Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 264
Filtrar
1.
Brain ; 144(2): 665-681, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33367648

RESUMO

Opioids such as morphine are mainstay treatments for clinical pain conditions. Itch is a common side effect of opioids, particularly as a result of epidural or intrathecal administration. Recent progress has advanced our understanding of itch circuits in the spinal cord. However, the mechanisms underlying opioid-induced itch are not fully understood, although an interaction between µ-opioid receptor (MOR) and gastrin-releasing peptide receptor (GRPR) in spinal GRPR-expressing neurons has been implicated. In this study we investigated the cellular mechanisms of intrathecal opioid-induced itch by conditional deletion of MOR-encoding Oprm1 in distinct populations of interneurons and sensory neurons. We found that intrathecal injection of the MOR agonists morphine or DAMGO elicited dose-dependent scratching as well as licking and biting, but this pruritus was totally abolished in mice with a specific Oprm1 deletion in Vgat+ neurons [Oprm1-Vgat (Slc32a1)]. Loss of MOR in somatostatin+ interneurons and TRPV1+ sensory neurons did not affect morphine-induced itch but impaired morphine-induced antinociception. In situ hybridization revealed Oprm1 expression in 30% of inhibitory and 20% of excitatory interneurons in the spinal dorsal horn. Whole-cell recordings from spinal cord slices showed that DAMGO induced outward currents in 9 of 19 Vgat+ interneurons examined. Morphine also inhibited action potentials in Vgat+ interneurons. Furthermore, morphine suppressed evoked inhibitory postsynaptic currents in postsynaptic Vgat- excitatory neurons, suggesting a mechanism of disinhibition by MOR agonists. Notably, morphine-elicited itch was suppressed by intrathecal administration of NPY and abolished by spinal ablation of GRPR+ neurons with intrathecal injection of bombesin-saporin, whereas intrathecal GRP-induced itch response remained intact in mice lacking Oprm1-Vgat. Intrathecal bombesin-saporin treatment reduced the number of GRPR+ neurons by 97% in the lumber spinal cord and 91% in the cervical spinal cord, without changing the number of Oprm1+ neurons. Additionally, chronic itch from DNFB-induced allergic contact dermatitis was decreased by Oprm1-Vgat deletion. Finally, naloxone, but not peripherally restricted naloxone methiodide, inhibited chronic itch in the DNFB model and the CTCL model, indicating a contribution of central MOR signalling to chronic itch. Our findings demonstrate that intrathecal morphine elicits itch via acting on MOR on spinal inhibitory interneurons, leading to disinhibition of the spinal itch circuit. Our data also provide mechanistic insights into the current treatment of chronic itch with opioid receptor antagonist such as naloxone.


Assuntos
Analgésicos/administração & dosagem , Morfina/administração & dosagem , Prurido/induzido quimicamente , Prurido/fisiopatologia , Receptores Opioides mu/fisiologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiopatologia , Animais , Dermatite/fisiopatologia , Feminino , Injeções Espinhais , Interneurônios/efeitos dos fármacos , Interneurônios/fisiologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Receptores da Bombesina/fisiologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia , Canais de Cátion TRPV/fisiologia , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/fisiologia
2.
J Neurosci ; 40(34): 6522-6535, 2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32690613

RESUMO

Chronic pain caused by spinal cord injury (SCI) is notoriously resistant to treatment, particularly by opioids. After SCI, DRG neurons show hyperactivity and chronic depolarization of resting membrane potential (RMP) that is maintained by cAMP signaling through PKA and EPAC. Importantly, SCI also reduces the negative regulation by Gαi of adenylyl cyclase and its production of cAMP, independent of alterations in G protein-coupled receptors and/or G proteins. Opioid reduction of pain depends on coupling of opioid receptors to Gαi/o family members. Combining high-content imaging and cluster analysis, we show that in male rats SCI decreases opioid responsiveness in vitro within a specific subset of small-diameter nociceptors that bind isolectin B4. This SCI effect is mimicked in nociceptors from naive animals by a modest 5 min depolarization of RMP (15 mm K+; -45 mV), reducing inhibition of cAMP signaling by µ-opioid receptor agonists DAMGO and morphine. Disinhibition and activation of C-Raf by depolarization-dependent phosphorylation are central to these effects. Expression of an activated C-Raf reduces sensitivity of adenylyl cyclase to opioids in nonexcitable HEK293 cells, whereas inhibition of C-Raf or treatment with the hyperpolarizing drug retigabine restores opioid responsiveness and blocks spontaneous activity of nociceptors after SCI. Inhibition of ERK downstream of C-Raf also blocks SCI-induced hyperexcitability and depolarization, without direct effects on opioid responsiveness. Thus, depolarization-dependent C-Raf and downstream ERK activity maintain a depolarized RMP and nociceptor hyperactivity after SCI, providing a self-reinforcing mechanism to persistently promote nociceptor hyperexcitability and limit the therapeutic effectiveness of opioids.SIGNIFICANCE STATEMENT Chronic pain induced by spinal cord injury (SCI) is often permanent and debilitating, and usually refractory to treatment with analgesics, including opioids. SCI-induced pain in a rat model has been shown to depend on persistent hyperactivity in primary nociceptors (injury-detecting sensory neurons), associated with a decrease in the sensitivity of adenylyl cyclase production of cAMP to inhibitory Gαi proteins in DRGs. This study shows that SCI and one consequence of SCI (chronic depolarization of resting membrane potential) decrease sensitivity to opioid-mediated inhibition of cAMP and promote hyperactivity of nociceptors by enhancing C-Raf activity. ERK activation downstream of C-Raf is necessary for maintaining ongoing depolarization and hyperactivity, demonstrating an unexpected positive feedback loop to persistently promote pain.


Assuntos
Dor Crônica/fisiopatologia , Nociceptores/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Receptores Opioides mu/fisiologia , Transdução de Sinais , Traumatismos da Medula Espinal/fisiopatologia , Animais , Células Cultivadas , Dor Crônica/complicações , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiopatologia , Células HEK293 , Humanos , Masculino , Potenciais da Membrana , Ratos Sprague-Dawley , Receptores Opioides mu/agonistas , Traumatismos da Medula Espinal/complicações
3.
Bull Exp Biol Med ; 168(6): 727-729, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32337663

RESUMO

We studied the role of opioid receptor subtypes in improvement of the functional state of the heart during reperfusion after adaptation to continuous normobaric hypoxia. To this end, male Wistar rats were subjected to continuous normobaric hypoxia (12% O2). Then, the hearts were isolated and exposed to total 45-min ischemia followed by 30-min reperfusion. Opioid receptor antagonists were added to the perfusion solution prior to ischemia. It was found that continuous normobaric hypoxia reduced the release of creatine phosphokinase into the effluent, increased myocardial contractile force, and decreased the end-diastolic pressure during reperfusion; these positive effects were related to activation of cardiac δ2- and µ-opioid receptors.


Assuntos
Hipóxia/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides delta/fisiologia , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/fisiologia , Adaptação Fisiológica/efeitos dos fármacos , Animais , Compostos de Benzilideno/farmacologia , Creatina Quinase/metabolismo , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Oligopeptídeos/farmacologia , Técnicas de Cultura de Órgãos , Peptídeos , Ratos , Ratos Wistar , Receptores Opioides mu/antagonistas & inibidores , Tetra-Hidroisoquinolinas/farmacologia
4.
Behav Brain Res ; 374: 112123, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31376441

RESUMO

Previous studies have uncovered a potential role of the opioid system in iron hemostasis and dopamine metabolism. Abnormalities in both of these systems have been noted in human RLS. Autopsy studies of human RLS have shown an endogenous opioid deficiency in the thalamus. Opioids, particularly prolonged-release oxycodone/naloxone, have been approved in Europe to be a second-line therapy for severe restless legs syndrome (RLS). To study the role of opioid receptors in the pathogenesis of RLS, we used a triple knockout (KO) mouse strain that lack mu, delta, and kappa opioid receptors and explored the behavioral and biochemical parameters relevant to RLS. The triple KO mice showed hyperactivity and a trend of increased probability of waking during the rest period (day) akin to that in human RLS (night). Surprisingly, triple KO mice also exhibit decreased serum iron concentration, evidence of anemia, a significant dysfunction in dopamine metabolism akin to that noted in human RLS, as well as an increased latency in response to thermal stimuli. To our knowledge, this is the first study to demonstrate that the endogenous opioid system may play a role in iron metabolism and subsequently in the pathogenesis of anemia. It is also the first study showing that opioid receptors are involved in the production of motor restlessness with a circadian predominance. Our findings support the role of endogenous opioids in the pathogenesis of RLS, and the triple KO mice can be used to understand the relationship between iron deficiency, anemia, dopaminergic dysfunction, and RLS.


Assuntos
Deficiências de Ferro , Ferro/metabolismo , Receptores Opioides mu/fisiologia , Analgésicos Opioides/uso terapêutico , Anemia/metabolismo , Anemia Ferropriva/metabolismo , Animais , Dopamina/metabolismo , Dopamina/fisiologia , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Naloxona/uso terapêutico , Peptídeos Opioides/uso terapêutico , Agitação Psicomotora/tratamento farmacológico , Receptores Opioides mu/genética , Síndrome das Pernas Inquietas/metabolismo , Síndrome das Pernas Inquietas/fisiopatologia
5.
Vitam Horm ; 111: 227-246, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31421702

RESUMO

The organization of estrogenic signaling in the CNS is exceedingly complex. It is comprised of peripherally and centrally synthesized estrogens, and a plethora of types of estrogen receptor that can localize to both the nucleus and the plasma membrane. Moreover, CNS estrogen receptors can exist independent of aromatase (aka estrogen synthase) as well as oligomerize with it, along with a host of other membrane signaling proteins. This ability of CNS estrogen receptors to either to physically pair or exist separately enables locally produced estrogens to act on multiple spatial levels, with a high degree of gradated regulation and plasticity, signaling either in-phase or out-of phase with circulating estrogens. This complexity explains the numerous contradictory findings regarding sex-dependent pain processing and sexually dimorphic opioid antinociception. This review highlights the increasing awareness that estrogens are major endogenous arbiters of both opioid analgesic actions and the mechanisms used to achieve them. This behooves us to understand, and possibly intercede at, the points of intersection of estrogenic signaling and opioid functionality. Factors that integrate estrogenic actions at subcellular, synaptic, and CNS regional levels are likely to be prime drug targets for novel pharmacotherapies designed to modulate CNS estrogen-dependent opioid functionalities and possibly circumvent the current opioid epidemic.


Assuntos
Analgésicos Opioides/farmacologia , Estrogênios/fisiologia , Reprodução/fisiologia , Caracteres Sexuais , Analgesia , Animais , Aromatase , Encéfalo/fisiologia , Dinorfinas/fisiologia , Feminino , Humanos , Masculino , Sistemas Neurossecretores/fisiologia , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Receptores de Estrogênio/fisiologia , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/fisiologia , Transdução de Sinais/fisiologia
6.
Br J Anaesth ; 122(6): e157-e167, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30915986

RESUMO

BACKGROUND: Opioid receptors are implicated in cancer progression and long-term patient outcomes. However, the prognostic significance, underlying mechanisms, and therapeutic value of mu-opioid receptor (MOP) in hepatocellular carcinoma (HCC) remain unclear. METHODS: MOP expression in human biopsy HCC samples was evaluated using RNA microarrays, quantitative real-time polymerase chain reaction (qRT-PCR), and immunochemical analyses. Molecular and cellular techniques, including siRNA-mediated depletion and lentiviral vector-mediated overexpression, were used to elucidate the functions and mechanisms of MOP. The effect of the MOP agonist morphine in HCC was evaluated both in vitro and in vivo. The therapeutic value of MOP inhibitors in HCC progression and metastasis was investigated with in vitro experiments and subcutaneous and orthotopic HCC mouse models in vivo. RESULTS: Through microarray analysis and qRT-PCR, we identified that MOP is highly expressed in human HCC tumours. High MOP expression in HCC tumours was confirmed by immunocytochemistry and correlated with aggressive clinicopathological features and a worse prognosis. Depletion of MOP suppressed cell proliferation, migration, and invasion, whereas overexpression of MOP promoted cell growth and metastasis in human HCC cell lines. Both clinical and biological evidence revealed that MOP-mediated epithelial-mesenchymal transition promotes HCC metastasis and poor prognosis. Morphine promotes cell proliferation, migration, and invasion in vitro and in vivo in mouse models. More importantly, MOP inhibitors suppressed cell growth, invasion, and metastasis in vitro and in the subcutaneous and orthotopic xenograft models. CONCLUSIONS: MOP plays a key oncogenic function in hepatocarcinogenesis. Its overexpression is associated with poor prognosis in patients with HCC. Furthermore, MOP inhibitors may be a promising strategy for HCC therapy.


Assuntos
Biomarcadores Tumorais/biossíntese , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores Opioides mu/biossíntese , Adolescente , Adulto , Idoso , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Animais , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/secundário , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Progressão da Doença , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Terapia de Alvo Molecular/métodos , Morfina/efeitos adversos , Morfina/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Invasividade Neoplásica , Prognóstico , RNA Mensageiro/genética , RNA Neoplásico/genética , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/genética , Receptores Opioides mu/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adulto Jovem
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(9): 1066-1070, 2018 Aug 30.
Artigo em Chinês | MEDLINE | ID: mdl-30377099

RESUMO

OBJECTIVE: To observe descending inhibition of cardiac nociception induced by microinjection of endomorphin-1 (EM1) in the ventrolateral periaqueductal gray (VLPAG) in rats effect and explore the role of µ-opioid receptor in mediating this effect. METHODS: Male SD rats were randomized into electromyography (EMG) group and c-Fos group, both of which were further divided into 5 subgroups, namely 0.9% NaCl group, bradykinin (BK) group, BK+EM1 group, BK+CTOP group, and BK+CTOP+EM1 group. Rat models of cardiac nociception were established by intrapericardial injection of BK. The changes of cardiaosomatic motor reflex induced by BK were observed by assessing EMG responses of the dorsal spinotrapezius muscle; c-Fos expression in the spinal dorsal horn at levels T3-T5 was tested. RESULTS: Compared with 0.9% NaCl, intrapericardial BK injection induced obvious EMG activities and significantly increased c-Fos expression in the spinal dorsal horn at T3-T5 (P < 0.05). Compared with BK injection, microinjection of EM1 in the VLPAG dose-dependently inhibited EMG activities and significantly decreased c-Fos expression (P < 0.05); microinjection of CTOP in the VLPAG produced no significant effect on EMG or c-Fos expression (P > 0.05). Microinjection of CTOP obviously reversed EM1-induced inhibition of EMG activities and c-Fos expression (P < 0.05). CONCLUSIONS: Microinjection of EM1 in the VLPAG produces descending inhibition of cardiac nociception in rats by activating µ-opioid receptor.


Assuntos
Analgésicos Opioides/administração & dosagem , Coração/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Oligopeptídeos/administração & dosagem , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Animais , Eletromiografia , Coração/fisiologia , Masculino , Microinjeções , Nociceptividade/fisiologia , Oligopeptídeos/farmacologia , Substância Cinzenta Periaquedutal/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/fisiologia , Cloreto de Sódio/administração & dosagem , Cloreto de Sódio/farmacologia , Somatostatina/administração & dosagem , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Músculos Superficiais do Dorso/efeitos dos fármacos , Músculos Superficiais do Dorso/fisiologia
8.
J Pharmacol Sci ; 137(1): 67-75, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29716811

RESUMO

Oxytocin (OT) is a 9-amine neuropeptide that plays an essential role in mammalian labor, lactation, maternal bonding, and social affiliation. OT has been reported to exert an analgesic effect in both humans and animals, and the results of certain animal experiments have shown that the analgesic effect of OT is partially blocked by opioid receptor antagonists. To investigate the relationship between OT and µ opioid receptor (MOR), we evaluated how OT affects MOR in vitro by performing an electrical impedance-based receptor biosensor assay (CellKey™ assay), an intracellular cAMP assay, and a competitive receptor-binding analysis by using cells stably expressing human MOR and OT receptor. In both the CellKey™ assay and the intracellular cAMP assay, OT alone exerted no direct agonistic effect on human MOR, but treatment with 10-6 M OT markedly enhanced the MOR signaling induced by 10-6 M endomorphin-1, ß-endorphin, morphine, fentanyl, and DAMGO. Moreover, in the competitive receptor-binding assay, 10-6 M OT did not alter the affinity of endomorphin-1 or morphine for MOR. These results suggest that OT could function as a positive allosteric modulator that regulates the efficacy of MOR signaling, and thus OT might represent a previously unrecognized candidate analgesic agent.


Assuntos
Regulação Alostérica/efeitos dos fármacos , Neuropeptídeos/farmacologia , Ocitocina/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Analgésicos , Animais , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Ocitocina/fisiologia , Receptores Opioides mu/fisiologia , Estimulação Química
9.
Anesthesiology ; 128(6): 1207-1219, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29461271

RESUMO

BACKGROUND: Patients with early stage tongue cancer do not frequently complain of tongue pain. Endothelin-1 signaling is upregulated in the cancerous tongue at the early stage. We tested the hypothesis that endothelin-1 signaling contributes to the modulation of tongue nociception. METHODS: Squamous cell carcinoma cells were inoculated into the tongue under general anesthesia. Lingual mechanical sensitivity under light anesthesia using forceps from days 1 to 21 (n = 8) and the amounts of endothelin-1 and ß-endorphin in the tongue on days 6, 14, and 21 (n = 5 to 7) were examined after the inoculation. The effect of endothelin-A or µ-opioid receptor antagonism on the mechanical sensitivity was examined (n = 5 to 7). RESULTS: Lingual mechanical sensitivity did not change at the early stage (days 5 to 6) but increased at the late stage (days 13 to 14). The amount of endothelin-1 increased (25.4 ± 4.8 pg/ml vs. 15.0 ± 5.2 pg/ml; P = 0.008), and endothelin-A receptor antagonism in the tongue induced mechanical hypersensitivity at the early stage (51 ± 9 g vs. 81 ± 6 g; P = 0.0001). The µ-opioid receptor antagonism enhanced mechanical hypersensitivity (39 ± 7 g vs. 81 ± 6 g; P < 0.0001), and the amount of ß-endorphin increased at the early stage. CONCLUSIONS: ß-Endorphin released from the cancer cells via endothelin-1 signaling is involved in analgesic action in mechanical hypersensitivity at the early stage.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Endotelina-1/metabolismo , Nociceptividade/fisiologia , Transdução de Sinais/fisiologia , Neoplasias da Língua/metabolismo , Animais , Carcinoma de Células Escamosas/patologia , Masculino , Antagonistas de Entorpecentes/farmacologia , Estadiamento de Neoplasias/métodos , Nociceptividade/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/fisiologia , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Língua/patologia
10.
Toxins (Basel) ; 10(1)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29342943

RESUMO

The synthetic peptide PnPP-19 comprehends 19 amino acid residues and it represents part of the primary structure of the toxin δ-CNTX-Pn1c (PnTx2-6), isolated from the venom of the spider Phoneutria nigriventer. Behavioural tests suggest that PnPP-19 induces antinociception by activation of CB1, µ and δ opioid receptors. Since the peripheral and central antinociception induced by PnPP-19 involves opioid activation, the aim of this work was to identify whether this synthetic peptide could directly activate opioid receptors and investigate the subtype selectivity for µ-, δ- and/or κ-opioid receptors. Furthermore, we also studied the modulation of calcium influx driven by PnPP-19 in dorsal root ganglion neurons, and analyzed whether this modulation was opioid-mediated. PnPP-19 selectively activates µ-opioid receptors inducing indirectly inhibition of calcium channels and hereby impairing calcium influx in dorsal root ganglion (DRG) neurons. Interestingly, notwithstanding the activation of opioid receptors, PnPP-19 does not induce ß-arrestin2 recruitment. PnPP-19 is the first spider toxin derivative that, among opioid receptors, selectively activates µ-opioid receptors. The lack of ß-arrestin2 recruitment highlights its potential for the design of new improved opioid agonists.


Assuntos
Canais de Cálcio/fisiologia , Peptídeos/farmacologia , Receptores Opioides mu/fisiologia , Venenos de Aranha/farmacologia , Animais , Gânglios Espinais/fisiologia , Células HEK293 , Humanos , Neurônios/fisiologia , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Ratos Wistar , Xenopus laevis
11.
Anesthesiology ; 128(5): 967-983, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29334500

RESUMO

BACKGROUND: The current study used recombinant herpes simplex virus type I to increase expression of µ-opiate receptors and the opioid ligand preproenkephalin in peripheral nerve fibers in a mouse model of neuropathic pain. It was predicted that viral vector delivery of a combination of genes encoding the µ-opioid receptor and preproenkephalin would attenuate neuropathic pain and enhance opioid analgesia. The behavioral effects would be paralleled by changes in response properties of primary afferent neurons. METHODS: Recombinant herpes simplex virus type 1 containing cDNA sequences of the µ-opioid receptor, human preproenkephalin, a combination, or Escherichia coli lacZ gene marker (as a control) was used to investigate the role of peripheral opioids in neuropathic pain behaviors. RESULTS: Inoculation with the µ-opioid receptor viral vector (n = 13) reversed mechanical allodynia and thermal hyperalgesia and produced leftward shifts in loperamide (ED50 = 0.6 ± 0.2 mg/kg vs. ED50 = 0.9 ± 0.2 mg/kg for control group, n = 8, means ± SD) and morphine dose-response curves (ED50 = 0.3 ± 0.5 mg/kg vs. ED50 = 1.1 ± 0.1 mg/kg for control group). In µ-opioid receptor viral vector inoculated C-fibers, heat-evoked responses (n = 12) and ongoing spontaneous activity (n = 18) were decreased after morphine application. Inoculation with both µ-opioid receptor and preproenkephalin viral vectors did not alter mechanical and thermal responses. CONCLUSIONS: Increasing primary afferent expression of opioid receptors can decrease neuropathic pain-associated behaviors and increase systemic opioid analgesia through inhibition of peripheral afferent fiber activity.


Assuntos
Analgésicos Opioides/farmacologia , Encefalinas/fisiologia , Neuralgia/prevenção & controle , Neurônios Aferentes/fisiologia , Receptores Opioides mu/fisiologia , Analgesia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Herpesvirus Humano 1/genética , Masculino , Camundongos , Morfina/farmacologia , Proteínas Proto-Oncogênicas c-fos/análise , Receptores Opioides mu/análise
12.
Mol Pharmacol ; 93(2): 36-48, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29237725

RESUMO

The µ-opioid receptor (MOR) is a Gi/o protein-coupled receptor that mediates analgesic, euphoric, and reward effects. Using a bacterial two-hybrid screen, we reported that the carboxyl tail of the rat MOR associates with A20-binding inhibitor of nuclear factor κB (ABIN-1). This interaction was confirmed by direct protein-protein binding and coimmunoprecipitation of MOR and ABIN-1 proteins in cell lysates. Saturation binding studies showed that ABIN-1 had no effect on MOR binding. However, the interaction of ABIN-1 and MOR inhibited the activation of G proteins induced by DAMGO ([d-Ala2,N-Me-Phe4,Gly5-ol]-Enkephalin). MOR phosphorylation, ubiquitination, and internalization induced by DAMGO were decreased in Chinese hamster ovary cells that coexpressed MOR and ABIN-1. The suppression of forskolin-stimulated adenylyl cyclase by DAMGO was also inhibited by the interaction of ABIN-1 with MOR. In addition, extracellular signal-regulated kinase activation was also negatively regulated by overexpression of ABIN-1. These data suggest that ABIN-1 is a negative coregulator of MOR activation, phosphorylation, and internalization in vitro. ABIN-1 also inhibited morphine-induced hyperlocomotion in zebrafish larvae (AB strain). By utilization of an antisense morpholino oligonucleotide (MO) gene knockdown technology, the ABIN-1 MO-injected zebrafish larvae showed a significant increase (approximately 60%) in distance moved compared with control MO-injected larvae after acute morphine treatment (P < 0.01). Taken together, ABIN-1 negatively regulates MOR function in vitro and in vivo.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Receptores Opioides mu/fisiologia , Analgésicos Opioides/farmacologia , Animais , Sítios de Ligação , Células CHO , Linhagem Celular Tumoral , Cricetulus , AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endocitose , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Larva , Ligantes , Proteínas Luminescentes , Fosforilação , Ratos , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Transdução de Sinais , Radioisótopos de Enxofre/metabolismo , Ubiquitinação , Peixe-Zebra/crescimento & desenvolvimento
14.
Alcohol Clin Exp Res ; 40(10): 2114-2123, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27508965

RESUMO

BACKGROUND: The nucleus accumbens shell is a key brain area mediating the reinforcing effects of ethanol (EtOH). Previously, it has been shown that the density of µ-opioid receptors in the nucleus accumbens shell is higher in alcohol-preferring Alko Alcohol (AA) rats than in alcohol-avoiding Alko Non-Alcohol rats. In addition, EtOH releases opioid peptides in the nucleus accumbens and opioid receptor antagonists are able to modify EtOH intake, all suggesting an opioidergic mechanism in the control of EtOH consumption. As the exact mechanisms of opioidergic involvement remains to be elucidated, the aim of this study was to clarify the role of accumbal µ- and κ-opioid receptors in controlling EtOH intake in alcohol-preferring AA rats. METHODS: Microinfusions of the µ-opioid receptor antagonist CTOP (0.3 and 1 µg/site), µ-opioid receptor agonist DAMGO (0.03 and 0.1 µg/site), nonselective opioid receptor agonist morphine (30 µg/site), and κ-opioid receptor agonist U50488H (0.3 and 1 µg/site) were administered via bilateral guide cannulas into the nucleus accumbens shell of AA rats that voluntarily consumed 10% EtOH solution in an intermittent, time-restricted (90-minute) 2-bottle choice access paradigm. RESULTS: CTOP (1 µg/site) significantly increased EtOH intake. Conversely, DAMGO resulted in a decreasing trend in EtOH intake. Neither morphine nor U50488H had any effect on EtOH intake in the used paradigm. CONCLUSIONS: The results provide further evidence for the role of accumbens shell µ-opioid receptors but not κ-opioid receptors in mediating reinforcing effects of EtOH and in regulating EtOH consumption. The results also provide support for views suggesting that the nucleus accumbens shell has a major role in mediating EtOH reward.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Etanol/administração & dosagem , Núcleo Accumbens/fisiologia , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/fisiologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Masculino , Microinjeções , Morfina/administração & dosagem , Morfina/farmacologia , Ratos , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Recompensa , Somatostatina/administração & dosagem , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Especificidade da Espécie
15.
Behav Brain Res ; 312: 163-8, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27312267

RESUMO

Pentazocine, a mixed-action kappa opioid receptor (KOR) agonist, has high affinity for both KOR and the mu opioid receptor (MOR), and has been shown clinically to alleviate pain with a pronounced effect in women. However, whether local application of pentazocine in the spinal cord produces antinociception and the contribution of spinal KOR and MOR in mediating the effect of pentazocine in female rats remain unknown. Also, it is not known whether pentazocine-induced antinociception in females is estrogen-dependent. Hence, we investigated whether intrathecal (i.t.) (-)-pentazocine produces thermal antinociception and whether estrogen modulates the drug effect in female rats. Only the highest dose of pentazocine (500 nmol) was effective in producing antinociception in ovariectomized (OVX) rats. In contrast, pentazocine produced antinociception in estradiol-treated ovariectomized females (OVX+E) rats with the lowest effective dose being 250nmol. KOR or MOR mediated the effect of the lowest effective dose in OVX+E rats; however, MOR blockade extended the KOR-mediated effect of 500nmol pentazocine in both groups. In normally cycling females, the 250nmol dose was effective in producing antinociception at the proestrous, but not at the diestrous stage of the estrous cycle. Thus, estrogen facilitates and KOR or MOR mediates. the antinociceptive effect of i.t. (-)-pentazocine in female rats. Selective doses of (-)-pentazocine, with or without MOR blockade, may have a therapeutic benefit.


Assuntos
Analgésicos/administração & dosagem , Estradiol/administração & dosagem , Antagonistas de Estrogênios/administração & dosagem , Nociceptividade/efeitos dos fármacos , Pentazocina/administração & dosagem , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/fisiologia , Animais , Ciclo Estral , Feminino , Temperatura Alta , Injeções Espinhais , Ovariectomia , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Medula Espinal/efeitos dos fármacos
16.
eNeuro ; 3(1)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27022625

RESUMO

Touch plays a significant role in human social behavior and social communication, and its rewarding nature has been suggested to involve opioids. Opioid blockade in monkeys leads to increased solicitation and receipt of grooming, suggesting heightened enjoyment of touch. We sought to study the role of endogenous opioids in perception of affective touch in healthy adults and in patients with fibromyalgia, a chronic pain condition shown to involve reduced opioid receptor availability. The pleasantness of touch has been linked to the activation of C-tactile fibers, which respond maximally to slow gentle touch and correlate with ratings of pleasantness. We administered naloxone to patients and healthy controls to directly observe the consequences of µ-opioid blockade on the perceived pleasantness and intensity of touch. We found that at baseline chronic pain patients showed a blunted distinction between slow and fast brushing for both intensity and pleasantness, suggesting reduced C-tactile touch processing. In addition, we found a differential effect of opioid blockade on touch perception in healthy subjects and pain patients. In healthy individuals, opioid blockade showed a trend toward increased ratings of touch pleasantness, while in chronic pain patients it significantly decreased ratings of touch intensity. Further, in healthy individuals, naloxone-induced increase in touch pleasantness was associated with naloxone-induced decreased preference for slow touch, suggesting a possible effect of opioid levels on processing of C-tactile fiber input. These findings suggest a role for endogenous opioids in touch processing, and provide further evidence for altered opioid functioning in chronic pain patients.


Assuntos
Dor Crônica/fisiopatologia , Fibromialgia/fisiopatologia , Receptores Opioides mu/fisiologia , Percepção do Tato/fisiologia , Adolescente , Adulto , Dor Crônica/psicologia , Feminino , Fibromialgia/psicologia , Humanos , Masculino , Pessoa de Meia-Idade , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Estimulação Física , Receptores Opioides mu/antagonistas & inibidores , Percepção do Tato/efeitos dos fármacos , Adulto Jovem
17.
J Oral Facial Pain Headache ; 30(1): 61-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26817034

RESUMO

AIMS: To investigate whether the protective effect of testosterone on the development of temporomandibular joint (TMJ) nociception in male rats is mediated by the activation of central opioid mechanisms. METHODS: Experiments were performed on 156 male Wistar rats. A pharmacologic approach was used to assess the ability of opioid receptor antagonists infused into the dorsal portion of the brainstem and adjacent to the caudal component (subnucleus caudalis) of the spinal trigeminal nucleus to block the protective effect of testosterone in male rats. The TMJ injection of 0.5% formalin was used as a nociceptive stimulus. One-way or two-way ANOVA was used for data analyses. RESULTS: The injection of 0.5% formalin into the TMJ induced a significant nociceptive behavior in gonadectomized male rats (P < .05), but not in naïve, sham, and testosterone-replaced gonadectomized rats, confirming that testosterone prevents the development of TMJ nociception. The injection of either the nonselective opioid receptor antagonist naloxone (15 µg) or the simultaneous injection of the µ-opioid receptor antagonist Cys2, Tyr3, Orn5, Pen7amide (CTOP, 30 µg) and the κ-opioid receptor antagonist Nor-Binaltorphimine (Nor-BNI, 90 µg) significantly increased the 0.5% formalin-induced behavioral response in sham and testosterone-replaced gonadectomized rats (P < .05) but had no effect in gonadectomized rats. However, the injection of each selective opioid receptor antagonist alone or the simultaneous injection of µ- or κ- and δ-opioid receptor antagonists had no effect. CONCLUSION: These findings indicate that the protective effect of endogenous testosterone on the development of TMJ nociception in male rats is mediated by the activation of central opioid mechanisms. Furthermore, the coactivation of central µ- and κ-opioid receptors is necessary for testosterone to protect male rats from developing TMJ nociception.


Assuntos
Nociceptividade/fisiologia , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/fisiologia , Articulação Temporomandibular/fisiologia , Testosterona/fisiologia , Animais , Tronco Encefálico/efeitos dos fármacos , Dor Facial/prevenção & controle , Formaldeído/efeitos adversos , Masculino , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Nociceptividade/efeitos dos fármacos , Dor Nociceptiva/induzido quimicamente , Orquiectomia , Ratos , Ratos Wistar , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides mu/antagonistas & inibidores , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Transtornos da Articulação Temporomandibular/prevenção & controle , Testosterona/farmacologia , Núcleo Inferior Caudal do Nervo Trigêmeo/efeitos dos fármacos
18.
Mol Psychiatry ; 21(8): 1057-62, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26460230

RESUMO

Positron emission tomography (PET) studies suggest opioidergic system dysfunction in morbid obesity, while evidence for the role of the dopaminergic system is less consistent. Whether opioid dysfunction represents a state or trait in obesity remains unresolved, but could be assessed in obese subjects undergoing weight loss. Here we measured brain µ-opioid receptor (MOR) and dopamine D2 receptor (D2R) availability in 16 morbidly obese women twice-before and 6 months after bariatric surgery-using PET with [(11)C]carfentanil and [(11)C]raclopride. Data were compared with those from 14 lean control subjects. Receptor-binding potentials (BPND) were compared between the groups and between the pre- and postoperative scans among the obese subjects. Brain MOR availability was initially lower among obese subjects, but weight loss (mean=26.1 kg, s.d.=7.6 kg) reversed this and resulted in ~23% higher MOR availability in the postoperative versus preoperative scan. Changes were observed in areas implicated in reward processing, including ventral striatum, insula, amygdala and thalamus (P's<0.005). Weight loss did not influence D2R availability in any brain region. Taken together, the endogenous opioid system plays an important role in the pathophysiology of human obesity. Because bariatric surgery and concomitant weight loss recover downregulated MOR availability, lowered MOR availability is associated with an obese phenotype and may mediate excessive energy uptake. Our results highlight that understanding the opioidergic contribution to overeating is critical for developing new treatments for obesity.


Assuntos
Obesidade Mórbida/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores Opioides mu/metabolismo , Adulto , Cirurgia Bariátrica , Encéfalo/metabolismo , Dopamina/metabolismo , Feminino , Fentanila/análogos & derivados , Humanos , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons/métodos , Receptores de Dopamina D2/fisiologia , Receptores Opioides/metabolismo , Receptores Opioides/fisiologia , Receptores Opioides mu/fisiologia , Redução de Peso
19.
Cancer ; 121(16): 2681-8, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26043235

RESUMO

Mu opioids are among the most widely used drugs for patients with cancer with both acute and chronic pain as well as in the perioperative period. Several retrospective studies have suggested that opioid use might promote tumor progression and as a result negatively impact survival in patients with advanced cancer; however, in the absence of appropriate prospective validation, any changes in recommendations for opioid use are not warranted. In this review, the authors present preclinical and clinical data that support their hypothesis that the mu opioid receptor is a potential target for cancer therapy because of its plausible role in tumor progression. The authors also propose the hypothesis that peripheral opioid antagonists such as methylnaltrexone, which reverses the peripheral effects of mu opioids but maintains centrally mediated analgesia and is approved by the US Food and Drug Administration for the treatment of opioid-induced constipation, can be used to target the mu opioid receptor.


Assuntos
Antagonistas de Entorpecentes/uso terapêutico , Neoplasias/tratamento farmacológico , Receptores Opioides mu/fisiologia , Animais , Humanos , Camundongos , Terapia de Alvo Molecular , Naltrexona/análogos & derivados , Naltrexona/uso terapêutico , Compostos de Amônio Quaternário/uso terapêutico , Receptores Opioides mu/genética
20.
J Neurosci ; 35(19): 7317-25, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25972162

RESUMO

The amygdala is a key region for the processing of information underlying fear, anxiety, and fear extinction. Within the local neuronal networks of the amygdala, a population of inhibitory, intercalated neurons (ITCs) modulates the flow of information among various nuclei of amygdala, including the basal nucleus (BA) and the centromedial nucleus (CeM) of the amygdala. These ITCs have been shown to be important during fear extinction and are target of a variety of neurotransmitters and neuropeptides. Here we provide evidence that the activation of µ-opioid receptors (MORs) by the specific agonist DAMGO ([D-Ala2,N-Me-Phe4,Gly5-ol]-Enkephalin) hyperpolarizes medially located ITCs (mITCs) in acute brain slices of mice. Moreover, we use whole-cell patch-clamp recordings in combination with local electrical stimulation or glutamate uncaging to analyze the effect of MOR activation on local microcircuits. We show that the GABAergic transmission between mITCs and CeM neurons is attenuated by DAMGO, whereas the glutamatergic transmission on CeM neurons and mITCs is unaffected. Furthermore, MOR activation induced by theta burst stimulation in BA suppresses plastic changes of feedforward inhibitory transmission onto CeM neurons as revealed by the MOR antagonist CTAP d-Phe-Cys-Tyr-d-Trp-Arg-Thr-Pen-Thr-NH2. In summary, the mITCs constitute a target for the opioid system, and therefore, the activation of MOR in ITCs might play a central role in the modulation of the information processing between the basolateral complex of the amygdala and central nuclei of the amygdala.


Assuntos
Núcleo Central da Amígdala/citologia , Inibição Neural/fisiologia , Neurônios/fisiologia , Receptores Opioides mu/fisiologia , Transmissão Sináptica/fisiologia , Analgésicos Opioides/farmacologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Antagonistas de Entorpecentes/farmacologia , Inibição Neural/efeitos dos fármacos , Inibição Neural/genética , Neurônios/efeitos dos fármacos , Peptídeos/farmacologia , Quinoxalinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Tetrodotoxina/farmacologia , Valina/análogos & derivados , Valina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA