Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Handb Exp Pharmacol ; 247: 73-85, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28689302

RESUMO

Delta opioid receptors (δORs) regulate a number of physiological functions, and agonists for this receptor are being pursued for the treatment of mood disorders, chronic pain, and migraine. A major challenge to the development of these compounds is that, like many G-protein coupled receptors (GPCRs), agonists at the δOR can induce very different signaling and receptor trafficking events. This concept, known as ligand-directed signaling, functional selectivity, or biased agonism, can result in different agonists producing highly distinct behavioral consequences. In this chapter, we highlight the in vitro and in vivo evidence for ligand-directed signaling and trafficking at the δOR. A number of biological implications of agonist-directed signaling at the δOR have been demonstrated. Importantly, ligand-specific effects can impact both acute behavioral effects of delta agonists, as well as the long-term adaptations induced by chronic drug treatment. A better understanding of the specific signaling cascades that regulate these differential behavioral effects would help to guide rational drug design, ultimately resulting in δOR agonists with fewer adverse effects.


Assuntos
Receptores Opioides delta/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Desenho de Fármacos , Humanos , Técnicas In Vitro , Ligantes
2.
J Neurosci ; 36(12): 3541-51, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27013682

RESUMO

Ligand-specific recruitment of arrestins facilitates functional selectivity of G-protein-coupled receptor signaling. Here, we describe agonist-selective recruitment of different arrestin isoforms to the delta opioid receptor in mice. A high-internalizing delta opioid receptor agonist (SNC80) preferentially recruited arrestin 2 and, in arrestin 2 knock-outs (KOs), we observed a significant increase in the potency of SNC80 to inhibit mechanical hyperalgesia and decreased acute tolerance. In contrast, the low-internalizing delta agonists (ARM390, JNJ20788560) preferentially recruited arrestin 3 with unaltered behavioral effects in arrestin 2 KOs. Surprisingly, arrestin 3 KO revealed an acute tolerance to these low-internalizing agonists, an effect never observed in wild-type animals. Furthermore, we examined delta opioid receptor-Ca(2+)channel coupling in dorsal root ganglia desensitized by ARM390 and the rate of resensitization was correspondingly decreased in arrestin 3 KOs. Live-cell imaging in HEK293 cells revealed that delta opioid receptors are in pre-engaged complexes with arrestin 3 at the cell membrane and that ARM390 strengthens this membrane interaction. The disruption of these complexes in arrestin 3 KOs likely accounts for the altered responses to low-internalizing agonists. Together, our results show agonist-selective recruitment of arrestin isoforms and reveal a novel endogenous role of arrestin 3 as a facilitator of resensitization and an inhibitor of tolerance mechanisms. SIGNIFICANCE STATEMENT: Agonists that bind to the same receptor can produce highly distinct signaling events and arrestins are a major mediator of this ligand bias. Here, we demonstrate that delta opioid receptor agonists differentially recruit arrestin isoforms. We found that the high-internalizing agonist SNC80 preferentially recruits arrestin 2 and knock-out (KO) of this protein results in increased efficacy of SNC80. In contrast, low-internalizing agonists (ARM390 and JNJ20788560) preferentially recruit arrestin 3 and, surprisingly, KO of arrestin 3 produces acute tolerance and impaired receptor resensitization to these agonists. Arrestin 3 is in pre-engaged complexes with the delta opioid receptor at the cell membrane and low-internalizing agonists promote this interaction. This study reveals a novel role for arrestin 3 as a facilitator of receptor resensitization.


Assuntos
Arrestinas/metabolismo , Benzamidas/administração & dosagem , Hiperalgesia/fisiopatologia , Percepção da Dor , Piperazinas/administração & dosagem , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/metabolismo , Animais , Tolerância a Medicamentos , Feminino , Masculino , Camundongos , Camundongos Knockout , Isoformas de Proteínas
3.
Cell Mol Life Sci ; 68(17): 2933-49, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21153910

RESUMO

A series of pharmacological and physiological studies have demonstrated the functional cross-regulation between MOR and NMDAR. These receptors coexist at postsynaptic sites in midbrain periaqueductal grey (PAG) neurons, an area implicated in the analgesic effects of opioids like morphine. In this study, we found that the MOR-associated histidine triad nucleotide-binding protein 1 (HINT1) is essential for maintaining the connection between the NMDAR and MOR. Morphine-induced analgesic tolerance is prevented and even rescued by inhibiting PKC or by antagonizing NMDAR. However, in the absence of HINT1, the MOR becomes supersensitive to morphine before suffering a profound and lasting desensitization that is refractory to PKC inhibition or NMDAR antagonism. Thus, HINT1 emerges as a key protein that is critical for sustaining NMDAR-mediated regulation of MOR signaling strength. Thus, HINT1 deficiency may contribute to opioid-intractable pain syndromes by causing long-term MOR desensitization via mechanisms independent of NMDAR.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides mu/metabolismo , Animais , Dimerização , Masculino , Camundongos , Camundongos Knockout , Morfina/farmacologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Proteínas RGS/química , Proteínas RGS/genética , Proteínas RGS/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Transdução de Sinais
4.
Mol Pain ; 5: 11, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19284549

RESUMO

BACKGROUND: Although the systemic administration of cannabinoids produces antinociception, their chronic use leads to analgesic tolerance as well as cross-tolerance to morphine. These effects are mediated by cannabinoids binding to peripheral, spinal and supraspinal CB1 and CB2 receptors, making it difficult to determine the relevance of each receptor type to these phenomena. However, in the brain, the CB1 receptors (CB1Rs) are expressed at high levels in neurons, whereas the expression of CB2Rs is marginal. Thus, CB1Rs mediate the effects of smoked cannabis and are also implicated in emotional behaviors. We have analyzed the production of supraspinal analgesia and the development of tolerance at CB1Rs by the direct injection of a series of cannabinoids into the brain. The influence of the activation of CB1Rs on supraspinal analgesia evoked by morphine was also evaluated. RESULTS: Intracerebroventricular (icv) administration of cannabinoid receptor agonists, WIN55,212-2, ACEA or methanandamide, generated a dose-dependent analgesia. Notably, a single administration of these compounds brought about profound analgesic tolerance that lasted for more than 14 days. This decrease in the effect of cannabinoid receptor agonists was not mediated by depletion of CB1Rs or the loss of regulated G proteins, but, nevertheless, it was accompanied by reduced morphine analgesia. On the other hand, acute morphine administration produced tolerance that lasted only 3 days and did not affect the CB1R. We found that both neural mu-opioid receptors (MORs) and CB1Rs interact with the HINT1-RGSZ module, thereby regulating pertussis toxin-insensitive Gz proteins. In mice with reduced levels of these Gz proteins, the CB1R agonists produced no such desensitization or morphine cross-tolerance. On the other hand, experimental enhancement of Gz signaling enabled an acute icv administration of morphine to produce a long-lasting tolerance at MORs that persisted for more than 2 weeks, and it also impaired the analgesic effects of cannabinoids. CONCLUSION: In the brain, cannabinoids can produce analgesic tolerance that is not associated with the loss of surface CB1Rs or their uncoupling from regulated transduction. Neural specific Gz proteins are essential mediators of the analgesic effects of supraspinal CB1R agonists and morphine. These Gz proteins are also responsible for the long-term analgesic tolerance produced by single doses of these agonists, as well as for the cross-tolerance between CB1Rs and MORs.


Assuntos
Tolerância a Medicamentos , Subunidades alfa de Proteínas de Ligação ao GTP/fisiologia , Morfina/farmacologia , Receptor CB1 de Canabinoide/agonistas , Receptores Opioides mu/efeitos dos fármacos , Analgesia , Animais , Química Encefálica , Canabinoides/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptor CB1 de Canabinoide/fisiologia
5.
Int J Lab Hematol ; 41(1): 109-117, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30290085

RESUMO

INTRODUCTION: Diagnosis of myelodysplastic syndromes (MDSs) when anemia is the only abnormality can be complicated. The aim of our study was to investigate the primary causes of anemia and/or macrocytosis of uncertain etiology. METHODS: We conducted a multicenter, prospective study over 4 months in three hematology laboratories. In step 1, we used an automated informatics system to screen 137 453 hemograms for cases of anemia and/or macrocytosis (n = 2702). In step 2, we excluded all patients whose anemia appeared to be due to a known cause. This left 290 patients had anemia of uncertain etiology. In step 3, we conducted further investigations, including a peripheral blood smear, and analysis of iron, vitamin B12, folate, and thyroid hormone levels. RESULTS: A differential diagnosis was obtained in 139 patients (48%). The primary causes of anemia were iron deficiency (n = 59) and megaloblastic anemia (n = 39). In total, 25 hematologic disorders were diagnosed, including 14 patients with MDS (56%). The median age of MDS patients was 80 years, 12 had anemia as an isolated cytopenia, and most (n = 10) had lower-risk disease (IPSS-R ≤ 3.5). SF3B1 mutations were most frequent (n = 6) and correlated with the presence of ring sideroblasts (100%) and associated with better prognosis (P = 0.001). CONCLUSIONS: Our prospective, four-step approach is an efficient and logical strategy to facilitate the diagnosis of MDS on the basis of unexplained anemia and/or macrocytosis, and may allow the early diagnosis of the most serious causes of anemia. Molecular analysis of genes related to MDS could be a promising diagnostic and prognostic approach.


Assuntos
Anemia/etiologia , Síndromes Mielodisplásicas/diagnóstico , Idoso , Idoso de 80 Anos ou mais , Anemia/complicações , Anemia/diagnóstico , Anemia Macrocítica , Anemia Megaloblástica , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Mutação , Síndromes Mielodisplásicas/complicações , Fosfoproteínas/genética , Estudos Prospectivos , Fatores de Processamento de RNA/genética
6.
Br J Pharmacol ; 175(14): 3050-3059, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29722902

RESUMO

BACKGROUND AND PURPOSE: Opioid δ receptor agonists are potent antihyperalgesics in chronic pain models, but tolerance develops after prolonged use. Previous evidence indicates that distinct forms of tolerance occur depending on the internalization properties of δ receptor agonists. As arrestins are important in receptor internalization, we investigated the role of arrestin 2 (ß-arrestin 1) in mediating the development of tolerance induced by high- and low-internalizing δ receptor agonists. EXPERIMENTAL APPROACH: We evaluated the effect of two δ receptor agonists with similar analgesic potencies, but either high-(SNC80) or low-(ARM390) internalization properties in wild-type (WT) and arrestin 2 knockout (KO) mice. We compared tolerance to the antihyperalgesic effects of these compounds in a model of inflammatory pain. We also examined tolerance to the convulsant effect of SNC80. Furthermore, effect of chronic treatment with SNC80 on δ agonist-stimulated [35 S]-GTPγS binding was determined in WT and KO mice. KEY RESULTS: Arrestin 2 KO resulted in increased drug potency, duration of action and decreased acute tolerance to the antihyperalgesic effects of SNC80. In contrast, ARM390 produced similar effects in both WT and KO animals. Following chronic treatment, we found a marked decrease in the extent of tolerance to SNC80-induced antihyperalgesia and convulsions in arrestin 2 KO mice. Accordingly, δ receptors remained functionally coupled to G proteins in arrestin 2 KO mice chronically treated with SNC80. CONCLUSIONS AND IMPLICATIONS: Overall, these results suggest that δ receptor agonists interact with arrestins in a ligand-specific manner, and tolerance to high- but not low-internalizing agonists are preferentially regulated by arrestin 2.


Assuntos
Analgésicos/farmacologia , Benzamidas/farmacologia , Tolerância a Medicamentos/fisiologia , Piperazinas/farmacologia , Receptores Opioides delta/agonistas , beta-Arrestina 1/metabolismo , Analgésicos/uso terapêutico , Animais , Benzamidas/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Camundongos Knockout , Dor/tratamento farmacológico , Dor/metabolismo , Piperazinas/uso terapêutico , Receptores Opioides delta/metabolismo , Convulsões/induzido quimicamente , beta-Arrestina 1/genética
7.
Neuroscience ; 338: 145-159, 2016 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-27349452

RESUMO

In recent years, the delta opioid receptor has attracted increasing interest as a target for the treatment of chronic pain and emotional disorders. Due to their therapeutic potential, numerous tools have been developed to study the delta opioid receptor from both a molecular and a functional perspective. This review summarizes the most commonly available tools, with an emphasis on their use and limitations. Here, we describe (1) the cell-based assays used to study the delta opioid receptor. (2) The features of several delta opioid receptor ligands, including peptide and non-peptide drugs. (3) The existing approaches to detect delta opioid receptors in fixed tissue, and debates that surround these techniques. (4) Behavioral assays used to study the in vivo effects of delta opioid receptor agonists; including locomotor stimulation and convulsions that are induced by some ligands, but not others. (5) The characterization of genetically modified mice used specifically to study the delta opioid receptor. Overall, this review aims to provide a guideline for the use of these tools with the final goal of increasing our understanding of delta opioid receptor physiology.


Assuntos
Receptores Opioides delta/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos Transgênicos , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/genética , Projetos de Pesquisa
8.
Neuropharmacology ; 89: 412-23, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25445489

RESUMO

In the nervous system, the glutamate N-methyl-D-aspartate receptor (NMDAR) restricts the activity of the mu-opioid receptor (MOR). Both receptors are present in midbrain periaqueductal grey (PAG) neurons, an area that plays a central role in the supraspinal antinociceptive effects of opioids. The cross-talk that occurs between these receptors is sustained by the MOR-associated histidine triad nucleotide binding protein 1 (HINT1), which displays nucleoside phosphoramidase and acyl-AMP hydrolase activity. Here we report that the inhibitor of HINT1 enzymatic activity guanosine-5'-tryptamine carbamate (TpGc) significantly enhanced morphine antinociception while preventing the development of tolerance. At the molecular level, TpGc reduced the capacity of MORs to recruit NMDAR activity to negatively regulate opioid signaling. In mice suffering from chronic constriction injury concurrent with increased NMDAR activity, a single intracerebroventricular administration of TpGc attenuated NMDAR function and alleviated mechanical allodynia for several days. These data suggest a potential therapeutic role for HINT1 inhibitors in the clinical management of acute and neuropathic pain.


Assuntos
Analgésicos Opioides/uso terapêutico , Hiperalgesia/tratamento farmacológico , Morfina/uso terapêutico , Proteínas do Tecido Nervoso/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Modelos Animais de Doenças , Tolerância a Medicamentos , Comportamento Exploratório/efeitos dos fármacos , Hiperalgesia/etiologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Traumatismos dos Nervos Periféricos/complicações , Receptores de N-Metil-D-Aspartato/genética , Natação/psicologia
9.
Antioxid Redox Signal ; 19(15): 1766-82, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-23600761

RESUMO

AIMS: Overactivation of glutamate N-methyl-D-aspartate receptor (NMDAR) increases the cytosolic concentrations of calcium and zinc, which significantly contributes to neural death. Since cannabinoids prevent the NMDAR-mediated increase in cytosolic calcium, we investigated whether they also control the rise of potentially toxic free zinc ions, as well as the processes implicated in this phenomenon. RESULTS: The cannabinoid receptors type 1 (CNR1) and NMDARs are cross-regulated in different regions of the nervous system. Cannabinoids abrogated the stimulation of the nitric oxide-zinc pathway by NMDAR, an effect that required the histidine triad nucleotide-binding protein 1 (HINT1). Conversely, NMDAR antagonism reduced the analgesia promoted by the CNR1 agonist WIN55,212-2 and impaired its capacity to internalize CNR1s. At the cell surface, CNR1s co-immunoprecipitated with the NR1 subunits of NMDARs, an association that diminished after the administration of NMDA in vivo or as a consequence of neuropathic overactivation of NMDARs, both situations in which cannabinoids do not control NMDAR activity. Under these circumstances, inhibition of protein kinase A (PKA) restored the association between CNR1s and NR1 subunits, and cannabinoids regained control over NMDAR activity. Notably, CNR1 and NR1 associated poorly in HINT1(-/-) mice, in which there was little cross-regulation between these receptors. INNOVATION: The CNR1 can regulate NMDAR function when the receptor is coupled to HINT1. Thus, internalization of CNR1s drives the co-internalization of the NR1 subunits, neutralizing the overactivation of NMDARs. CONCLUSION: Cannabinoids require the HINT1 protein to counteract the toxic effects of NMDAR-mediated NO production and zinc release. This study situates the HINT1 protein at the forefront of cannabinoid protection against NMDAR-mediated brain damage.


Assuntos
Ácido Glutâmico/metabolismo , Óxido Nítrico/biossíntese , Receptores de Canabinoides/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Zinco/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Canabinoides/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ácido Glutâmico/farmacologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Sistema Nervoso/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteína Quinase C/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptores de Canabinoides/química , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/química , Transdução de Sinais
10.
Mol Brain ; 6: 42, 2013 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-24093505

RESUMO

BACKGROUND: G protein-coupled receptors (GPCRs) are the targets of a large number of drugs currently in therapeutic use. Likewise, the glutamate ionotropic N-methyl-D-aspartate receptor (NMDAR) has been implicated in certain neurological disorders, such as neurodegeration, neuropathic pain and mood disorders, as well as psychosis and schizophrenia. Thus, there is now an important need to characterize the interactions between GPCRs and NMDARs. Indeed, these interactions can produce distinct effects, and whereas the activation of Mu-opioid receptor (MOR) increases the calcium fluxes associated to NMDARs, that of type 1 cannabinoid receptor (CNR1) antagonizes their permeation. Notably, a series of proteins interact with these receptors affecting their responses and interactions, and then emerge as novel therapeutic targets for the aforementioned pathologies. RESULTS: We found that in the presence of GPCRs, the HINT1 protein influences the activity of NMDARs, whereby NMDAR activation was enhanced in CNR1+/+/HINT1-/- cortical neurons and the cannabinoid agonist WIN55,212-2 provided these cells with no protection against a NMDA insult. NMDAR activity was normalized in these cells by the lentiviral expression of HINT1, which also restored the neuroprotection mediated by cannabinoids. NMDAR activity was also enhanced in CNR1-/-/HINT1+/+ neurons, although this activity was dampened by the expression of GPCRs like the MOR, CNR1 or serotonin 1A (5HT1AR). CONCLUSIONS: The HINT1 protein plays an essential role in the GPCR-NMDAR connection. In the absence of receptor activation, GPCRs collaborate with HINT1 proteins to negatively control NMDAR activity. When activated, most GPCRs release the control of HINT1 and NMDAR responsiveness is enhanced. However, cannabinoids that act through CNR1 maintain the negative control of HINT1 on NMDAR function and their protection against glutamate excitotoxic insult persists.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Benzoxazinas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Lentivirus/metabolismo , Camundongos , Morfolinas/farmacologia , N-Metilaspartato/toxicidade , Naftalenos/farmacologia , Proteínas do Tecido Nervoso/deficiência , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Ligação Proteica/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Receptor CB1 de Canabinoide/deficiência , Receptores Opioides mu/metabolismo , Zinco/metabolismo
11.
Neuropsychopharmacology ; 37(2): 338-49, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21814188

RESUMO

The capacity of opioids to alleviate inflammatory pain is negatively regulated by the glutamate-binding N-methyl-D-aspartate receptor (NMDAR). Increased activity of this receptor complicates the clinical use of opioids to treat persistent neuropathic pain. Immunohistochemical and ultrastructural studies have demonstrated the coexistence of both receptors within single neurons of the CNS, including those in the mesencephalic periaqueductal gray (PAG), a region that is implicated in the opioid control of nociception. We now report that mu-opioid receptors (MOR) and NMDAR NR1 subunits associate in the postsynaptic structures of PAG neurons. Morphine disrupts this complex by protein kinase-C (PKC)-mediated phosphorylation of the NR1 C1 segment and potentiates the NMDAR-CaMKII, pathway that is implicated in morphine tolerance. Inhibition of PKC, but not PKA or GRK2, restored the MOR-NR1 association and rescued the analgesic effect of morphine as well. The administration of N-methyl-D-aspartic acid separated the MOR-NR1 complex, increased MOR Ser phosphorylation, reduced the association of the MOR with G-proteins, and diminished the antinociceptive capacity of morphine. Inhibition of PKA, but not PKC, CaMKII, or GRK2, blocked these effects and preserved morphine antinociception. Thus, the opposing activities of the MOR and NMDAR in pain control affect their relation within neurons of structures such as the PAG. This finding could be exploited in developing bifunctional drugs that would act exclusively on those NMDARs associated with MORs.


Assuntos
Dor/fisiopatologia , Substância Cinzenta Periaquedutal/metabolismo , Agregação de Receptores/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides mu/metabolismo , Animais , Células CHO , Cricetinae , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Tolerância a Medicamentos/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos ICR , Morfina/administração & dosagem , Morfina/antagonistas & inibidores , Morfina/farmacologia , N-Metilaspartato/farmacologia , Neurônios/metabolismo , Dor/prevenção & controle , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/fisiologia , Agregação de Receptores/efeitos dos fármacos
12.
Antioxid Redox Signal ; 15(4): 873-87, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21348811

RESUMO

UNLABELLED: Morphine increases the production of nitric oxide (NO) via the phosphoinositide 3-kinase/Akt/neural nitric oxide synthase (nNOS) pathway. Subsequently, NO enhances N-methyl-D-aspartate receptor (NMDAR)/calmodulin-dependent protein kinase II (CaMKII) cascade, diminishing the strength of morphine-activated Mu-opioid receptor (MOR) signaling. During this process, NO signaling is restricted by the association of nNOS to the MOR. AIMS: Here, we examined how nNOS/NO signaling is downregulated by the morphine-activated MOR and how this regulation affects antinociception. RESULTS: Accordingly, we show that the MOR-NMDAR regulatory loop relies on the negative control of nNOS activity exerted by RGSZ2, a protein physically coupled to the MOR. This regulation requires binding of the nNOS N terminal PDZ domain to the RGSZ2 PDZ binding motifs that lie upstream of the RGS box. INNOVATION: Indeed, in RGSZ2-deficient mice morphine over-stimulates the nNOS/NO/NMDAR/CaMKII pathway, causing analgesic tolerance to develop rapidly. Recovery of RGSZ2 levels or inhibition of nNOS, protein kinase C, NMDAR, or CaMKII function restores MOR signaling and morphine recovers its full analgesic potency. CONCLUSION: This RGSZ2-dependent regulation of NMDAR activity is relevant to persistent pain disorders associated with heightened NMDAR-mediated glutamate responses and the reduced antinociceptive capacity of opioids.


Assuntos
Analgésicos Opioides/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Morfina/farmacologia , Óxido Nítrico Sintase Tipo I/metabolismo , Proteínas RGS/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides mu/agonistas , Sequência de Aminoácidos , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Tolerância a Medicamentos , Masculino , Redes e Vias Metabólicas , Camundongos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Domínios PDZ , Proteínas RGS/genética , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores Opioides mu/metabolismo , Transdução de Sinais
13.
PLoS One ; 6(12): e28557, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22163035

RESUMO

The RGSZ2 gene, a regulator of G protein signaling, has been implicated in cognition, Alzheimer's disease, panic disorder, schizophrenia and several human cancers. This 210 amino acid protein is a GTPase accelerating protein (GAP) on Gαi/o/z subunits, binds to the N terminal of neural nitric oxide synthase (nNOS) negatively regulating the production of nitric oxide, and binds to the histidine triad nucleotide-binding protein 1 at the C terminus of different G protein-coupled receptors (GPCRs). We now describe a novel regulatory mechanism of RGS GAP function through the covalent incorporation of Small Ubiquitin-like MOdifiers (SUMO) into RGSZ2 RGS box (RH) and the SUMO non covalent binding with SUMO-interacting motifs (SIM): one upstream of the RH and a second within this region. The covalent attachment of SUMO does not affect RGSZ2 binding to GPCR-activated GαGTP subunits but abolishes its GAP activity. By contrast, non-covalent binding of SUMO with RH SIM impedes RGSZ2 from interacting with GαGTP subunits. Binding of SUMO to the RGSZ2 SIM that lies outside the RH does not affect GαGTP binding or GAP activity, but it could lead to regulatory interactions with sumoylated proteins. Thus, sumoylation and SUMO-SIM interactions constitute a new regulatory mechanism of RGS GAP function and therefore of GPCR cell signaling as well.


Assuntos
Regulação da Expressão Gênica , Proteínas RGS/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Motivos de Aminoácidos , Animais , Células CHO , Cricetinae , Proteínas Ativadoras de GTPase/metabolismo , Masculino , Camundongos , Modelos Biológicos , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/métodos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Sinaptossomos/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa