mammalian brain. In single-cell experiments with various mutant versions of the mGlu2 receptor, we showed that stimulation of cells expressing mGlu2-5-HT2A heteromers with an mGlu2 agonist led to activation of Gq/11 proteins by the 5-HT2A receptors. For this crosstalk to occur, one of the mGlu2 subunits had to couple to Gi/o proteins, and we determined the relative location of the Gi/o-contacting subunit within the mGlu2 homodimer of the heteromeric complex. Additionally, mGlu2-dependent activation of Gq/11, but not Gi/o, was reduced in the frontal cortex of 5-HT2A knockout mice and was reduced in the frontal cortex of postmortem brains from schizophrenic patients. These findings offer structural insights into this important target in molecular psychiatry.


Assuntos
Multimerização Proteica , Receptor 5-HT2A de Serotonina/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Esquizofrenia/metabolismo , Transdução de Sinais , Regulação Alostérica , Animais , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Receptor 5-HT2A de Serotonina/genética , Receptores de Glutamato Metabotrópico/genética , Esquizofrenia/genética
11.
PLoS One ; 10(4): e0122604, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25849355

RESUMO

The activation of G-protein coupled receptors by agonist compounds results in diverse biological responses in cells, such as the endocytosis process consisting in the translocation of receptors from the plasma membrane to the cytoplasm within internalizing vesicles or endosomes. In order to functionally evaluate endocytosis events resulted from pharmacological responses, we have developed an image analysis method -the Q-Endosomes algorithm- that specifically discriminates the fluorescent signal originated at endosomes from that one observed at the plasma membrane in images obtained from living cells by fluorescence microscopy. Mu opioid (MOP) receptor tagged at the carboxy-terminus with yellow fluorescent protein (YFP) and permanently expressed in HEK293 cells was used as experimental model to validate this methodology. Time-course experiments performed with several agonists resulted in different sigmoid curves depending on the drug used to initiate MOP receptor endocytosis. Thus, endocytosis resulting from the simultaneous activation of co-expressed MOP and serotonin 5-HT2C receptors by morphine plus serotonin was significantly different, in kinetics as well as in maximal response parameters, from the one caused by DAMGO, sufentanyl or methadone. Therefore, this analytical tool permits the pharmacological characterization of receptor endocytosis in living cells with functional and temporal resolution.


Assuntos
Endocitose/efeitos dos fármacos , Receptor 5-HT2C de Serotonina/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Células HEK293 , Meia-Vida , Células HeLa , Humanos , Cinética , Microscopia de Fluorescência , Transporte Proteico , Serotonina/farmacologia , Agonistas do Receptor 5-HT2 de Serotonina/farmacologia , Vesículas Transportadoras/metabolismo
12.
Exp Brain Res ; 230(4): 395-406, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23864045

RESUMO

Earlier autoradiographic studies with the 5-HT2 receptor agonist [(125)I](±)DOI in human brain showed unexpected biphasic competition curves for various 5-HT2A antagonists. We have performed similar studies in rat brain regions with selective 5-HT2A (M100907) and 5-HT2C (SB242084) antagonists together with ketanserin and mesulergine. The effect of GTP analogues on antagonist competition was also studied. Increasing concentrations of Gpp(NH)p or GTPγS resulted in a maximal inhibition of [(125)I](±)DOI-specific binding of approximately 50 %. M100907 competed biphasically in all regions. In the presence of 100 µM Gpp(NH)p, M100907 still displaced biphasically the remaining [(125)I](±)DOI binding. Ketanserin showed biphasic curves in some regions and monophasic curves in others. In the latter, Gpp(NH)p evidenced an additional high-affinity site. SB242084 competed biphasically in brainstem nuclei and monophasically in the other regions. In most areas, SB242084 affinities were not notably altered by Gpp(NH)p. Mesulergine competed monophasically in all regions without alteration by Gpp(NH)p. These results conform with the extended ternary complex model of receptor action: receptor exists as an equilibrium of multiple conformations, i.e. ground (R), partly activated (R*) and activated G-protein-coupled (R*G) conformation/s. Thus, [(125)I](±)DOI would label multiple conformations of both 5-HT2A and 5-HT2C receptors in rat brain, and M100907 and ketanserin would recognise these conformations with different affinities.


Assuntos
Encéfalo/efeitos dos fármacos , Receptor 5-HT2A de Serotonina/química , Receptor 5-HT2C de Serotonina/química , Aminopiridinas/química , Aminopiridinas/farmacologia , Animais , Autorradiografia/métodos , Encéfalo/metabolismo , Ergolinas/química , Ergolinas/farmacologia , Indóis/química , Indóis/farmacologia , Ketanserina/química , Ketanserina/farmacologia , Masculino , Ratos , Ratos Wistar , Receptor 5-HT2A de Serotonina/metabolismo , Receptor 5-HT2C de Serotonina/metabolismo , Serotonina/metabolismo
13.
J Biol Chem ; 287(53): 44301-19, 2012 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-23129762

RESUMO

Serotonin and glutamate G protein-coupled receptor (GPCR) neurotransmission affects cognition and perception in humans and rodents. GPCRs are capable of forming heteromeric complexes that differentially alter cell signaling, but the role of this structural arrangement in modulating behavior remains unknown. Here, we identified three residues located at the intracellular end of transmembrane domain four that are necessary for the metabotropic glutamate 2 (mGlu2) receptor to be assembled as a GPCR heteromer with the serotonin 5-hydroxytryptamine 2A (5-HT(2A)) receptor in the mouse frontal cortex. Substitution of these residues (Ala-677(4.40), Ala-681(4.44), and Ala-685(4.48)) leads to absence of 5-HT(2A)·mGlu2 receptor complex formation, an effect that is associated with a decrease in their heteromeric ligand binding interaction. Disruption of heteromeric expression with mGlu2 attenuates the psychosis-like effects induced in mice by hallucinogenic 5-HT(2A) agonists. Furthermore, the ligand binding interaction between the components of the 5-HT(2A)·mGlu2 receptor heterocomplex is up-regulated in the frontal cortex of schizophrenic subjects as compared with controls. Together, these findings provide structural evidence for the unique behavioral function of a GPCR heteromer.


Assuntos
Receptor 5-HT2A de Serotonina/metabolismo , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/metabolismo , Esquizofrenia/metabolismo , Psicologia do Esquizofrênico , Adulto , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Comportamento , Estudos de Casos e Controles , Dimerização , Feminino , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Pessoa de Meia-Idade , Dados de Sequência Molecular , Ligação Proteica , Receptor 5-HT2A de Serotonina/genética , Receptores de Glutamato Metabotrópico/genética , Esquizofrenia/genética , Alinhamento de Sequência , Adulto Jovem
14.
CNS Neurol Disord Drug Targets ; 9(5): 616-26, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20632966

RESUMO

Internalisation of the mu opioid receptor from the surface of cells is generally achieved by receptor occupancy with agonist ligands of high efficacy. However, in many situations the potent analgesic morphine fails to promote internalisation effectively and whether there is a direct link between this and the propensity for the sustained use of morphine to result in both tolerance and dependence has been studied intensely. Although frequently described as a partial agonist, this characteristic appears insufficient to explain the poor capacity of morphine to promote internalisation of the mu opioid receptor. Experiments performed using both transfected cell systems and ex vivo/in vivo models have provided evidence that when morphine can promote internalisation of the mu receptor there is a decrease in the development of tolerance and dependence. Although aspects of this model are controversial, such observations suggest a number of approaches to further enhance the use of morphine as an analgesic.


Assuntos
Tolerância a Medicamentos/fisiologia , Endocitose/efeitos dos fármacos , Morfina/farmacologia , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Sequência de Aminoácidos/genética , Animais , Endocitose/fisiologia , Humanos , Modelos Biológicos , Receptores Opioides mu/genética
15.
Mol Pharmacol ; 75(6): 1380-91, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19279328

RESUMO

The serotonin (5-hydroxytryptamine; 5-HT) 2A receptor is a cell surface class A G protein-coupled receptor that regulates a multitude of physiological functions of the body and is a target for antipsychotic drugs. Here we found by means of fluorescence resonance energy transfer and immunoprecipitation studies that the 5-HT(2A)-receptor homodimerized in live cells, which we linked with its antagonist-dependent fingerprint in both binding and receptor signaling. Some antagonists, like the atypical antipsychotics clozapine and risperidone, differentiate themselves from others, like the typical antipsychotic haloperidol, antagonizing these 5-HT(2A) receptor-mediated functions in a pathway-specific manner, explained here by a new model of multiple active interconvertible conformations at dimeric receptors.


Assuntos
Antagonistas do Receptor 5-HT2 de Serotonina , Animais , Linhagem Celular , Cricetinae , Cricetulus , Transferência Ressonante de Energia de Fluorescência , Humanos , Imunoprecipitação , Modelos Biológicos , Conformação Proteica , Multimerização Proteica , Receptor 5-HT2A de Serotonina/fisiologia , Transdução de Sinais
16.
Biochem J ; 417(1): 161-72, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18764782

RESUMO

Many G-protein-coupled receptors, including the alpha(1b)-adrenoceptor, form homo-dimers or oligomers. Mutation of hydrophobic residues in transmembrane domains I and IV alters the organization of the alpha(1b)-adrenoceptor oligomer, with transmembrane domain IV playing a critical role. These mutations also result in endoplasmic reticulum trapping of the receptor. Following stable expression of this alpha(1b)-adrenoceptor mutant, cell surface delivery, receptor function and structural organization were recovered by treatment with a range of alpha(1b)-adrenoceptor antagonists that acted at the level of the endoplasmic reticulum. This was accompanied by maturation of the mutant receptor to a terminally N-glycosylated form, and only this mature form was trafficked to the cell surface. Co-expression of the mutant receptor with an otherwise wild-type form of the alpha(1b)-adrenoceptor that is unable to bind ligands resulted in this wild-type variant also being retained in the endoplasmic reticulum. Ligand-induced cell surface delivery of the mutant alpha(1b)-adrenoceptor now allowed co-recovery to the plasma membrane of the ligand-binding-deficient mutant. These results demonstrate that interactions between alpha(1b)-adrenoceptor monomers occur at an early stage in protein synthesis, that ligands of the alpha(1b)-adrenoceptor can act as pharmacological chaperones to allow a structurally compromised form of the receptor to pass cellular quality control, that the mutated receptor is not inherently deficient in function and that an oligomeric assembly of ligand-binding-competent and -incompetent forms of the alpha(1b)-adrenoceptor can be trafficked to the cell surface by binding of a ligand to only one component of the receptor oligomer.


Assuntos
Membrana Celular/metabolismo , Chaperonas Moleculares/farmacologia , Receptores Adrenérgicos alfa/metabolismo , Antagonistas Adrenérgicos alfa/farmacologia , Transporte Biológico/efeitos dos fármacos , Biotinilação/efeitos dos fármacos , Brefeldina A/farmacologia , Linhagem Celular , Dimerização , Retículo Endoplasmático/metabolismo , Ensaio de Imunoadsorção Enzimática , Glicosilação/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Mutação , Prazosina/farmacologia , Ligação Proteica/efeitos dos fármacos , Receptores Adrenérgicos alfa/química , Receptores Adrenérgicos alfa/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
17.
Mol Pharmacol ; 74(5): 1278-91, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18703670

RESUMO

Analysis of the distribution of mRNA encoding the serotonin (5-hydroxytryptamine) 5-HT(2A) receptor and the mu opioid peptide receptor in rat brain demonstrated their coexpression in neurons in several distinct regions. These regions included the periaqueductal gray, an area that plays an important role in morphine-induced analgesia but also in the development of tolerance to morphine. To explore potential cross-regulation between these G protein-coupled receptors, the human mu opioid peptide receptor was expressed stably and constitutively in Flp-In T-REx human embryonic kidney 293 cells that harbored the human 5-HT(2A) receptor at the inducible Flp-In locus. In the absence of the 5-HT(2A) receptor, pretreatment with the enkephalin agonist [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin but not with the alkaloid agonist morphine produced desensitization, internalization, and down-regulation of the mu opioid peptide receptor. Induction of 5-HT(2A) receptor expression in these cells resulted in up-regulation of mu opioid peptide receptor levels that was blocked by both a 5-HT(2A) receptor inverse agonist and selective inhibition of signaling via Galpha(q)/Galpha(11) G proteins. After induction of the 5-HT(2A) receptor, coaddition of 5-HT with morphine now also resulted in desensitization, receptor internalization, and down-regulation of the mu opioid peptide receptor. It has been argued that enhancement of mu opioid peptide receptor internalization in response to morphine would limit the development of tolerance without limiting analgesia. These data suggest that selective activation of the 5-HT(2A) receptor in concert with treatment with morphine might achieve this aim.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Morfina/farmacologia , Receptor 5-HT2A de Serotonina/metabolismo , Receptores Opioides mu/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Linhagem Celular , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Masculino , Hibridização de Ácido Nucleico , RNA Mensageiro/genética , Ensaio Radioligante , Ratos , Ratos Wistar , Receptor 5-HT2A de Serotonina/genética , Agonistas do Receptor 5-HT2 de Serotonina , Agonistas do Receptor de Serotonina/farmacologia
18.
Nature ; 452(7183): 93-7, 2008 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-18297054

RESUMO

The psychosis associated with schizophrenia is characterized by alterations in sensory processing and perception. Some antipsychotic drugs were identified by their high affinity for serotonin 5-HT2A receptors (2AR). Drugs that interact with metabotropic glutamate receptors (mGluR) also have potential for the treatment of schizophrenia. The effects of hallucinogenic drugs, such as psilocybin and lysergic acid diethylamide, require the 2AR and resemble some of the core symptoms of schizophrenia. Here we show that the mGluR2 interacts through specific transmembrane helix domains with the 2AR, a member of an unrelated G-protein-coupled receptor family, to form functional complexes in brain cortex. The 2AR-mGluR2 complex triggers unique cellular responses when targeted by hallucinogenic drugs, and activation of mGluR2 abolishes hallucinogen-specific signalling and behavioural responses. In post-mortem human brain from untreated schizophrenic subjects, the 2AR is upregulated and the mGluR2 is downregulated, a pattern that could predispose to psychosis. These regulatory changes indicate that the 2AR-mGluR2 complex may be involved in the altered cortical processes of schizophrenia, and this complex is therefore a promising new target for the treatment of psychosis.


Assuntos
Transtornos Psicóticos/metabolismo , Receptor 5-HT2A de Serotonina/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Linhagem Celular , Células Cultivadas , Regulação para Baixo , Alucinógenos/metabolismo , Alucinógenos/farmacologia , Humanos , Camundongos , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transtornos Psicóticos/tratamento farmacológico , Transtornos Psicóticos/genética , Receptor 5-HT2A de Serotonina/análise , Receptor 5-HT2A de Serotonina/deficiência , Receptor 5-HT2A de Serotonina/genética , Receptores de Glutamato Metabotrópico/análise , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/genética , Esquizofrenia/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima
19.
Mol Pharmacol ; 71(4): 1015-29, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17220353

RESUMO

Approaches to identify G protein-coupled receptor oligomers rather than dimers have been lacking. Using concatamers of fluorescent proteins, we established conditions to monitor sequential three-color fluorescence resonance energy transfer (3-FRET) and used these to detect oligomeric complexes of the alpha(1b)-adrenoceptor in single living cells. Mutation of putative key hydrophobic residues in transmembrane domains I and IV resulted in substantial reduction of sequential 3-FRET and was associated with lack of protein maturation, prevention of plasma membrane delivery, and elimination of signaling function. Although these mutations prevented cell surface delivery, bimolecular fluorescence complementation studies indicated that they did not ablate protein-protein interactions and confirmed endoplasmic reticulum/Golgi retention of the transmembrane domain I plus transmembrane domain IV mutated receptor. The transmembrane domain I plus transmembrane domain IV mutated receptor was a "dominant-negative" in blocking cell surface delivery of the wild-type receptor. Mutations only in transmembrane domain I did not result in a reduction in 3-FRET, whereas restricting mutation to transmembrane domain IV did result in reduced 3-FRET. Mutations in either transmembrane domain I or transmembrane domain IV, however, were sufficient to eliminate cell surface delivery. Terminal N-glycosylation is insufficient to determine cell surface delivery because both transmembrane domain I and transmembrane domain IV mutants matured as effectively as the wild-type receptor. These data indicate that the alpha(1b)-adrenoceptor is able to form oligomeric rather than only simple dimeric complexes and that disruption of effective oligomerization by introducing mutations into transmembrane domain IV has profound consequences for cell surface delivery and function.


Assuntos
Receptores Adrenérgicos alfa 1/química , Receptores Adrenérgicos alfa 1/fisiologia , Linhagem Celular , Membrana Celular/metabolismo , Dimerização , Transferência Ressonante de Energia de Fluorescência , Glicosilação , Humanos , Mutagênese Sítio-Dirigida , Transporte Proteico , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Receptores de Superfície Celular , Transfecção
20.
Vision Res ; 46(27): 4434-41, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17005232

RESUMO

The structural basis of the quaternary organization of rhodopsin has recently been explored and modeled. Because information obtained from studying rhodopsin has frequently been directly applicable to other G protein-coupled receptors we wished to ascertain if dimeric and/or oligomeric forms of the alpha(1b)-adrenoceptor could be observed and if so whether rhodopsin might provide insights into the quaternary structure of this receptor. Co-immunoprecipitation and both conventional and time-resolved fluorescence resonance energy transfer studies demonstrated quaternary structure of the alpha(1b)-adrenoceptor and, in concert with the reconstitution of fragments of this receptor, provided information on the molecular basis of these interactions. Development of three color fluorescence resonance energy transfer (FRET) allowed the imaging of alpha(1b)-adrenoceptor oligomers in single living cells. Mutation of hydrophobic residues in transmembrane domains I and IV of the receptor resulted in marked reduction in three color FRET suggesting an alteration in oligomeric organization and potential similarities with rhodopsin. The mutated alpha(1b)-adrenoceptor was unable to reach the cell surface, did not become terminally N-glycosylated and was unable to signal.


Assuntos
Receptores Adrenérgicos alfa 1/química , Rodopsina/química , Animais , Membrana Celular/metabolismo , Dimerização , Humanos , Mutação , Estrutura Quaternária de Proteína , Receptores Adrenérgicos alfa 1/genética , Segmento Externo da Célula Bastonete/metabolismo , Visão Ocular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...