Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 180(24): 3160-3174, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37489013

RESUMO

BACKGROUND AND PURPOSE: Opioid-induced respiratory depression limits the use of µ-opioid receptor agonists in clinical settings and is the main cause of opioid overdose fatalities. The relative potential of different opioid agonists to induce respiratory depression at doses exceeding those producing analgesia is understudied despite its relevance to assessments of opioid safety. Here we evaluated the respiratory depressant and anti-nociceptive effects of three novel opioids and relate these measurements to their in vitro efficacy. EXPERIMENTAL APPROACH: Respiration was measured in awake, freely moving male CD-1 mice using whole body plethysmography. Anti-nociception was measured using the hot plate test. Morphine, oliceridine and tianeptine were administered intraperitoneally, whereas methadone, oxycodone and SR-17018 were administered orally. Receptor activation and arrestin-3 recruitment were measured in HEK293 cells using BRET assays. KEY RESULTS: Across the dose ranges examined, all opioids studied depressed respiration in a dose-dependent manner, with similar effects at the highest doses, and with tianeptine and oliceridine showing reduced duration of effect, when compared with morphine, oxycodone, methadone and SR-17018. When administered at doses that induced similar respiratory depression, all opioids induced similar anti-nociception, with tianeptine and oliceridine again showing reduced duration of effect. These data were consistent with the in vitro agonist activity of the tested compounds. CONCLUSION AND IMPLICATIONS: In addition to providing effective anti-nociception, the novel opioids, oliceridine, tianeptine and SR-17018 depress respiration in male mice. However, the different potencies and kinetics of effect between these novel opioids may be relevant to their therapeutic application in different clinical settings.


Assuntos
Analgésicos Opioides , Insuficiência Respiratória , Masculino , Humanos , Animais , Camundongos , Oxicodona/farmacologia , Células HEK293 , Morfina/farmacologia , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/tratamento farmacológico , Metadona/efeitos adversos
2.
Br J Pharmacol ; 180(7): 943-957, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-33245558

RESUMO

BACKGROUND AND PURPOSE: G protein-biased µ opioid receptor agonists have the potential to induce less receptor desensitisation and tolerance than balanced opioids. Here, we investigated if the cyclic endomorphin analogue Tyr-c[D-Lys-Phe-Tyr-Gly] (Compound 1) is a G protein-biased µ agonist and characterised its ability to induce rapid receptor desensitisation in mammalian neurones. EXPERIMENTAL APPROACH: The signalling and trafficking properties of opioids were characterised using bioluminescence resonance energy transfer assays, enzyme-linked immunosorbent assay and phosphosite-specific immunoblotting in human embryonic kidney 293 cells. Desensitisation of opioid-induced currents were studied in rat locus coeruleus neurones using whole-cell patch-clamp electrophysiology. The mechanism of Compound 1-induced µ receptor desensitisation was probed using kinase inhibitors. KEY RESULTS: Compound 1 has similar intrinsic activity for G protein signalling as morphine. As predicted for a G protein-biased µ agonist, Compound 1 induced minimal agonist-induced internalisation and phosphorylation at intracellular µ receptor serine/threonine residues known to be involved in G protein-coupled receptor kinase (GRK)-mediated desensitisation. However, Compound 1 induced robust rapid µ receptor desensitisation in locus coeruleus neurons, to a greater degree than morphine. The extent of Compound 1-induced desensitisation was unaffected by activation or inhibition of protein kinase C (PKC) but was significantly reduced by inhibition of GRK. CONCLUSION AND IMPLICATIONS: Compound 1 is a novel G protein-biased µ agonist that induces substantial rapid receptor desensitisation in mammalian neurons. Surprisingly, Compound 1-induced desensitisation was demonstrated to be GRK dependent despite its G protein bias. Our findings refute the assumption that G protein-biased agonists will evade receptor desensitisation and tolerance. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.


Assuntos
Analgésicos Opioides , Receptores Opioides mu , Ratos , Humanos , Animais , Analgésicos Opioides/farmacologia , Receptores Opioides mu/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Morfina/farmacologia , Transdução de Sinais , Quinases de Receptores Acoplados a Proteína G/metabolismo , Mamíferos/metabolismo
3.
Adv Drug Alcohol Res ; 12022 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-35909497

RESUMO

Background: Opioid users regularly consume other drugs such as alcohol (ethanol). Acute administration of ethanol rapidly reverses tolerance to morphine-induced respiratory depression. However, recent research has suggested that the primary metabolite of ethanol, acetaldehyde, may play a key role in mediating the CNS effects seen after ethanol consumption. This research investigated the role of acetaldehyde in ethanol reversal of tolerance to morphine-induced respiratory depression. Methods: Tolerance was induced in mice by 6-days implantation of a 75 mg morphine pellet with control mice implanted with a placebo pellet. Tolerance was assessed by acute morphine administration on day 6 and respiration measured by plethysmography. Levels of acetaldehyde were inhibited or enhanced by pre-treatments with the acetaldehyde chelator D-penicillamine and the inhibitor of acetaldehyde dehydrogenase disulfiram respectively. Results: Morphine pellet implanted mice displayed tolerance to an acute dose of morphine compared to placebo pellet implanted controls. Acute acetaldehyde administration dose-dependently reversed tolerance to morphine respiratory depression. As previously demonstrated, ethanol reversed morphine tolerance, and this was inhibited by D-penicillamine pre-treatment. An acute, low dose of ethanol that did not significantly reverse morphine tolerance was able to do so following disulfiram pre-treatment. Conclusion: These data suggest that acetaldehyde, the primary metabolite of ethanol, is responsible for the reversal of morphine tolerance observed following ethanol administration.

4.
Br J Pharmacol ; 179(14): 3875-3885, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35297034

RESUMO

BACKGROUND AND PURPOSE: Mitragynine, the major alkaloid in Mitragyna speciosa (kratom), is a partial agonist at the µ opioid receptor. CYP3A-dependent oxidation of mitragynine yields the metabolite 7-OH mitragynine, a more efficacious µ receptor agonist. While both mitragynine and 7-OH mitragynine can induce anti-nociception in mice, recent evidence suggests that 7-OH mitragynine formed as a metabolite is sufficient to explain the anti-nociceptive effects of mitragynine. However, the ability of 7-OH mitragynine to induce µ receptor-dependent respiratory depression has not yet been studied. EXPERIMENTAL APPROACH: Respiration was measured in awake, freely moving, male CD-1 mice, using whole body plethysmography. Anti-nociception was measured using the hot plate assay. Morphine, mitragynine, 7-OH mitragynine and the CYP3A inhibitor ketoconazole were administered orally. KEY RESULTS: The respiratory depressant effects of mitragynine showed a ceiling effect, whereby doses higher than 10 mg·kg-1 produced the same level of effect. In contrast, 7-OH mitragynine induced a dose-dependent effect on mouse respiration. At equi-depressant doses, both mitragynine and 7-OH mitragynine induced prolonged anti-nociception. Inhibition of CYP3A reduced mitragynine-induced respiratory depression and anti-nociception without affecting the effects of 7-OH mitragynine. CONCLUSIONS AND IMPLICATIONS: Both the anti-nociceptive effects and the respiratory depressant effects of mitragynine are partly due to its metabolic conversion to 7-OH mitragynine. The limiting rate of conversion of mitragynine into its active metabolite results in a built-in ceiling effect of the mitragynine-induced respiratory depression. These data suggest that such 'metabolic saturation' at high doses may underlie the improved safety profile of mitragynine as an opioid analgesic.


Assuntos
Mitragyna , Insuficiência Respiratória , Alcaloides de Triptamina e Secologanina , Animais , Citocromo P-450 CYP3A , Masculino , Camundongos , Receptores Opioides mu/agonistas , Alcaloides de Triptamina e Secologanina/farmacologia
5.
Pharmacol Ther ; 230: 107961, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34256067

RESUMO

Morphine and other mu-opioid receptor (MOR) agonists remain the mainstay treatment of acute and prolonged pain states worldwide. The major limiting factor for continued use of these current opioids is the high incidence of side effects that result in loss of life and loss of quality of life. The development of novel opioids bereft, or much less potent, at inducing these side effects remains an intensive area of research, with multiple pharmacological strategies being explored. However, as with many G protein-coupled receptors (GPCRs), translation of promising candidates from in vitro characterisation to successful clinical candidates still represents a major challenge and attrition point. This review summarises the preclinical animal models used to evaluate the key opioid-induced behaviours of antinociception, respiratory depression, constipation and opioid-induced hyperalgesia and tolerance. We highlight the influence of distinct variables in the experimental protocols, as well as the potential implications for differences in receptor reserve in each system. Finally, we discuss how methods to assess opioid action in vivo and in vitro relate to each other in the context of bridging the translational gap in opioid drug discovery.


Assuntos
Analgésicos Opioides , Qualidade de Vida , Analgésicos Opioides/efeitos adversos , Animais , Humanos , Morfina/efeitos adversos , Dor/tratamento farmacológico , Receptores Opioides mu/agonistas
6.
Br J Pharmacol ; 177(13): 2923-2931, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32052419

RESUMO

BACKGROUND AND PURPOSE: GPCRs can signal through both G proteins and ß-arrestin2. For the µ-opioid receptor, early experimental evidence from a single study suggested that G protein signalling mediates analgesia, whereas ß-arrestin2 signalling mediates respiratory depression and constipation. Consequently, for more than a decade, much research effort has been focused on developing biased µ-opioid agonists that preferentially target G protein signalling over ß-arrestin signalling, as it was believed that such drugs would be analgesics devoid of respiratory depressant activity. However, the prototypical compounds that have been developed based on this concept have so far failed in clinical and preclinical development. EXPERIMENTAL APPROACH: The present study was set up to re-examine opioid-induced respiratory depression in ß-arrestin2 knockout mice. To this end, a consortium was formed consisting of three different laboratories located in different countries to evaluate independently opioid-induced respiratory depression. KEY RESULTS: Our consensus results unequivocally demonstrate that the prototypical µ-opioid agonist morphine (3.75-100 mg·kg-1 s.c. or 3-30 mg·kg-1 i.p.) as well as the potent opioid fentanyl (0.05-0.35 mg·kg-1 s.c.) do indeed induce respiratory depression and constipation in ß-arrestin2 knockout mice in a dose-dependent manner indistinguishable from that observed in wild-type mice. CONCLUSION AND IMPLICATIONS: Our findings do not support the original suggestion that ß-arrestin2 signalling plays a key role in opioid-induced respiratory depression and call into question the concept of developing G protein-biased µ-opioid receptor agonists as a strategy for the development of safer opioid analgesic drugs.


Assuntos
Morfina , Insuficiência Respiratória , Analgésicos Opioides/toxicidade , Animais , Fentanila , Camundongos , Morfina/farmacologia , Receptores Opioides mu/metabolismo , Insuficiência Respiratória/induzido quimicamente , beta-Arrestina 2/metabolismo
7.
Platelets ; 31(5): 641-645, 2020 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-31684789

RESUMO

The principal demonstrated role of the nonvisual arrestins in vivo is to limit G protein-coupled receptor (GPCR) signaling. Nonetheless, a direct demonstration of this fundamental ability in platelets remains lacking, despite the prominent role played by GPCRs in platelet activation. This paper describes the basic characterization of the activatory responses of platelets from mice lacking arrestin-3 (arr3-/-), revealing pleiotropic roles dependent on GPCR ligand. Functionally, arrestin-3 acts as a brake on platelet aggregation regardless of ligand tested. Downstream of P2Y receptors, arr3-/- mice show increased secretion and integrin activation mirrored by enhanced intracellular calcium signaling and global PKC-dependent phosphorylation. Furthermore, P2Y12 receptor (P2Y12R) activity as assessed by ADP-mediated reduction of VASP phosphorylation is enhanced in arr3-/-mice. Downstream of PAR receptors there are similar increases in secretion and integrin activation in arr3-/- mice, together with enhanced PKC activity. Last, in arr3-/- mice the TP receptor displays unaltered PKC activity but markedly reduced calcium responses, which together with the kinetics of the aggregation response suggested a unique positive regulatory role for arrestin-3 in TP signaling. Overall, this paper reveals pleiotropic roles for arrestin-3 dependent on GPCR ligand describing for the first time a negative regulatory function for arrestin-3 in platelets.


Assuntos
Arrestinas/metabolismo , Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Plaquetas/citologia , Humanos , Camundongos
8.
J Pharmacol Exp Ther ; 372(2): 224-236, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31594792

RESUMO

Agonists at the δ opioid receptor are known to be potent antihyperalgesics in chronic pain models and effective in models of anxiety and depression. However, some δ opioid agonists have proconvulsant properties while tolerance to the therapeutic effects can develop. Previous evidence indicates that different agonists acting at the δ opioid receptor differentially engage signaling and regulatory pathways with significant effects on behavioral outcomes. As such, interest is now growing in the development of biased agonists as a potential means to target specific signaling pathways and potentially improve the therapeutic profile of δ opioid agonists. Here, we report on PN6047 (3-[[4-(dimethylcarbamoyl)phenyl]-[1-(thiazol-5-ylmethyl)-4-piperidylidene]methyl]benzamide), a novel G protein-biased and selective δ opioid agonist. In cell-based assays, PN6047 fully engages G protein signaling but is a partial agonist in both the arrestin recruitment and internalization assays. PN6047 is effective in rodent models of chronic pain but shows no detectable analgesic tolerance following prolonged treatment. In addition, PN6047 exhibited antidepressant-like activity in the forced swim test, and importantly, the drug had no effect on chemically induced seizures. PN6047 did not exhibit reward-like properties in the conditioned place preference test or induce respiratory depression. Thus, δ opioid ligands with limited arrestin signaling such as PN6047 may be therapeutically beneficial in the treatment of chronic pain states. SIGNIFICANCE STATEMENT: PN6047 (3-[[4-(dimethylcarbamoyl)phenyl]-[1-(thiazol-5-ylmethyl)-4-piperidylidene]methyl]benzamide) is a selective, G protein-biased δ opioid agonist with efficacy in preclinical models of chronic pain. No analgesic tolerance was observed after prolonged treatment, and PN6047 does not display proconvulsant activity or other opioid-mediated adverse effects. Our data suggest that δ opioid ligands with limited arrestin signaling will be beneficial in the treatment of chronic pain.


Assuntos
Analgésicos Opioides/metabolismo , Antidepressivos/química , Benzamidas/química , Benzamidas/farmacocinética , Dor Crônica/tratamento farmacológico , Proteínas de Ligação ao GTP/metabolismo , Receptores Opioides delta/metabolismo , Animais , Antidepressivos/administração & dosagem , Antidepressivos/efeitos adversos , Antidepressivos/farmacocinética , Arrestina/metabolismo , Benzamidas/administração & dosagem , Benzamidas/efeitos adversos , Tolerância a Medicamentos , Células HEK293 , Humanos , Masculino , Camundongos , Modelos Animais , Ratos Wistar , Resultado do Tratamento
9.
Br J Pharmacol ; 177(2): 254-266, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31499594

RESUMO

BACKGROUND AND PURPOSE: Fentanyl overdose deaths have reached "epidemic" levels in North America. Death in opioid overdose invariably results from respiratory depression. In the present work, we have characterized how fentanyl depresses respiration, and by comparing fentanyl with heroin and morphine, the active breakdown product of heroin, we have sought to determine the factors, in addition to high potency, that contribute to the lethality of fentanyl. EXPERIMENTAL APPROACH: Respiration (rate and tidal volume) was measured in awake, freely moving mice by whole body plethysmography. KEY RESULTS: Intravenously administered fentanyl produced more rapid depression of respiration than equipotent doses of heroin or morphine. Fentanyl depressed both respiratory rate and tidal volume. Fentanyl did not depress respiration in µ-opioid receptor knockout mice. Naloxone, the opioid antagonist widely used to treat opioid overdose, reversed the depression of respiration by morphine more readily than that by fentanyl, whereas diprenorphine, a more lipophilic antagonist, was equipotent in reversing fentanyl and morphine depression of respiration. Prolonged treatment with morphine induced tolerance to respiratory depression, but the degree of cross tolerance to fentanyl was less than the tolerance to morphine itself. CONCLUSION AND IMPLICATIONS: We propose that several factors (potency, rate of onset, lowered sensitivity to naloxone, and lowered cross tolerance to heroin) combine to make fentanyl more likely to cause opioid overdose deaths than other commonly abused opioids. Lipophilic antagonists such as diprenorphine may be better antidotes than naloxone to treat fentanyl overdose.


Assuntos
Analgésicos Opioides/toxicidade , Fentanila/toxicidade , Heroína/toxicidade , Pulmão/efeitos dos fármacos , Morfina/toxicidade , Respiração/efeitos dos fármacos , Insuficiência Respiratória/induzido quimicamente , Analgésicos Opioides/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Overdose de Drogas , Tolerância a Medicamentos , Feminino , Fentanila/administração & dosagem , Heroína/administração & dosagem , Injeções Intraperitoneais , Injeções Intravenosas , Pulmão/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Insuficiência Respiratória/tratamento farmacológico , Insuficiência Respiratória/metabolismo , Insuficiência Respiratória/fisiopatologia , Taxa Respiratória/efeitos dos fármacos , Medição de Risco , Volume de Ventilação Pulmonar/efeitos dos fármacos
10.
Drug Alcohol Depend ; 205: 107674, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31715438

RESUMO

BACKGROUND: Opioid users regularly consume other drugs such as alcohol (ethanol). Acute administration of ethanol can rapidly reverse tolerance to morphine-induced respiratory depression. However, alcohol consumption by opioid users is likely to occur over prolonged time periods. We have therefore sought to determine the effect of prolonged alcohol consumption on the development of tolerance to opioid respiratory depression. METHODS: Mice were fed control or ethanol (5%) liquid diet for 16 days. On days 9-16 morphine tolerance was induced by administration of 3 priming injections of morphine followed by subcutaneous implantation of a morphine-filled osmotic mini-pump. Control mice received saline. Respiration was measured by plethysmography and the effect of an acute morphine challenge dose was measured on day 16 to assess the development of morphine tolerance. RESULTS: Prolonged ethanol consumption for 14 days did not alter the respiratory depressant effect of an acute dose of morphine. Control mice treated with prolonged morphine developed tolerance to acute morphine respiratory depression whereas ethanol diet fed mice treated with prolonged morphine showed significant respiratory depression during morphine-pump treatment and remained sensitive to the respiratory depressant effect of the acute challenge dose of morphine. The ethanol consumption did not alter blood or brain levels of morphine, whilst conversely prolonged morphine treatment did not alter blood levels of ethanol. CONCLUSIONS: Prolonged ethanol consumption prevents the development and maintenance of tolerance to the respiratory depressant effect of morphine. These data suggest that ethanol inhibition of tolerance will greatly increase the risk of fatal heroin overdose in humans.


Assuntos
Analgésicos Opioides/administração & dosagem , Tolerância a Medicamentos , Etanol/administração & dosagem , Morfina/administração & dosagem , Insuficiência Respiratória/induzido quimicamente , Mecânica Respiratória/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Overdose de Drogas/fisiopatologia , Overdose de Drogas/prevenção & controle , Tolerância a Medicamentos/fisiologia , Masculino , Camundongos , Distribuição Aleatória , Insuficiência Respiratória/fisiopatologia , Mecânica Respiratória/fisiologia
11.
Br J Pharmacol ; 175(13): 2653-2661, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29582414

RESUMO

BACKGROUND AND PURPOSE: PZM21 is a novel µ-opioid receptor ligand that has been reported to induce minimal arrestin recruitment and be devoid of the respiratory depressant effects characteristic of classical µ receptor ligands such as morphine. We have re-examined the signalling profile of PZM21 and its ability to depress respiration. EXPERIMENTAL APPROACH: G protein (Gi ) activation and arrestin-3 translocation were measured in vitro, using BRET assays, in HEK 293 cells expressing µ receptors. Respiration (rate and tidal volume) was measured in awake, freely moving mice by whole-body plethysmography, and antinociception was measured by the hot plate test. KEY RESULTS: PZM21 (10-9 - 3 × 10-5  M) produced concentration-dependent Gi activation and arrestin-3 translocation. Comparison with responses evoked by morphine and DAMGO revealed that PZM21 was a low efficacy agonist in both signalling assays. PZM21 (10-80 mg·kg-1 ) depressed respiration in a dose-dependent manner. The respiratory depression was due to a decrease in the rate of breathing not a decrease in tidal volume. On repeated daily administration of PZM21 (twice daily doses of 40 mg·kg-1 ), complete tolerance developed to the antinociceptive effect of PZM21 over 3 days but no tolerance developed to its respiratory depressant effect. CONCLUSION AND IMPLICATIONS: These data demonstrate that PZM21 is a low efficacy µ receptor agonist for both G protein and arrestin signalling. Contrary to a previous report, PZM21 depresses respiration in a manner similar to morphine, the classical opioid receptor agonist.


Assuntos
Analgésicos Opioides/farmacologia , Depressão/tratamento farmacológico , Tolerância a Medicamentos , Receptores Opioides mu/agonistas , Tiofenos/farmacologia , Ureia/análogos & derivados , Animais , Depressão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Opioides mu/metabolismo , Ureia/farmacologia
12.
Br J Pharmacol ; 175(12): 2492-2503, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29574756

RESUMO

BACKGROUND AND PURPOSE: Oxycodone, a prescription opioid, is a major drug of abuse, especially in the USA, and contributes significantly to opioid overdose deaths each year. Overdose deaths result primarily from respiratory depression. We have studied respiratory depression by oxycodone and have characterized how tolerance develops on prolonged exposure to the drug. We have investigated the role of PKC in maintaining tolerance and have examined whether ethanol or pregabalin reverses oxycodone-induced tolerance. EXPERIMENTAL APPROACH: Respiration was measured in male CD-1 mice by whole-body plethysmography. Mice were preinjected with oxycodone then implanted with mini-pumps (s.c.) delivering 20, 45 or 120 mg·kg-1 ·day-1 oxycodone for 6 days and subsequently challenged with oxycodone (3 mg·kg-1 , i.p.) or morphine (10 mg·kg-1 , i.p.) to assess the level of tolerance. KEY RESULTS: Oxycodone-treated mice developed tolerance to oxycodone and cross tolerance to morphine-induced respiratory depression. Tolerance was less with 20 mg·kg-1 ·day-1 than with 45 or 120 mg·kg-1 ·day-1 oxycodone treatment. At doses that do not depress respiration, ethanol (0.3 g·kg-1 ), pregabalin (20 mg·kg-1 ) and calphostin C (45 µg·kg-1 ) all reversed oxycodone-induced tolerance resulting in significant respiratory depression. Reversal of tolerance was less in mice treated with oxycodone (120 mg·kg-1 ·day-1 ). In mice receiving ethanol and calphostin C or ethanol and pregabalin, there was no greater reversal of tolerance than seen with either drug alone. CONCLUSION AND IMPLICATIONS: These data suggest that oxycodone-induced tolerance is mediated by PKC and that reversal of tolerance by ethanol or pregabalin may be a contributory factor in oxycodone overdose deaths.


Assuntos
Analgésicos Opioides/farmacologia , Etanol/farmacologia , Oxicodona/farmacologia , Pregabalina/farmacologia , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Insuficiência Respiratória/tratamento farmacológico , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/análise , Animais , Etanol/administração & dosagem , Masculino , Camundongos , Morfina , Oxicodona/administração & dosagem , Oxicodona/análise , Pregabalina/administração & dosagem , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Insuficiência Respiratória/induzido quimicamente
13.
Addiction ; 112(9): 1580-1589, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28493329

RESUMO

AIM: To examine the risk to heroin users of also using gabapentin or pregabalin (gabapentoids). DESIGN: Multi-disciplinary study: we (a) examined trends in drug-related deaths and gabapentoid prescription data in England and Wales to test for evidence that any increase in deaths mentioning gabapentin or pregabalin is associated with trends in gabapentoid prescribing and is concomitant with opioid use; (b) interviewed people with a history of heroin use about their polydrug use involving gabapentin and pregabalin; and (c) studied the respiratory depressant effects of pregabalin in the absence and presence of morphine in mice to determine whether concomitant exposure increased the degree of respiratory depression observed. SETTING: England and Wales. PARTICIPANTS: Interviews were conducted with 30 participants (19 males, 11 female). MEASUREMENTS: (a) Office of National Statistics drug-related deaths from 1 January 2004 to 31 December 2015 that mention both an opioid and pregabalin or gabapentin; (b) subjective views on the availability, use, interactions and effects of polydrug use involving pregabalin and gabapentin; and (c) rate and depth of respiration. RESULTS: Pregabalin and gabapentin prescriptions increased approximately 24% per year from 1 million in 2004 to 10.5 million in 2015. The number of deaths involving gabapentoids increased from fewer than one per year prior to 2009 to 137 in 2015; 79% of these deaths also involved opioids. The increase in deaths was correlated highly with the increase in prescribing (correlation coefficient 0.94; 5% increase in deaths per 100 000 increase in prescriptions). Heroin users described pregabalin as easy to obtain. They suggested that the combination of heroin and pregabalin reinforced the effects of heroin but were concerned it induced 'blackouts' and increased the risk of overdose. In mice, a low dose of S-pregabalin (20 mg/kg) that did not itself depress respiration reversed tolerance to morphine depression of respiration (resulting in 35% depression of respiration, P < 0.05), whereas a high dose of S-pregabalin (200 mg/kg) alone depressed respiration and this effect summated with that of morphine. CONCLUSIONS: For heroin users, the combination of opioids with gabapentin or pregabalin potentially increases the risk of acute overdose death through either reversal of tolerance or an additive effect of the drugs to depress respiration.


Assuntos
Aminas/farmacologia , Analgésicos/farmacologia , Ácidos Cicloexanocarboxílicos/farmacologia , Overdose de Drogas/mortalidade , Usuários de Drogas/estatística & dados numéricos , Dependência de Heroína/mortalidade , Pregabalina/farmacologia , Ácido gama-Aminobutírico/farmacologia , Adulto , Animais , Modelos Animais de Doenças , Interações Medicamentosas , Inglaterra/epidemiologia , Feminino , Gabapentina , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Risco , País de Gales/epidemiologia , Adulto Jovem
14.
J Pharmacol Exp Ther ; 361(1): 51-59, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28130265

RESUMO

Respiratory depression is the major cause of death in opioid overdose. We have previously shown that prolonged treatment of mice with morphine induces profound tolerance to the respiratory-depressant effects of the drug (Hill et al., 2016). The aim of the present study was to investigate whether tolerance to opioid-induced respiratory depression is mediated by protein kinase C (PKC) and/or c-Jun N-terminal kinase (JNK). We found that although mice treated for up to 6 days with morphine developed tolerance, as measured by the reduced responsiveness to an acute challenge dose of morphine, administration of the brain-penetrant PKC inhibitors tamoxifen and calphostin C restored the ability of acute morphine to produce respiratory depression in morphine-treated mice. Importantly, reversal of opioid tolerance was dependent on the nature of the opioid ligand used to induce tolerance, as these PKC inhibitors did not reverse tolerance induced by prolonged treatment of mice with methadone nor did they reverse the protection to acute morphine-induced respiratory depression afforded by prolonged treatment with buprenorphine. We found no evidence for the involvement of JNK in morphine-induced tolerance to respiratory depression. These results indicate that PKC represents a major mechanism underlying morphine tolerance, that the mechanism of opioid tolerance to respiratory depression is ligand-dependent, and that coadministration of drugs with PKC-inhibitory activity and morphine (as well as heroin, largely metabolized to morphine in the body) may render individuals more susceptible to overdose death by reversing tolerance to the effects of morphine.


Assuntos
Analgésicos Opioides/toxicidade , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Insuficiência Respiratória/enzimologia , Tamoxifeno/uso terapêutico , Analgésicos Opioides/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Tolerância a Medicamentos/fisiologia , Bombas de Infusão Implantáveis , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Masculino , Camundongos , Proteína Quinase C/antagonistas & inibidores , Distribuição Aleatória , Insuficiência Respiratória/induzido quimicamente , Tamoxifeno/farmacologia , Resultado do Tratamento
15.
PLoS One ; 11(4): e0153187, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27077912

RESUMO

Opioids are widely used medicinally as analgesics and abused for hedonic effects, actions that are each complicated by substantial risks such as cardiorespiratory depression. These drugs mimic peptides such as ß-endorphin, which has a key role in endogenous analgesia. The ß-endorphin in the central nervous system originates from pro-opiomelanocortin (POMC) neurons in the arcuate nucleus and nucleus of the solitary tract (NTS). Relatively little is known about the NTSPOMC neurons but their position within the sensory nucleus of the vagus led us to test the hypothesis that they play a role in modulation of cardiorespiratory and nociceptive control. The NTSPOMC neurons were targeted using viral vectors in a POMC-Cre mouse line to express either opto-genetic (channelrhodopsin-2) or chemo-genetic (Pharmacologically Selective Actuator Modules). Opto-genetic activation of the NTSPOMC neurons in the working heart brainstem preparation (n = 21) evoked a reliable, titratable and time-locked respiratory inhibition (120% increase in inter-breath interval) with a bradycardia (125±26 beats per minute) and augmented respiratory sinus arrhythmia (58% increase). Chemo-genetic activation of NTSPOMC neurons in vivo was anti-nociceptive in the tail flick assay (latency increased by 126±65%, p<0.001; n = 8). All effects of NTSPOMC activation were blocked by systemic naloxone (opioid antagonist) but not by SHU9119 (melanocortin receptor antagonist). The NTSPOMC neurons were found to project to key brainstem structures involved in cardiorespiratory control (nucleus ambiguus and ventral respiratory group) and endogenous analgesia (periaqueductal gray and midline raphe). Thus the NTSPOMC neurons may be capable of tuning behaviour by an opioidergic modulation of nociceptive, respiratory and cardiac control.


Assuntos
Analgesia , Bradicardia/metabolismo , Tronco Encefálico/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Insuficiência Respiratória/metabolismo , Analgésicos Opioides/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Tronco Encefálico/citologia , Tronco Encefálico/efeitos dos fármacos , Channelrhodopsins , Feminino , Masculino , Hormônios Estimuladores de Melanócitos/farmacologia , Camundongos Transgênicos , Microscopia Confocal , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Núcleo Solitário/citologia , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/metabolismo
16.
Neuropsychopharmacology ; 41(3): 762-73, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26171718

RESUMO

Opioids are the most common drugs associated with unintentional drug overdose. Death results from respiratory depression. Prolonged use of opioids results in the development of tolerance but the degree of tolerance is thought to vary between different effects of the drugs. Many opioid addicts regularly consume alcohol (ethanol), and post-mortem analyses of opioid overdose deaths have revealed an inverse correlation between blood morphine and ethanol levels. In the present study, we determined whether ethanol reduced tolerance to the respiratory depressant effects of opioids. Mice were treated with opioids (morphine, methadone, or buprenorphine) for up to 6 days. Respiration was measured in freely moving animals breathing 5% CO2 in air in plethysmograph chambers. Antinociception (analgesia) was measured as the latency to remove the tail from a thermal stimulus. Opioid tolerance was assessed by measuring the response to a challenge dose of morphine (10 mg/kg i.p.). Tolerance developed to the respiratory depressant effect of morphine but at a slower rate than tolerance to its antinociceptive effect. A low dose of ethanol (0.3 mg/kg) alone did not depress respiration but in prolonged morphine-treated animals respiratory depression was observed when ethanol was co-administered with the morphine challenge. Ethanol did not alter the brain levels of morphine. In contrast, in methadone- or buprenorphine-treated animals no respiratory depression was observed when ethanol was co-administered along with the morphine challenge. As heroin is converted to morphine in man, selective reversal of morphine tolerance by ethanol may be a contributory factor in heroin overdose deaths.


Assuntos
Analgésicos Opioides/farmacologia , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Morfina/farmacologia , Medicamentos para o Sistema Respiratório/farmacologia , Analgésicos Opioides/farmacocinética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Buprenorfina/farmacologia , Corticosterona/sangue , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Comportamento Exploratório/efeitos dos fármacos , Temperatura Alta , Masculino , Metadona/farmacologia , Camundongos , Morfina/farmacocinética , Atividade Motora/efeitos dos fármacos , Dor Nociceptiva/tratamento farmacológico , Limiar da Dor/efeitos dos fármacos , Pletismografia , Respiração , Medicamentos para o Sistema Respiratório/farmacocinética , Volume de Ventilação Pulmonar/efeitos dos fármacos
17.
Cancer Cell ; 2(2): 157-64, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12204536

RESUMO

Birt-Hogg-Dubé (BHD) syndrome is a rare inherited genodermatosis characterized by hair follicle hamartomas, kidney tumors, and spontaneous pneumothorax. Recombination mapping in BHD families delineated the susceptibility locus to 700 kb on chromosome 17p11.2. Protein-truncating mutations were identified in a novel candidate gene in a panel of BHD families, with a 44% frequency of insertion/deletion mutations within a hypermutable C(8) tract. Tissue expression of the 3.8 kb transcript was widespread, including kidney, lung, and skin. The full-length BHD sequence predicted a novel protein, folliculin, that was highly conserved across species. Discovery of disease-causing mutations in BHD, a novel kidney cancer gene associated with renal oncocytoma or chromophobe renal cancer, will contribute to understanding the role of folliculin in pathways common to skin, lung, and kidney development.


Assuntos
Estrona/genética , Folículo Piloso/patologia , Hamartoma/genética , Neoplasias Renais/genética , Mutação/genética , Pneumotórax/genética , Sequência de Aminoácidos , Sequência de Bases , Cromossomos Humanos Par 17/genética , Sequência Conservada , Análise Mutacional de DNA , Estrona/química , Éxons/genética , Feminino , Mutação da Fase de Leitura/genética , Predisposição Genética para Doença , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Mapeamento Físico do Cromossomo , Pneumotórax/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA