Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Pharmacol Exp Ther ; 385(3): 180-192, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37019472

RESUMO

Mitragynine, an opioidergic alkaloid present in Mitragyna speciosa (kratom), is metabolized by cytochrome P450 3A (CYP3A) to 7-hydroxymitragynine, a more potent opioid receptor agonist. The extent to which conversion to 7-hydroxymitragynine mediates the in vivo effects of mitragynine is unclear. The current study examined how CYP3A inhibition (ketoconazole) modifies the pharmacokinetics of mitragynine in rat liver microsomes in vitro. The study further examined how ketoconazole modifies the discriminative stimulus and antinociceptive effects of mitragynine in rats. Ketoconazole [30 mg/kg, oral gavage (o.g.)] increased systemic exposure to mitragynine (13.3 mg/kg, o.g.) by 120% and 7-hydroxymitragynine exposure by 130%. The unexpected increase in exposure to 7-hydroxymitragynine suggested that ketoconazole inhibits metabolism of both mitragynine and 7-hydroxymitragynine, a finding confirmed in rat liver microsomes. In rats discriminating 3.2 mg/kg morphine from vehicle under a fixed-ratio schedule of food delivery, ketoconazole pretreatment increased the potency of both mitragynine (4.7-fold) and 7-hydroxymitragynine (9.7-fold). Ketoconazole did not affect morphine's potency. Ketoconazole increased the antinociceptive potency of 7-hydroxymitragynine by 4.1-fold. Mitragynine (up to 56 mg/kg, i.p.) lacked antinociceptive effects both in the presence and absence of ketoconazole. These results suggest that both mitragynine and 7-hydroxymitragynine are cleared via CYP3A and that 7-hydroxymitragynine is formed as a metabolite of mitragynine by other routes. These results have implications for kratom use in combination with numerous medications and citrus juices that inhibit CYP3A. SIGNIFICANCE STATEMENT: Mitragynine is an abundant kratom alkaloid that exhibits low efficacy at the µ-opioid receptor (MOR). Its metabolite, 7-hydroxymitragynine, is also an MOR agonist but with higher affinity and efficacy than mitragynine. Our results in rats demonstrate that cytochrome P450 3A (CYP3A) inhibition can increase the systematic exposure of both mitragynine and 7-hydroxymitragynine and their potency to produce MOR-mediated behavioral effects. These data highlight potential interactions between kratom and CYP3A inhibitors, which include numerous medications and citrus juices.


Assuntos
Citocromo P-450 CYP3A , Alcaloides de Triptamina e Secologanina , Ratos , Animais , Cetoconazol/farmacologia , Alcaloides de Triptamina e Secologanina/metabolismo , Morfina/farmacologia , Analgésicos Opioides/farmacologia
2.
J Pharmacol Exp Ther ; 383(3): 182-198, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36153006

RESUMO

The primary kratom alkaloid mitragynine is proposed to act through multiple mechanisms, including actions at µ-opioid receptors (MORs) and adrenergic-α 2 receptors (Aα 2Rs), as well as conversion in vivo to a MOR agonist metabolite (i.e., 7-hydroxymitragynine). Aα 2R and MOR agonists can produce antinociceptive synergism. Here, contributions of both receptors to produce mitragynine-related effects were assessed by measuring receptor binding in cell membranes and, in rats, pharmacological behavioral effect antagonism studies. Mitragynine displayed binding affinity at both receptors, whereas 7-hydroxymitragynine only displayed MOR binding affinity. Compounds were tested for their capacity to decrease food-maintained responding and rectal temperature and to produce antinociception in a hotplate test. Prototypical MOR agonists and 7-hydroxymitragynine, but not mitragynine, produced antinociception. MOR agonist and 7-hydroxymitragynine rate-deceasing and antinociceptive effects were antagonized by the opioid antagonist naltrexone but not by the Aα 2R antagonist yohimbine. Hypothermia only resulted from reference Aα 2R agonists. The rate-deceasing and hypothermic effects of reference Aα 2R agonists were antagonized by yohimbine but not naltrexone. Neither naltrexone nor yohimbine antagonized the rate-decreasing effects of mitragynine. Mitragynine and 7-hydroxymitragynine increased the potency of the antinociceptive effects of Aα 2R but not MOR reference agonists. Only mitragynine produced hypothermic effects. Isobolographic analyses for the rate-decreasing effects of the reference Aα 2R and MOR agonists were also conducted. These results suggest mitragynine and 7-hydroxymitragynine may produce antinociceptive synergism with Aα 2R and MOR agonists. When combined with Aα 2R agonists, mitragynine could also produce hypothermic synergism. SIGNIFICANCE STATEMENT: Mitragynine is proposed to target the µ-opioid receptor (MOR) and adrenergic-α2 receptor (Aα2R) and to produce behavioral effects through conversion to its MOR agonist metabolite 7-hydroxymitragynine. Isobolographic analyses indicated supra-additivity in some dose ratio combinations. This study suggests mitragynine and 7-hydroxymitragynine may produce antinociceptive synergism with Aα2R and MOR agonists. When combined with Aα2R agonists, mitragynine could also produce hypothermic synergism.


Assuntos
Mitragyna , Alcaloides de Triptamina e Secologanina , Animais , Ratos , Agonistas de Receptores Adrenérgicos alfa 2 , Analgésicos Opioides/farmacologia , Mitragyna/química , Naltrexona/farmacologia , Receptores Adrenérgicos alfa 2 , Receptores Opioides mu/agonistas , Alcaloides de Triptamina e Secologanina/farmacologia , Ioimbina/farmacologia
3.
Behav Pharmacol ; 33(6): 427-434, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35947069

RESUMO

Baclofen and γ-hydroxybutyrate (GHB) exert γ-aminobutyric acid (GABA)B receptor agonism and have therapeutic utility but possess different pharmacological activities. We examined whether separate groups of mice could be trained to discriminate either baclofen or GHB, and the contribution of GABAB receptors to discriminative stimulus effects. Male C57BL/6J mice were trained to discriminate either baclofen (3.2 mg/kg, intraperitoneal) or GHB (178 mg/kg, intraperitoneal) from saline under a fixed-ratio 10 schedule. The GABAB antagonist 3-aminopropyl(diethoxymethyl)phosphinic acid (CGP 35348) was used to pharmacologically assess GABAB receptor involvement. The selectivity of the resulting discriminations was assessed with the opioid agonist morphine and the benzodiazepine midazolam. In baclofen-trained mice, both baclofen and GHB were readily discriminated. Baclofen produced a maximum of 86% baclofen-appropriate responding. CGP 35348 (320 mg/kg, i.p.) produced a 4.7-fold rightward shift in the dose-effect function. GHB produced a maximum of 85.8% baclofen-appropriate responding. In GHB-trained mice, both GHB and baclofen were readily discriminated. In GHB-trained mice, GHB produced a maximum of 85.3% drug-appropriate responding; CGP 35348 (320 mg/kg, i.p.) produced a 1.8-fold rightward shift in the GHB discrimination dose-effect function. Baclofen produced up to 70.0% GHB-appropriate responding. CGP 35348 (320 mg/kg, i.p.) significantly antagonized baclofen discrimination and baclofen produced up to 37% GHB-appropriate responding up to doses that disrupted operant responding. Morphine did not produce substitution for either baclofen or GHB. Midazolam produced partial substitution for both. GHB and baclofen discrimination assays in mice provide a useful approach for examining different receptor types mediating the effects of these two drugs.


Assuntos
Oxibato de Sódio , Animais , Baclofeno/farmacologia , Agonistas GABAérgicos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Midazolam/farmacologia , Derivados da Morfina , Receptores de GABA-B/fisiologia , Oxibato de Sódio/farmacologia
4.
J Pharmacol Exp Ther ; 376(3): 410-427, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33384303

RESUMO

Relationships between µ-opioid receptor (MOR) efficacy and effects of mitragynine and 7-hydroxymitragynine are not fully established. We assessed in vitro binding affinity and efficacy and discriminative stimulus effects together with antinociception in rats. The binding affinities of mitragynine and 7-hydroxymitragynine at MOR (Ki values 77.9 and 709 nM, respectively) were higher than their binding affinities at κ-opioid receptor (KOR) or δ-opioid receptor (DOR). [35S]guanosine 5'-O-[γ-thio]triphosphate stimulation at MOR demonstrated that mitragynine was an antagonist, whereas 7-hydroxymitragynine was a partial agonist (Emax = 41.3%). In separate groups of rats discriminating either morphine (3.2 mg/kg) or mitragynine (32 mg/kg), mitragynine produced a maximum of 72.3% morphine-lever responding, and morphine produced a maximum of 65.4% mitragynine-lever responding. Other MOR agonists produced high percentages of drug-lever responding in the morphine and mitragynine discrimination assays: 7-hydroxymitragynine (99.7% and 98.1%, respectively), fentanyl (99.7% and 80.1%, respectively), buprenorphine (99.8% and 79.4%, respectively), and nalbuphine (99.4% and 98.3%, respectively). In the morphine and mitragynine discrimination assays, the KOR agonist U69,593 produced maximums of 72.3% and 22.3%, respectively, and the DOR agonist SNC 80 produced maximums of 34.3% and 23.0%, respectively. 7-Hydroxymitragynine produced antinociception; mitragynine did not. Naltrexone antagonized all of the effects of morphine and 7-hydroxymitragynine; naltrexone antagonized the discriminative stimulus effects of mitragynine but not its rate-decreasing effects. Mitragynine increased the potency of the morphine discrimination yet decreased morphine antinociception. Here we illustrate striking differences in MOR efficacy, with mitragynine having less than 7-hydroxymitragynine. SIGNIFICANCE STATEMENT: At human µ-opioid receptor (MOR) in vitro, mitragynine has low affinity and is an antagonist, whereas 7-hydroxymitragynine has 9-fold higher affinity than mitragynine and is an MOR partial agonist. In rats, intraperitoneal mitragynine exhibits a complex pharmacology including MOR agonism; 7-hydroxymitragynine has higher MOR potency and efficacy than mitragynine. These results are consistent with 7-hydroxymitragynine being a highly selective MOR agonist and with mitragynine having a complex pharmacology that combines low efficacy MOR agonism with activity at nonopioid receptors.


Assuntos
Comportamento Animal/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Alcaloides de Triptamina e Secologanina/metabolismo , Alcaloides de Triptamina e Secologanina/farmacologia , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Células CHO , Cricetulus , Aprendizagem por Discriminação/efeitos dos fármacos , Células HEK293 , Humanos , Ligação Proteica , Ratos
5.
Mol Pharmacol ; 98(4): 328-342, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32690626

RESUMO

Epibatidine is a potent analgetic agent with very high affinity for brain nicotinic acetylcholine receptors (nAChR). We determined the activity profiles of three epibatidine derivatives, RTI-36, RTI-76, and RTI-102, which have affinity for brain nAChR equivalent to that of epibatidine but reduced analgetic activity. RNAs coding for nAChR monomeric subunits and/or concatamers were injected into Xenopus oocytes to obtain receptors of defined subunit composition and stoichiometry. The epibatidine analogs produced protracted activation of high sensitivity (HS) α4- and α2-containing receptors with the stoichiometry of 2alpha:3beta subunits but not low sensitivity (LS) receptors with the reverse ratio of alpha and beta subunits. Although not strongly activated by the epibatidine analogs, LS α4- and α2-containing receptors were potently desensitized by the epibatidine analogs. In general, the responses of α4(2)ß2(2)α5 and ß3α4ß2α6ß2 receptors were similar to those of the HS α4ß2 receptors. RTI-36, the analog closest in structure to epibatidine, was the most efficacious of the three compounds, also effectively activating α7 and α3ß4 receptors, albeit with lower potency and less desensitizing effect. Although not the most efficacious agonist, RTI-76 was the most potent desensitizer of α4- and α2-containing receptors. RTI-102, a strong partial agonist for HS α4ß2 receptors, was effectively an antagonist for LS α4ß2 receptors. Our results highlight the importance of subunit stoichiometry and the presence or absence of specific accessory subunits for determining the activity of these drugs on brain nAChR, affecting the interpretation of in vivo studies since in most cases these structural details are not known. SIGNIFICANCE STATEMENT: Epibatidine and related compounds are potent ligands for the high-affinity nicotine receptors of the brain, which are therapeutic targets and mediators of nicotine addiction. Far from being a homogeneous population, these receptors are diverse in subunit composition and vary in subunit stoichiometry. We show the importance of these structural details for drug activity profiles, which present a challenge for the interpretation of in vivo experiments since conventional methods, such as in situ hybridization and immunohistochemistry, cannot illuminate these details.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Agonistas Nicotínicos/farmacologia , Subunidades Proteicas/metabolismo , Piridinas/química , Receptores Nicotínicos/metabolismo , Animais , Animais Geneticamente Modificados , Encéfalo/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/química , Humanos , Estrutura Molecular , Complexos Multiproteicos/metabolismo , Agonistas Nicotínicos/química , Subunidades Proteicas/genética , Receptores Nicotínicos/genética , Tropanos/química , Tropanos/farmacologia , Xenopus/genética
6.
J Pharmacol Exp Ther ; 370(3): 380-389, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31235534

RESUMO

Better therapeutic options are needed for pain. Baclofen, buspirone, and morphine are characterized as having analgesic properties. However, little is known about potential interactions between analgesic effects of these drugs when combined. Furthermore, it is not known if the magnitude of these potential interactions will be similar for all drug effects. Thus, we tested the effects of these drugs alone and in combination for their capacity to produce thermal antinociception and to decrease food-maintained responding. Four male and four female Sprague-Dawley rats responded for food under a fixed-ratio 10 schedule; afterward they were immediately placed on a 52°C hot plate. Morphine, baclofen, and buspirone were examined alone and in 1:1 combinations, based upon ED50 values. Morphine and baclofen effects were evaluated with the opioid antagonist naltrexone and the GABAB antagonist (3-Aminopropyl)(diethoxymethyl)phosphinic acid (CGP35348), respectively. Morphine, baclofen, and buspirone dose dependently decreased operant responding, with the calculated ED50 values being 7.09, 3.42, and 0.57 mg/kg, respectively. The respective antinociception ED50 values were 16.15, 8.75, and 2.20 mg/kg. Analysis of 1:1 combinations showed the effects of morphine plus baclofen to decrease schedule-controlled responding and to produce thermal antinociception were synergistic. Effects of morphine plus buspirone and baclofen plus buspirone to decrease schedule-controlled responding were additive. Effects of the two combinations to produce thermal antinociception were synergistic. Naltrexone and CGP35348 antagonized the effects of morphine and baclofen, respectively. Synergistic antinociceptive effects, in conjunction with additive effects on food-maintained responding, highlight the therapeutic utility of opioid and non-opioid drug combinations.


Assuntos
Analgésicos/farmacologia , Baclofeno/farmacologia , Buspirona/farmacologia , Morfina/farmacologia , Tempo de Reação/efeitos dos fármacos , Temperatura , Animais , Relação Dose-Resposta a Droga , Interações Medicamentosas , Tolerância a Medicamentos , Feminino , Antagonistas de Receptores de GABA-B/farmacologia , Masculino , Compostos Organofosforados/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-B/metabolismo
7.
J Pharmacol Exp Ther ; 366(3): 509-518, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29945931

RESUMO

The immunomodulatory prodrug 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol (FTY720), which acts as an agonist for sphingosine-1-phosphate (S1P) receptors (S1PR) when phosphorylated, is proposed as a novel pain therapeutic. In this study, we assessed FTY720-mediated antinociception in the radiant heat tail-flick test and in the chronic constriction injury (CCI) model of neuropathic pain in mice. FTY720 produced antinociception and antiallodynia, respectively, and these effects were dose-dependent and mimicked by the S1PR1-selective agonist CYM-5442. Repeated administration of FTY720 for 1 week produced tolerance to acute thermal antinociception, but not to antiallodynia in the CCI model. S1PR-stimulated [35S]GTPγS autoradiography revealed apparent desensitization of G protein activation by S1P or the S1PR1 agonist 5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)phenyl]-1,2,4-oxadiazole (SEW-2871) throughout the brain. Similar results were seen in spinal cord membranes, whereby the Emax value of S1PR-stimulated [35S]GTPγS binding was greatly reduced in repeated FTY720-treated mice. These results suggest that S1PR1 is a primary target of FTY720 in alleviating both acute thermal nociception and chronic neuropathic nociception. Furthermore, the finding that tolerance develops to antinociception in the tail-flick test but not in chronic neuropathic pain suggests a differential mechanism of FTY720 action between these models. The observation that repeated FTY720 administration led to desensitized S1PR1 signaling throughout the central nervous system suggests the possibility that S1PR1 activation drives the acute thermal antinociceptive effects, whereas S1PR1 desensitization mediates the following: 1) tolerance to thermal antinociceptive actions of FTY720 and 2) the persistent antiallodynic effects of FTY720 in neuropathic pain by producing functional antagonism of pronociceptive S1PR1 signaling.


Assuntos
Cloridrato de Fingolimode/farmacologia , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Peptídeos Opioides/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/metabolismo , Temperatura , Animais , Modelos Animais de Doenças , Cloridrato de Fingolimode/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos ICR , Neuralgia/fisiopatologia , Receptores de Lisoesfingolipídeo/agonistas , Nociceptina
8.
J Pharmacol Exp Ther ; 366(1): 169-183, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29540562

RESUMO

Although paclitaxel effectively treats various cancers, its debilitating peripheral neuropathic pain side effects often persist long after treatment has ended. Therefore, a compelling need exists for the identification of novel pharmacologic strategies to mitigate this condition. As inhibitors of monoacylglycerol lipase (MAGL), the primary hydrolytic enzyme of the endogenous cannabinoid, 2-arachidonyolglycerol, produces antinociceptive effects in numerous rodent models of pain, we investigated whether inhibitors of this enzyme (i.e., JZL184 and MJN110) would reverse paclitaxel-induced mechanical allodynia in mice. These drugs dose dependently reversed allodynia with respective ED50 values (95% confidence limit) of 8.4 (5.2-13.6) and 1.8 (1.0-3.3) mg/kg. Complementary genetic and pharmacologic approaches revealed that the antiallodynic effects of each drug require both cannabinoid receptors, CB1 and CB2 MJN110 reduced paclitaxel-mediated increased expression of monocyte chemoattractant protein-1 (MCP-1, CCL2) and phospho-p38 MAPK in dorsal root ganglia as well as MCP-1 in spinal dorsal horn. Whereas the antinociceptive effects of high dose JZL184 (40 mg/kg) underwent tolerance following 6 days of repeated dosing, repeated administration of a threshold dose (i.e., 4 mg/kg) completely reversed paclitaxel-induced allodynia. In addition, we found that the administration of MJN110 to control mice lacked intrinsic rewarding effects in the conditioned place preference (CPP) paradigm. However, it produced a CPP in paclitaxel-treated animals, suggesting a reduced paclitaxel-induced aversive state. Importantly, JZL184 did not alter the antiproliferative and apoptotic effects of paclitaxel in A549 and H460 non-small cell lung cancer cells. Taken together, these data indicate that MAGL inhibitors reverse paclitaxel-induced neuropathic pain without interfering with chemotherapeutic efficacy.


Assuntos
Antineoplásicos/efeitos adversos , Inibidores Enzimáticos/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Monoacilglicerol Lipases/antagonistas & inibidores , Nociceptividade/efeitos dos fármacos , Paclitaxel/efeitos adversos , Animais , Apoptose/efeitos dos fármacos , Benzodioxóis/farmacologia , Benzodioxóis/uso terapêutico , Biomarcadores/metabolismo , Carbamatos/farmacologia , Carbamatos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/uso terapêutico , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Inflamação/metabolismo , Masculino , Camundongos , Fosfoproteínas/metabolismo , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Succinimidas/farmacologia , Succinimidas/uso terapêutico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
J Pharmacol Exp Ther ; 363(3): 394-401, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28970359

RESUMO

Diacylglycerol lipase (DAGL) α and ß, the major biosynthetic enzymes of the endogenous cannabinoid (endocannabinoid) 2-arachidonylglycerol (2-AG), are highly expressed in the nervous system and immune system, respectively. Genetic deletion or pharmacological inhibition of DAGL-ß protects against lipopolysaccharide (LPS)-induced inflammatory responses in mouse peritoneal macrophages and reverses LPS-induced allodynia in mice. To gain insight into the contribution of DAGL-α in LPS-induced allodynia, we tested global knockout mice as well as DO34, a dual DAGL-α/ß inhibitor. Intraperitoneal administration of DO34 (30 mg/kg) significantly decreased whole-brain levels of 2-AG (∼83%), anandamide (∼42%), and arachidonic acid (∼58%). DO34 dose-dependently reversed mechanical and cold allodynia, and these antinociceptive effects did not undergo tolerance after 6 days of repeated administration. In contrast, DO34 lacked acute thermal antinociceptive, motor, and hypothermal pharmacological effects in naive mice. As previously reported, DAGL-ß (-/-) mice displayed a protective phenotype from LPS-induced allodynia. However, DAGL-α (-/-) mice showed full allodynic responses, similar to their wild-type littermates. Interestingly, DO34 (30 mg/kg) fully reversed LPS-induced allodynia in DAGL-α (+/+) and (-/-) mice, but did not affect the antinociceptive phenotype of DAGL-ß (-/-) mice in this model, indicating a DAGL-α-independent site of action. These findings suggest that DAGL-α and DAGL-ß play distinct roles in LPS-induced nociception. Whereas DAGL-α appears to be dispensable for the development and expression of LPS-induced nociception, DAGL-ß inhibition represents a promising strategy to treat inflammatory pain.


Assuntos
Analgésicos/farmacologia , Lipopolissacarídeos/farmacologia , Lipase Lipoproteica/antagonistas & inibidores , Dor/enzimologia , Tiazóis/farmacologia , Ureia/análogos & derivados , Ureia/farmacologia , Analgésicos/uso terapêutico , Animais , Temperatura Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Tolerância a Medicamentos , Endocanabinoides/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Inflamação/fisiopatologia , Inflamação/psicologia , Lipase Lipoproteica/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Dor/tratamento farmacológico , Dor/fisiopatologia , Dor/psicologia , Tiazóis/uso terapêutico , Ureia/uso terapêutico
10.
J Pharmacol Exp Ther ; 359(2): 310-318, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27608657

RESUMO

Chemotherapy-induced peripheral neuropathy (CIPN) represents a serious complication associated with antineoplastic drugs. Although there are no medications available that effectively prevent CIPN, many classes of drugs have been used to treat this condition, including anticonvulsants, serotonin and noradrenaline reuptake inhibitors, and opioids. However, these therapeutic options yielded inconclusive results in CIPN clinical trials and produced assorted side effects with their prolonged use. Thus, there is an urgent need to develop efficacious and safe treatments for CIPN. In this report, we tested whether the endogenous lipid palmitoylethanolamide (PEA) alone or in combination with the anticonvulsant gabapentin would reduce allodynia in a mouse paclitaxel model of CIPN. Gabapentin and PEA reversed paclitaxel-induced allodynia with respective ED50 doses (95% confidence interval) of 67.4 (61.52-73.94) and 9.2 (8.39-10.16) mg/kg. Isobolographic analysis of these drugs in combination revealed synergistic antiallodynic effects. The PPAR-α antagonist receptor antagonist GW6471 [N-((2S)-2-(((1Z)-1-methyl-3-oxo-3-(4-(trifluoromethyl)phenyl)prop-1-enyl)amino)-3-(4-(2-(5-methyl-2-phenyl-1,3-oxazol-4-yl)ethoxy)phenyl)propyl)propanamide] completely blocked the antinociceptive effects of PEA. In addition, PEA administered via intraplantar injection into a paw, intrathecal injection, and intracerebroventricular injection reversed paclitaxel-induced allodynia, suggesting that it may act at multiple sites in the neuroaxis and periphery. Finally, repeated administration of PEA (30 mg/kg, 7 days) preserved the antiallodynic effects with no evidence of tolerance. These findings taken together suggest that PEA possesses potential to treat peripheral neuropathy in cancer patients undergoing chemotherapy.


Assuntos
Etanolaminas/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Paclitaxel/efeitos adversos , Ácidos Palmíticos/farmacologia , Amidas , Aminas/farmacologia , Animais , Ácidos Cicloexanocarboxílicos/farmacologia , Sinergismo Farmacológico , Etanolaminas/administração & dosagem , Etanolaminas/uso terapêutico , Gabapentina , Hiperalgesia/metabolismo , Masculino , Camundongos , PPAR alfa/metabolismo , Ácidos Palmíticos/administração & dosagem , Ácidos Palmíticos/uso terapêutico , Ácido gama-Aminobutírico/farmacologia
11.
J Pharmacol Exp Ther ; 357(1): 145-56, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26791602

RESUMO

Serious clinical liabilities associated with the prescription of opiates for pain control include constipation, respiratory depression, pruritus, tolerance, abuse, and addiction. A recognized strategy to circumvent these side effects is to combine opioids with other antinociceptive agents. The combination of opiates with the primary active constituent of cannabis (Δ(9)-tetrahydrocannabinol) produces enhanced antinociceptive actions, suggesting that cannabinoid receptor agonists can be opioid sparing. Here, we tested whether elevating the endogenous cannabinoid 2-arachidonoylglycerol through the inhibition of its primary hydrolytic enzyme monoacylglycerol lipase (MAGL), will produce opioid-sparing effects in the mouse chronic constriction injury (CCI) of the sciatic nerve model of neuropathic pain. The dose-response relationships of i.p. administration of morphine and the selective MAGL inhibitor 2,5-dioxopyrrolidin-1-yl 4-(bis(4-chlorophenyl)methyl)piperazine-1-carboxylate (MJN110) were tested alone and in combination at equieffective doses for reversal of CCI-induced mechanical allodynia and thermal hyperalgesia. The respective ED50 doses (95% confidence interval) of morphine and MJN110 were 2.4 (1.9-3.0) mg/kg and 0.43 (0.23-0.79) mg/kg. Isobolographic analysis of these drugs in combination revealed synergistic antiallodynic effects. Acute antinociceptive effects of the combination of morphine and MJN110 required µ-opioid, CB1, and CB2 receptors. This combination did not reduce gastric motility or produce subjective cannabimimetic effects in the drug discrimination assay. Importantly, combinations of MJN110 and morphine given repeatedly (i.e., twice a day for 6 days) continued to produce antiallodynic effects with no evidence of tolerance. Taken together, these findings suggest that MAGL inhibition produces opiate-sparing events with diminished tolerance, constipation, and cannabimimetic side effects.


Assuntos
Analgésicos Opioides/uso terapêutico , Carbamatos/farmacologia , Inibidores Enzimáticos/farmacologia , Monoacilglicerol Lipases/antagonistas & inibidores , Neuralgia/tratamento farmacológico , Succinimidas/farmacologia , Animais , Ácidos Araquidônicos/metabolismo , Comportamento Animal/efeitos dos fármacos , Constrição Patológica/complicações , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Endocanabinoides/metabolismo , Glicerídeos/metabolismo , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Morfina/uso terapêutico , Neuralgia/induzido quimicamente , Neuralgia/psicologia , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptor CB2 de Canabinoide/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos
12.
J Pharmacol Exp Ther ; 353(2): 424-32, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25762694

RESUMO

The endogenous cannabinoid 2-arachidonoylglycerol (2-AG) plays an important role in a variety of physiologic processes, but its rapid breakdown by monoacylglycerol lipase (MAGL) results in short-lived actions. Initial MAGL inhibitors were limited by poor selectivity and low potency. In this study, we tested JZL184 [4-nitrophenyl 4-[bis(2H-1,3-benzodioxol-5-yl)(hydroxy)methyl]piperidine-1-carboxylate] and MJN110 [2,5-dioxopyrrolidin-1-yl 4-(bis(4-chlorophenyl)methyl)piperazine-1-carboxylate], MAGL inhibitors that possess increased selectivity and potency, in mouse behavioral assays of neuropathic pain [chronic constriction injury (CCI) of the sciatic nerve], interoceptive cannabimimetic effects (drug-discrimination paradigm), and locomotor activity in an open field test. MJN110 (1.25 and 2.5 mg/kg) and JZL184 (16 and 40 mg/kg) significantly elevated 2-AG and decreased arachidonic acid but did not affect anandamide in whole brains. Both MAGL inhibitors significantly reduced CCI-induced mechanical allodynia with the following potencies [ED50 (95% confidence limit [CL]) values in mg/kg: MJN110 (0.43 [0.30-0.63]) > JZL184 (17.8 [11.6-27.4])] and also substituted for the potent cannabinoid receptor agonist CP55,940 [2-[(1R,2R,5R)-5-hydroxy-2-(3-hydroxypropyl)cyclohexyl]-5-(2-methyloctan-2-yl)phenol] in the drug-discrimination paradigm [ED50 (95% CL) values in mg/kg: MJN110 (0.84 [0.69-1.02]) > JZL184 (24.9 [14.6-42.5])]; however, these compounds elicited differential effects on locomotor behavior. Similar to cannabinoid 1 (CB1) receptor agonists, JZL184 produced hypomotility, whereas MJN110 increased locomotor behavior and did not produce catalepsy or hypothermia. Although both drugs substituted for CP55,940 in the drug discrimination assay, MJN110 was more potent in reversing allodynia in the CCI model than in producing CP55,940-like effects. Overall, these results suggest that MAGL inhibition may alleviate neuropathic pain, while displaying limited cannabimimetic effects compared with direct CB1 receptor agonists.


Assuntos
Analgésicos/farmacologia , Materiais Biomiméticos/farmacologia , Canabinoides/metabolismo , Inibidores Enzimáticos/farmacologia , Monoacilglicerol Lipases/antagonistas & inibidores , Analgésicos/uso terapêutico , Animais , Benzodioxóis/farmacologia , Benzodioxóis/uso terapêutico , Materiais Biomiméticos/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Carbamatos/farmacologia , Carbamatos/uso terapêutico , Constrição , Endocanabinoides/metabolismo , Inibidores Enzimáticos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Succinimidas/farmacologia , Succinimidas/uso terapêutico
13.
J Neuroinflammation ; 11: 92, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24884664

RESUMO

BACKGROUND: Peri-spinal subarachnoid (intrathecal; i.t.) injection of non-viral naked plasmid DNA encoding the anti-inflammatory cytokine, IL-10 (pDNA-IL-10) suppresses chronic neuropathic pain in animal models. However, two sequential i.t. pDNA injections are required within a discrete 5 to 72-hour period for prolonged efficacy. Previous reports identified phagocytic immune cells present in the peri-spinal milieu surrounding the i.t injection site that may play a role in transgene uptake resulting in subsequent IL-10 transgene expression. METHODS: In the present study, we aimed to examine whether factors known to induce pro-phagocytic anti-inflammatory properties of immune cells improve i.t. IL-10 transgene uptake using reduced naked pDNA-IL-10 doses previously determined ineffective. Both the synthetic glucocorticoid, dexamethasone, and the hexose sugar, D-mannose, were factors examined that could optimize i.t. pDNA-IL-10 uptake leading to enduring suppression of neuropathic pain as assessed by light touch sensitivity of the rat hindpaw (allodynia). RESULTS: Compared to dexamethasone, i.t. mannose pretreatment significantly and dose-dependently prolonged pDNA-IL-10 pain suppressive effects, reduced spinal IL-1ß and enhanced spinal and dorsal root ganglia IL-10 immunoreactivity. Macrophages exposed to D-mannose revealed reduced proinflammatory TNF-α, IL-1ß, and nitric oxide, and increased IL-10 protein release, while IL-4 revealed no improvement in transgene uptake. Separately, D-mannose dramatically increased pDNA-derived IL-10 protein release in culture supernatants. Lastly, a single i.t. co-injection of mannose with a 25-fold lower pDNA-IL-10 dose produced prolonged pain suppression in neuropathic rats. CONCLUSIONS: Peri-spinal treatment with D-mannose may optimize naked pDNA-IL-10 transgene uptake for suppression of allodynia, and is a novel approach to tune spinal immune cells toward pro-phagocytic phenotype for improved non-viral gene therapy.


Assuntos
Anti-Inflamatórios/uso terapêutico , Terapia Genética , Interleucina-10/uso terapêutico , Manose/uso terapêutico , Neuralgia/terapia , Limiar da Dor/fisiologia , Animais , Células Cultivadas , Doença Crônica , Constrição Patológica/complicações , Citocinas/genética , Citocinas/metabolismo , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Interleucina-10/biossíntese , Masculino , Camundongos , Microftalmia/tratamento farmacológico , Microftalmia/metabolismo , Neuralgia/etiologia , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Medula Espinal/fisiologia
14.
Front Mol Neurosci ; 17: 1355281, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38481473

RESUMO

The individual and global burden of migraine is of such significance that there are accelerated efforts to develop new therapies. New migraine therapeutics are needed to address the current deficiencies that exist in the efficacy and adherence rate of approved anti-migraine medications. The recent discovery of the calcitonin gene related peptide as an add-on to the role of serotonin has markedly increased the range of new treatment options for acute and chronic migraine. Despite this, tackling the complexity of migraine disorders requires a complete understanding of its pathophysiology. Preclinical animal models can shed light on disease-related pathophysiology, including migraine. Indeed, the use of animal models has been instrumental in developing many therapeutics. However, an animal model is limited by the predictive and face validity of that model, and this extends to preclinical migraine models. In this review, a summary of the current understanding of the pathophysiology of migraine is given from both a preclinical and clinical perspective, and an emphasis is placed on the animal models of migraine. We will discuss the strengths and pitfalls of common preclinical migraine models as well as experimental research areas to explore further.

15.
ACS Pharmacol Transl Sci ; 7(3): 654-666, 2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38481688

RESUMO

Opioids represent the most extensive category of abused substances in the United States, and the number of fatalities caused by these drugs exceeds those associated with all other drug overdoses combined. The administration of naltrexone, a potent pan-opioid receptor antagonist, to an individual dependent on opioids can trigger opioid withdrawal and induce severe side effects. There is a pressing demand for opioid antagonists free of opioid withdrawal effects. In our laboratory, we have identified a compound with affinity to mu, delta, and kappa opioid receptors in the range of 150-250 nM. This blood-brain barrier (BBB)-permeant compound was metabolically stable in vitro and in vivo. Our in vivo work demonstrated that 1-10 mg/kg intraperitoneal administration of our compound produces moderate efficacy in antagonizing morphine-induced antiallodynia effects in the chemotherapy-induced peripheral neuropathy (CIPN) model. The treatment was well-tolerated and did not cause behavioral changes. We have observed a fast elimination rate of this metabolically stable molecule. Furthermore, no organ toxicity was observed during the chronic administration of the compound over a 14-day period. Overall, we report a novel functional opioid antagonist holds promise for developing an opioid withdrawal therapeutic.

16.
Adv Drug Alcohol Res ; 3: 11622, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38389808

RESUMO

Nicotine is the primary psychoactive component responsible for maintaining tobacco dependence in humans. Chronic pain is often a consequence of tobacco-related pathologies, and the development of a dual therapeutic that could treat chronic pain and tobacco dependence would be advantageous. Epibatidine reliably substitutes for nicotine in the drug discrimination assay, and is a potent analgesic, but has a side-effect profile that limits its therapeutic potential. Thus, considerable efforts to produce epibatidine derivatives are underway. Here we tested three epibatidine derivatives, 2'-fluoro-3'-(4-nitrophenyl)deschloroepibatidine (RTI-7527-102; i.e., RTI-102), 2'-fluorodeschloroepibatidine (RTI-7527-36; i.e., RTI-36), and 3'-(3″-dimethylaminophenyl)-epibatidine (RTI-7527-76; i.e., RTI-76) in both the rat nicotine drug discrimination assay as well as in the rat chronic constriction injury (CCI) of the sciatic nerve neuropathic pain model. Male and female Sprague-Dawley rats were trained on a fixed-ratio 10 schedule to discriminate nicotine (0.32 mg/kg base) from vehicle. All compounds dose-dependently substituted for nicotine, without significant decreases in response rates. In the discrimination assay the rank order potency was RTI-36 > nicotine > RTI-102 > RTI-76. Evidence suggests the α4ß2* subtype is particularly important to nicotine-related abuse potential. Thus, here we utilized the antagonist dihydro-ß-erythroidine (DHßE) to examine relative ß2 subunit contribution. DHßE (3.2 mg/kg, s.c.) antagonized the discriminative stimulus effects of nicotine. However, relative to antagonism of nicotine, DHßE produced less antagonism of RTI-102 and RTI-76 and greater antagonism of RTI-36. It is likely that at nicotinic receptor subunits RTI-102, RTI-76 and RTI-36 possess differing activity. To confirm that the full discriminative stimulus of these compounds was due to nAChR activity beyond the ß2 subunit, we examined these compounds in the presence of the non-selective nicotinic receptor antagonist mecamylamine. Mecamylamine (0.56 mg/kg, s.c.) pretreatment abolished nicotine-paired lever responding for all compounds. In a separate cohort, male and female Sprague-Dawley rats underwent CCI surgery and tested for CCI-induced mechanical allodynia via the von Frey assay. Each compound produced CCI-induced mechanical allodynia reversal. RTI-36 displayed higher potency than either RTI-102 or RTI-76. These novel epibatidine analogs may prove to be useful tools in the fight against nicotine dependence as well as novel neuropathic pain analgesics.

17.
Pharmacol Rep ; 75(4): 937-950, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37243887

RESUMO

BACKGROUND: For many chemotherapy patients peripheral neuropathy is a debilitating side effect. Mitragyna speciosa (kratom) contains the alkaloid mitragynine (MG), which produces analgesia in multiple preclinical pain models. In humans, anecdotal reports suggest cannabidiol (CBD) may enhance kratom-related analgesia. We examined the interactive activity of MG and CBD in a mouse chemotherapy-induced peripheral neuropathy (CIPN) model. We also examined MG + CBD in acute antinociception and schedule-controlled responding assays, as well as examined underlying receptor mechanisms. METHODS: Male and female C57BL/6J mice received a cycle of intraperitoneal (ip) paclitaxel injections (cumulative dose 32 mg/kg). The von Frey assay was utilized to assess CIPN allodynia. In paclitaxel-naïve mice, schedule-controlled responding for food was conducted under a fixed ratio (FR)-10, and hot plate antinociception was examined. RESULTS: MG dose-relatedly attenuated CIPN allodynia (ED50 102.96 mg/kg, ip), reduced schedule-controlled responding (ED50 46.04 mg/kg, ip), and produced antinociception (ED50 68.83 mg/kg, ip). CBD attenuated allodynia (ED50 85.14 mg/kg, ip) but did not decrease schedule-controlled responding or produce antinociception. Isobolographic analysis revealed 1:1, 3:1 MG + CBD mixture ratios additively attenuated CIPN allodynia. All combinations decreased schedule-controlled responding and produced antinociception. WAY-100635 (serotonin 5-HT1A receptor antagonist) pretreatment (0.01 mg/kg, ip) antagonized CBD anti-allodynia. Naltrexone (pan opioid receptor antagonist) pretreatment (0.032 mg/kg, ip) antagonized MG anti-allodynia and acute antinociception but produced no change in MG-induced decreased schedule-controlled behavior. Yohimbine (α2 receptor antagonist) pretreatment (3.2 mg/kg, ip) antagonized MG anti-allodynia and produced no change in MG-induced acute antinociception or decreased schedule-controlled behavior. CONCLUSIONS: Although more optimization is needed, these data suggest CBD combined with MG may be useful as a novel CIPN therapeutic.


Assuntos
Canabidiol , Doenças do Sistema Nervoso Periférico , Camundongos , Humanos , Masculino , Feminino , Animais , Paclitaxel/toxicidade , Canabidiol/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Camundongos Endogâmicos C57BL , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Modelos Animais de Doenças , Dor/tratamento farmacológico
18.
Front Pharmacol ; 13: 881810, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35529444

RESUMO

Cannabinoids, including those found in cannabis, have shown promise as potential therapeutics for numerous health issues, including pathological pain and diseases that produce an impact on neurological processing and function. Thus, cannabis use for medicinal purposes has become accepted by a growing majority. However, clinical trials yielding satisfactory endpoints and unequivocal proof that medicinal cannabis should be considered a frontline therapeutic for most examined central nervous system indications remains largely elusive. Although cannabis contains over 100 + compounds, most preclinical and clinical research with well-controlled dosing and delivery methods utilize the various formulations of Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD), the two most abundant compounds in cannabis. These controlled dosing and delivery methods are in stark contrast to most clinical studies using whole plant cannabis products, as few clinical studies using whole plant cannabis profile the exact composition, including percentages of all compounds present within the studied product. This review will examine both preclinical and clinical evidence that supports or refutes the therapeutic utility of medicinal cannabis for the treatment of pathological pain, neurodegeneration, substance use disorders, as well as anxiety-related disorders. We will predominately focus on purified THC and CBD, as well as other compounds isolated from cannabis for the aforementioned reasons but will also include discussion over those studies where whole plant cannabis has been used. In this review we also consider the current challenges associated with the advancement of medicinal cannabis and its derived potential therapeutics into clinical applications.

19.
Sci Rep ; 12(1): 3344, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35228612

RESUMO

Incomplete functional recovery after peripheral nerve injury (PNI) often results in devastating physical disabilities in human patients. Despite improved progress in surgical and non-surgical approaches, achieving complete functional recovery following PNI remains a challenge. This study demonstrates that phentolamine may hold a significant promise in treating nerve injuries and denervation induced muscle atrophy following PNI. In a sciatic nerve crush injury mouse model, we found that phentolamine treatment enhanced motor and functional recovery, protected axon myelination, and attenuated injury-induced muscle atrophy in mice at 14 days post-injury (dpi) compared to saline treatment. In the soleus of phentolamine treated animals, we observed the downregulation of phosphorylated signal transducer and activator of transcription factor 3 (p-STAT3) as well as muscle atrophy-related genes Myogenin, muscle ring finger 1 (MuRF-1), and Forkhead box O proteins (FoxO1, FoxO3). Our results show that both nerve and muscle recovery are integral components of phentolamine treatment-induced global functional recovery in mice at 14 dpi. Moreover, phentolamine treatment improved locomotor functional recovery in the mice after spinal cord crush (SCC) injury. The fact that phentolamine is an FDA approved non-selective alpha-adrenergic blocker, clinically prescribed for oral anesthesia reversal, hypertension, and erectile dysfunction makes this drug a promising candidate for repurposing in restoring behavioral recovery following PNI and SCC injuries, axonal neuropathy, and muscle wasting disorders.


Assuntos
Traumatismos dos Nervos Periféricos , Neuropatia Ciática , Animais , Axônios/metabolismo , Humanos , Masculino , Camundongos , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Regeneração Nervosa , Fentolamina/uso terapêutico , Recuperação de Função Fisiológica/fisiologia , Nervo Isquiático/lesões
20.
Adv Pharmacol ; 93: 35-76, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35341571

RESUMO

Kratom products have been historically and anecdotally used in south Asian countries for centuries to manage pain and opioid withdrawal. The use of kratom products has dramatically increased in the United States. More than 45 kratom alkaloids have been isolated, yet the overall pharmacology of the individual alkaloids is still not well characterized. The purpose of this chapter is to summarize in vitro and in vivo opioid activities of the primary kratom alkaloid mitragynine and its more potent metabolite 7-hydroxymitragynine. Following are experimental procedures described to characterize opioid receptor activity; receptor binding and functional assays, antinociceptive assays, operant conditioning assays, and respiratory plethysmography. The capacity of kratom alkaloids to confer tolerance and physical dependence as well as their pharmacokinetic properties are also summarized. The data reviewed here suggest that kratom products and mitragynine possess low efficacy agonist activity at the mu-opioid receptor in vivo. In addition, kratom products and mitragynine have been demonstrated to antagonize the effects of high efficacy mu-opioid agonists. The data further suggest that 7-hydroxymitragynine formed in vivo by metabolism of mitragynine may be minimally involved in the overall behavioral profile of mitragynine and kratom, whereas 7-hydroxymitragynine itself, at sufficiently high doses administered exogenously, shares many of the same abuse- and dependence-related behavioral effects associated with traditional opioid agonists. The apparent low efficacy of kratom products and mitragynine at mu-opioid receptors supports the development of these ligands as effective and potentially safe medications for opioid use disorder.


Assuntos
Mitragyna , Transtornos Relacionados ao Uso de Substâncias , Analgésicos Opioides/farmacologia , Humanos , Mitragyna/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA