Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Pharmacol Biochem Behav ; 234: 173688, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38056696

RESUMO

There is accumulating evidence supporting the involvement of tissue-plasminogen activator (tPA) in the mechanisms underlying the effects of morphine and an enriched environment. This study was designed to investigate possible interactive roles of the glutamatergic and the dopaminergic systems regarding hippocampal tPA in the neurobiology of morphine dependence. For this purpose, Wistar albino rats, housed in either a standard- (SE) or an enriched environment (EE) were implanted subcutaneously with morphine (150 mg base) or placebo pellets. Behavioral and somatic signs of morphine abstinence precipitated by an opioid-receptor antagonist naloxone (1 mg/kg, i.p.) 72 h after the pellet implantation were observed individually for 15 min in all groups. Memantine (10 mg/kg i.p.), an antagonist of N-methyl-D-aspartic acid class of glutamatergic receptor-subtype decreased teeth-chattering, ptosis, diarrhea and the loss of body weight. SKF82958 (1 mg/kg, i.p.), a dopamine D1-receptor agonist decreased jumping and ptosis but increased rearing and loss of body weight. On the other hand, co-administration of SKF82958 with memantine prevented some of their effects that occur when administered alone at the same doses. Furthermore, the EE did not change the intensity of morphine abstinence. The level of hippocampal tPA mRNA was found to be lower in the SE morphine abstinence group than in the placebo group and close to the EE morphine abstinence group, whereas there was no significant alteration of its level in the memantine or SKF82958 groups. These findings suggest that the interaction between the glutamatergic and the dopaminergic systems may be an important component of the neurobiology of morphine dependence, and the role of tPA in this interaction should be further investigated.


Assuntos
Dependência de Morfina , Síndrome de Abstinência a Substâncias , Ratos , Animais , Morfina/farmacologia , Naloxona/farmacologia , Memantina/farmacologia , Dependência de Morfina/prevenção & controle , Ratos Wistar , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Peso Corporal
2.
Neurochem Int ; 150: 105157, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34390773

RESUMO

Besides their clinical application, chronic misuse of opioids has often been associated to drug addiction due to their addictive properties, underlying neuroadaptations of AMPA glutamate-receptor-dependent synaptic plasticity. Topiramate (TPM), an AMPAR antagonist, has been used to treat psychostimulants addiction, despite its harmful effects on memory. This study aimed to evaluate the effects of a novel topiramate nanosystem on molecular changes related to morphine reinstatement. Rats were previously exposed to morphine in conditioned place preference (CPP) paradigm and treated with topiramate-chitosan nanoparticles (TPM-CS-NP) or non-encapsulated topiramate in solution (S-TPM) during CPP extinction; following memory performance evaluation, they were re-exposed to morphine reinstatement. While morphine-CPP extinction was comparable among all experimental groups, TPM-CS-NP treatment prevented morphine reinstatement, preserving memory performance, which was impaired by both morphine-conditioning and S-TPM treatment. In the NAc, morphine increased D1R, D2R, D3R, DAT, GluA1 and MOR immunoreactivity. It also increased D1R, DAT, GluA1 and MOR in the dorsal hippocampus. TPM-CS-NP treatment decreased D1R, D3R and GluA1 and increased DAT in the NAc, decreasing GluA1 and increasing D2 and DAT in the dorsal hippocampus. Taken together, we may infer that TPM-CS-NP treatment was able to prevent the morphine reinstatement without memory impairment. Therefore, TPM-CS-NP may be considered an innovative therapeutic tool due to its property to prevent opioid reinstatement because it acts modifying both dopaminergic and glutamatergic neurotransmission, which are commonly related to morphine addiction.


Assuntos
Quitosana/administração & dosagem , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Dependência de Morfina/metabolismo , Nanopartículas/administração & dosagem , Topiramato/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Quimioterapia Combinada , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Masculino , Memória/efeitos dos fármacos , Memória/fisiologia , Morfina/farmacologia , Dependência de Morfina/prevenção & controle , Ratos , Ratos Wistar , Receptores de AMPA/metabolismo , Receptores Dopaminérgicos/metabolismo
3.
Pharm Res ; 38(7): 1263-1278, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34128146

RESUMO

A method to reproducibly mill abuse deterrent oxycodone hydrochloride (HCl) extended release (ER) tablets was developed for a nasal insufflation pharmacokinetic (PK) study. Several comminution methods were explored before determining that a conical mill resulted in controlled milling of tablets to a size range equal to or below 1000 µm. However, milling resulted in significant loss of oxycodone from abuse deterrent oxycodone HCl ER tablets compared to minimal oxycodone loss from oxycodone HCl immediate release (IR) tablets. Characterization of milled tablet powder showed that loss of oxycodone was not attributed to analytical procedures or oxycodone phase change during high intensity milling processes. The content uniformity of oxycodone in the milled tablet powder varied when ER and IR tablets were milled to a particle size distribution equal to or below 500 µm but did not vary when particles were sized above 500 µm to equal to or below 1000 µm. In addition, the initial excipient weight to drug substance weight ratio impacted the amount of oxycodone lost from the respective formulation. However, dissolution demonstrated that when oxycodone HCl ER tablets are milled, differences in excipient weight to drug substance weight ratio and particle size distribution of milled tablets did not result in significantly different release of oxycodone.


Assuntos
Formulações de Dissuasão de Abuso , Analgésicos Opioides/química , Composição de Medicamentos/métodos , Dependência de Morfina/prevenção & controle , Oxicodona/química , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacocinética , Química Farmacêutica , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacocinética , Liberação Controlada de Fármacos , Insuflação , Oxicodona/administração & dosagem , Oxicodona/farmacocinética , Pós , Imagem Individual de Molécula , Comprimidos
4.
Cell Mol Neurobiol ; 41(5): 1131-1143, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33433723

RESUMO

Chronic administration of opioids produces physical dependence and opioid-induced hyperalgesia. Users claim the Thai traditional tea "kratom" and component alkaloid mitragynine ameliorate opioid withdrawal without increased sensitivity to pain. Testing these claims, we assessed the combined kratom alkaloid extract (KAE) and two individual alkaloids, mitragynine (MG) and the analog mitragynine pseudoindoxyl (MP), evaluating their ability to produce physical dependence and induce hyperalgesia after chronic administration, and as treatments for withdrawal in morphine-dependent subjects. C57BL/6J mice (n = 10/drug) were administered repeated saline, or graded, escalating doses of morphine (intraperitoneal; i.p.), kratom alkaloid extract (orally, p.o.), mitragynine (p.o.), or MP (subcutaneously, s.c.) for 5 days. Mice treated chronically with morphine, KAE, or mitragynine demonstrated significant drug-induced hyperalgesia by day 5 in a 48 °C warm-water tail-withdrawal test. Mice were then administered naloxone (10 mg/kg, s.c.) and tested for opioid withdrawal signs. Kratom alkaloid extract and the two individual alkaloids demonstrated significantly fewer naloxone-precipitated withdrawal signs than morphine-treated mice. Additional C57BL/6J mice made physically dependent on morphine were then used to test the therapeutic potential of combined KAE, mitragynine, or MP given twice daily over the next 3 days at either a fixed dose or in graded, tapering descending doses. When administered naloxone, mice treated with KAE, mitragynine, or MP under either regimen demonstrated significantly fewer signs of precipitated withdrawal than control mice that continued to receive morphine. In conclusion, while retaining some liabilities, kratom, mitragynine, and mitragynine pseudoindoxyl produced significantly less physical dependence and ameliorated precipitated withdrawal in morphine-dependent animals, suggesting some clinical value.


Assuntos
Analgésicos Opioides/efeitos adversos , Mitragyna , Dependência de Morfina/prevenção & controle , Alcaloides de Triptamina e Secologanina/administração & dosagem , Alcaloides de Triptamina e Secologanina/síntese química , Síndrome de Abstinência a Substâncias/prevenção & controle , Analgésicos Opioides/administração & dosagem , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dependência de Morfina/metabolismo , Dependência de Morfina/psicologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Alcaloides de Triptamina e Secologanina/efeitos adversos , Alcaloides de Triptamina e Secologanina/isolamento & purificação , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/psicologia
5.
Behav Brain Res ; 402: 113104, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33417990

RESUMO

Oxidative stress and the nitric oxide (NO) pathway are involved in the development of opioid analgesic tolerance and dependence. Simvastatin modulates NO and oxidative stress, so the present study aimed to investigate its effect on the development and expression of morphine analgesic tolerance and withdrawal signs in mice. Morphine tolerance and dependence were induced by twice daily morphine injection (10 mg/kg, s.c.) for 5 consecutive days. Tolerance was assessed by the hot-plate test and dependence by naloxone challenge, on the sixth day. To determine if the NO is involved in the effects of simvastatin, mice were pre-treated with l-arginine (200 mg/kg) or the NO synthesis inhibitors (L-NAME; 30 mg/kg) along with simvastatin (300 mg/kg). The results showed that acute and chronic administration of simvastatin reversed the antinociceptive tolerance of morphine and attenuated withdrawal signs in morphine-dependent mice, and this effect is reversed by l-arginine and augmented by l-NAME. Also, the concentration of NO and oxidative stress factors such as malondialdehyde content, total thiol, and glutathione peroxidase (GPx) activity in brain tissues was evaluated. Chronic administration of simvastatin reduced NO and malondialdehyde, and increased total thiol and GPx levels in the cerebral cortex and hippocampus of morphine-dependent mice which were antagonized by l-arginine, and augmented by l-NAME. In summary, simvastatin attenuates morphine-induced antinociceptive tolerance and withdrawal symptoms, at least partly, through antioxidative properties and nitric oxide pathway.


Assuntos
Antioxidantes/farmacologia , Córtex Cerebral/efeitos dos fármacos , Tolerância a Medicamentos , Dependência de Morfina/prevenção & controle , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia , Síndrome de Abstinência a Substâncias/prevenção & controle , Animais , Hipocampo/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Camundongos , Dependência de Morfina/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo
6.
Neuropsychobiology ; 80(3): 264-270, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33207349

RESUMO

OBJECTIVE: Hydrogen sulfide is an endogenous gaseous mediator that has been indicated to have a role in pain mechanisms. In this study, we aimed to detect brain and spinal cord hydrogen sulfide levels during different phases of tolerance and dependence to morphine and to determine the effects of inhibition of endogenous hydrogen sulfide production on the development of tolerance and dependence. METHODS: Morphine tolerance and dependence was developed by subcutaneous injection of morphine (10 mg/kg) twice daily for 12 days. Physical dependence was determined by counting the jumps for 20 min, which is a withdrawal symptom occurring after a single dose of naloxone (5 mg/kg) administered intraperitoneally (i.p.). Propargylglycine (30 mg/kg, i.p.), a cystathionine-γ-lyase inhibitor, and hydroxylamine (12.5 mg/kg, i.p.), a cystathionine-ß-synthase inhibitor, were used as hydrogen sulfide synthase inhibitors. The tail-flick and hot-plate tests were used to determine the loss of antinociceptive effects of morphine and development of tolerance. RESULTS: It was found that chronic and acute uses of both propargylglycine and hydroxylamine prevented the development of tolerance to morphine, whereas they had no effect on morphine dependence. Chronic and acute administrations of hydrogen sulfide synthase inhibitors did not exert any difference in hydrogen sulfide levels in brain and spinal cords of both morphine-tolerant and -dependent animals. CONCLUSION: It has been concluded that hydrogen sulfide synthase inhibitors may have utility in preventing morphine tolerance.


Assuntos
Comportamento Animal/efeitos dos fármacos , Tolerância a Medicamentos , Inibidores Enzimáticos/farmacologia , Sulfeto de Hidrogênio/metabolismo , Dependência de Morfina/prevenção & controle , Alcinos/farmacologia , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/administração & dosagem , Glicina/análogos & derivados , Glicina/farmacologia , Hidroxilamina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Morfina/administração & dosagem , Entorpecentes/administração & dosagem
7.
Eur J Pharmacol ; 886: 173544, 2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-32896549

RESUMO

The opioid crisis has underscored the urgent need to identify safe and effective therapeutic strategies to overcome opioid-induced liabilities. We recently reported that LY2828360, a slowly signaling G protein-biased cannabinoid CB2 receptor agonist, suppresses neuropathic nociception and attenuates the development of tolerance to the opioid analgesic morphine in paclitaxel-treated mice. Whether beneficial effects of LY2828360 are dependent upon the presence of a pathological pain state are unknown and its impact on unwanted opioid-induced side-effects have never been investigated. Here, we asked whether LY2828360 would produce synergistic anti-allodynic effects with morphine in a paclitaxel model of chemotherapy-induced neuropathic pain and characterized its impact on opioid-induced reward and other unwanted side-effects associated with chronic opioid administration. Isobolographic analysis revealed that combinations of LY2828360 and morphine produced synergistic anti-allodynic effects in suppressing paclitaxel-induced mechanical allodynia. In wildtype (WT) mice, LY2828360 blocked morphine-induced reward in a conditioned place preference assay without producing reward or aversion when administered alone. The LY2828360-induced attenuation of morphine-induced reward was absent in CB2 knockout (CB2KO) mice. In the absence of a neuropathic pain state, LY2828360 partially attenuated naloxone-precipitated opioid withdrawal in morphine-dependent WT mice, and this withdrawal was itself markedly exacerbated in CB2KO mice. Moreover, LY2828360 did not reliably alter morphine-induced slowing of colonic transit or attenuate tolerance to morphine antinociceptive efficacy in the hot plate test of acute nociception. Our results suggest that cannabinoid CB2 receptor activation enhances the therapeutic properties of opioids while attenuating unwanted side-effects such as reward and dependence that occur with sustained opioid treatment.


Assuntos
Analgésicos Opioides/farmacologia , Analgésicos/uso terapêutico , Agonistas de Receptores de Canabinoides/farmacologia , Dependência de Morfina/prevenção & controle , Morfina/farmacologia , Neuralgia/tratamento farmacológico , Purinas/farmacologia , Piranos/farmacologia , Receptor CB2 de Canabinoide/agonistas , Recompensa , Animais , Agonistas de Receptores de Canabinoides/uso terapêutico , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuralgia/induzido quimicamente , Nociceptividade/efeitos dos fármacos , Paclitaxel , Purinas/uso terapêutico , Piranos/uso terapêutico , Receptor CB2 de Canabinoide/genética , Síndrome de Abstinência a Substâncias/tratamento farmacológico
8.
Neurosci Lett ; 724: 134875, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32114118

RESUMO

Orexin neuropeptides are involved in opiate-induced physical dependence and expression of withdrawal signs following drug abstinence. Orexin type-1 receptors (OXR1) are expressed throughout the rostroventrolateral medulla (RVLM), particularly in the lateral paragigantocellularis (LPGi) nucleus. The present study examined whether blocking OXR1 in LPGi region could affect the development of morphine dependence and so behavioral manifestations induced by morphine withdrawal in rats. Male Wistar rats weighing 250-300 g were used. In order to induce drug dependence, morphine was injected subcutaneously (s.c.) (6, 16, 26, 36, 46, 56, and 66 mg/kg, 2 ml/kg) at an interval of 24 h for 7 days. Animals were divided into experimental groups in which the orexin type-1 receptor antagonist, SB-334867 (0.2 µl, 3 mM), or its vehicle were injected into the LPGi nucleus for 7 days before each morphine injection. On day 8, naloxone (2.5 mg/kg, i.p.) was administered and morphine withdrawal behaviors were monitored for 25 min. Our results indicated that the inhibition of OXR1 in LPGi nucleus significantly reduces the development of morphine dependence and behavioral signs elicited by the administration of naloxone in morphine-dependent rats.


Assuntos
Bulbo/fisiologia , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/fisiologia , Dependência de Morfina/prevenção & controle , Morfina/efeitos adversos , Antagonistas dos Receptores de Orexina/administração & dosagem , Animais , Masculino , Bulbo/efeitos dos fármacos , Microinjeções/métodos , Morfina/administração & dosagem , Dependência de Morfina/fisiopatologia , Ratos , Ratos Wistar
10.
Acta Pharmacol Sin ; 40(5): 583-588, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30224637

RESUMO

Increasing preclinical evidence demonstrates that dopamine D3 receptor (D3R) antagonists are a potential option for the treatment of drug addiction. The reinstatement of the addiction can be triggered by environmental stimuli that acquire motivational salience through repeated associations with the drug's effects. YQA14 is a novel D3R antagonist that has exhibited pharmacotherapeutic efficacy in reducing cocaine and amphetamine reward and relapse to drug seeking in mice. In this study we investigated the effects of YQA14 on morphine-induced context-specific locomotor sensitization in mice. We showed that repeated injection of YQA14 (6.25-25 mg/kg every day ip) prior to morphine (10 mg/kg every day sc) not only inhibited the acquisition, but also significantly attenuated the expression of morphine-induced locomotor sensitization. Furthermore, in the expression phase, one single injection of YQA14 (6.25-25 mg/kg, ip) dose-dependently inhibited the expression of morphine-induced behavioral sensitization. Moreover, YQA14 inhibited the expression of morphine-induced behavioral sensitization in wild mice (WT), but not in D3R knockout (D3R-/-) mice in the expression phase. In addition, D3R-/- mice also displayed the reduction in the expression phase compared with WT mice. In summary, this study demonstrates that blockade or knockout of the D3R inhibits morphine-induced behavior sensitization, suggesting that D3R plays an important role in the pathogenesis and etiology of morphine addiction, and it might be a potential target for clinical management of opioid addiction.


Assuntos
Benzoxazóis/uso terapêutico , Antagonistas de Dopamina/uso terapêutico , Dependência de Morfina/prevenção & controle , Morfina/farmacologia , Atividade Motora/efeitos dos fármacos , Piperazinas/uso terapêutico , Receptores de Dopamina D3/antagonistas & inibidores , Animais , Benzoxazóis/administração & dosagem , Benzoxazóis/farmacocinética , Antagonistas de Dopamina/administração & dosagem , Antagonistas de Dopamina/farmacocinética , Comportamento de Procura de Droga , Injeções Intraperitoneais , Masculino , Camundongos Knockout , Morfina/administração & dosagem , Dependência de Morfina/etiologia , Dependência de Morfina/psicologia , Piperazinas/administração & dosagem , Piperazinas/farmacocinética , Receptores de Dopamina D3/genética
11.
Pharmacol Rep ; 70(5): 955-962, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30103176

RESUMO

BACKGROUND: Morphine - the main pillar of nociceptive pain management - systemic use is associated with development of tolerance and dependence. Tolerance and dependence lay a heavy burden in clinical pain management settings. An added weight to this dilemma is that effective, safe, and tolerable solution to this problem is still beyond reach. Antidepressants were reported as possible alleviators of opioid tolerance and dependence. One of the increasingly used antidepressant in clinical practice is bupropion given its high safety and tolerability profile. METHODS: The study was performed on male Balb-c mice weighing 20-30g. Hot plate test was used for assessment of bupropion (5mg/kg, ip) possible analgesic activity and enhancement of morphine acute analgesia (1 and 5mg/kg, sc). Repeated morphine (5mg/kg, sc) administration for 9days developed tolerance and dependence, bupropion (5mg/kg, ip) was concurrently administered to evaluate its potential to modulate these processes. We also biochemically analyzed bupropion effect on these phenomena through modulation of neurotransmitters (glutamate and norepinephrine), inflammatory status (nitric oxide), and pro-antioxidant balance (malondialdehyde and reduced glutathione). RESULTS: Bupropion was devoid of intrinsic analgesic activity and did not enhance morphine acute analgesia. However, bupropion significantly attenuated morphine tolerance and dependence development and abstinence syndrome with corresponding suppression of morphine induced changes in glutamate, norepinephrine, inflammatory status, and prooxidant-antioxidant balance. CONCLUSION: Bupropion efficacy in attenuation of morphine tolerance and dependence with its high safety and tolerability profile provide an alternative option to conventional agents e.g., ketamine and clonidine to modulate these phenomena.


Assuntos
Encéfalo/metabolismo , Bupropiona/farmacologia , Tolerância a Medicamentos , Ácido Glutâmico/metabolismo , Inflamação/metabolismo , Dependência de Morfina/prevenção & controle , Morfina/antagonistas & inibidores , Norepinefrina/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Glutationa/metabolismo , Masculino , Malondialdeído , Camundongos , Morfina/farmacologia , Naloxona/farmacologia , Óxido Nítrico/metabolismo , Compostos Organofosforados/metabolismo , Medição da Dor/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/prevenção & controle
12.
Pharmacol Biochem Behav ; 170: 71-78, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29782941

RESUMO

Adaptations within the nucleus accumbens (NAc) and caudate nucleus (CN) dopamine neurotransmission are involved in behavioral sensitization and enhanced incentive motivation towards drug paired stimuli which lead to drug addiction. Serotonin (5-hydroxytryptamine; 5-HT) can modulate dopamine neurotransmission to reduce rewarding effects of drugs of abuse. A recent study from our laboratory shows that rewarding effects of morphine are inhibited in rats co-treated with buspirone. To understand the neurochemical mechanism involved in morphine addiction and its inhibition with buspirone, present study determines the effects of buspirone, morphine and their co-administration on the metabolism of serotonin and dopamine in the NAc and CN. We find that rewarding effects of morphine are associated with an enhancement and attenuation of dopamine metabolism, respectively in the CN and NAc. Serotonin metabolism is enhanced in both regions. Co-administration of buspirone not only prevents rewarding effects of morphine, but its effects on the metabolism of dopamine and serotonin in the NAc and CN are also reversed. Results suggest that 5-HT1A receptor dependent modulation of dopamine neurotransmission in the CN and NAc is involved in the modulation of the rewarding effects of morphine in buspirone co-treated animals. The findings documenting an important role of 5-HT1A receptors in drug addiction suggest that synthetic opioid drugs with agonist activity of 5-HT1A receptors may prove non addictive analgesics.


Assuntos
Buspirona/farmacologia , Corpo Estriado/metabolismo , Dopamina/metabolismo , Dependência de Morfina/metabolismo , Morfina/farmacologia , Recompensa , Serotonina/metabolismo , Animais , Núcleo Caudado/metabolismo , Masculino , Dependência de Morfina/prevenção & controle , Núcleo Accumbens/metabolismo , Ratos , Ratos Wistar , Receptor 5-HT1A de Serotonina/fisiologia
13.
Exp Neurol ; 303: 134-141, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29453978

RESUMO

Opioid-induced neuroinflammation and the nitric oxide (NO) signal-transduction pathway are involved in the development of opioid analgesic tolerance. The antidepressant venlafaxine (VLF) modulates NO in nervous tissues, and so we investigated its effect on induced tolerance to morphine, neuroinflammation, and oxidative stress in mice. Tolerance to the analgesic effects of morphine were induced by injecting mice with morphine (50 mg/kg) once a day for three consecutive days; the effect of co-administration of VLF (5 or 40 mg/kg) with morphine was similarly tested in a separate group. To determine if the NO precursor l-arginine hydrochloride (l-arg) or NO are involved in the effects rendered by VLF, animals were pre-treated with l-arg (200 mg/kg), or the NO synthesis inhibitors N(ω)-nitro-l-arginine methyl ester (L-NAME; 30 mg/kg) or aminoguanidine hydrochloride (AG; 100 mg/kg), along with VLF (40 mg/kg) for three days before receiving morphine for another three days. Nociception was assessed with a hot-plate test on the fourth day, and the concentration of tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1ß), interleukin-6 (IL-6), interleukin-10, brain-derived neurotrophic factor, NO, and oxidative stress factors such as total thiol, malondialdehyde content, and glutathione peroxidase (GPx) activity in the brain was also determined. Co-administration of VLF with morphine attenuated morphine-induced analgesic tolerance and prevented the upregulation of proinflammatory cytokines (TNF-α, IL-1ß, and IL-6), NO, and malondialdehyde in brains of mice with induced morphine tolerance; chronic VLF administration inhibited this decrease in brain-derived neurotrophic factor, total thiol, and GPx levels. Moreover, repeated administration of l-arg before receipt of VLF antagonized the effects induced by VLF, while L-NAME and AG potentiated these effects. VLF attenuates morphine-induced analgesic tolerance, at least partly because of its anti-inflammatory and antioxidative properties. VLF also appears to suppress the development of morphine-induced analgesic tolerance through an l-arg-NO-mediated mechanism.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Citocinas/metabolismo , Dependência de Morfina/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Cloridrato de Venlafaxina/farmacologia , Analgésicos Opioides/efeitos adversos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Glutationa Peroxidase/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Morfina/efeitos adversos , NG-Nitroarginina Metil Éster/farmacologia , Nitritos/metabolismo , Medição da Dor
14.
Curr Med Res Opin ; 34(5): 893-901, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29368961

RESUMO

BACKGROUND: The development and use of abuse-deterrent (AD) opioids is part of a multifaceted strategy to reduce misuse, abuse, and diversion, while maintaining access for patients with severe pain who may benefit from their analgesic efficacy. Morphine AD, extended-release (ER), injection-molded tablets (morphine-ADER-IMT; ARYMO ER; Egalet US Inc., Wayne, PA) is approved by the FDA as an AD opioid. As part of the characterization of AD opioids, assessments of their human abuse potential (HAP) are required. Evidence from HAP studies can guide clinicians on the use of AD opioids in clinical practice. Herein, we describe HAP study design, and how specific AD features can impact the conduct of a study and interpretation of its results. OBJECTIVES: To review the design features and results of the oral and intranasal HAP studies with morphine-ADER-IMT in order to improve the understanding of key elements of HAP studies of AD opioids. CONCLUSIONS: Results from HAP studies with morphine-ADER-IMT and other AD opioids suggest that key study design features include the release profile (immediate-release vs extended-release) of the positive control, study drug doses, and the way the products are manipulated. These elements can directly impact the outcomes of the pharmacokinetic and pharmacodynamic (e.g. Maximum Drug Liking, Overall Drug Liking, and Take Drug Again) results. When evaluating HAP studies, it is important to understand study design features to assist in the interpretation of the results and understand the clinical relevance of the data to help guide clinical decision-making about the use of AD opioids.


Assuntos
Analgésicos Opioides/administração & dosagem , Morfina/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Administração Intranasal , Analgésicos Opioides/efeitos adversos , Preparações de Ação Retardada , Humanos , Injeções , Morfina/efeitos adversos , Dependência de Morfina/epidemiologia , Dependência de Morfina/prevenção & controle , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Dor/tratamento farmacológico , Comprimidos
15.
Addict Biol ; 23(5): 1055-1066, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-28961365

RESUMO

Accumulating evidence indicated that minocycline, a glial cell modulator, is able to modify a variety of morphine effects. Here, we investigated minocycline effects on electrical activity of nucleus accumbens (NAc) neurons using single unit recording in urethane-anesthetized rats. In addition, we investigated whether minocycline can modify the effects of morphine on NAc neural activity during reinstatement of morphine-seeking behavior. Minocycline increased the NAc firing activity in intact animals. Electrophysiological recording in morphine-treated animals was performed, following the acquisition of morphine-induced conditioned place preference (5 mg/kg, s.c., 3 days) and a drug-free extinction period. In acutely minocycline- treated animals, the neurons were recorded for 40 minutes following a single injection of either minocycline (50 µg/5 µl, i.c.v.) or saline. Then a priming dose of morphine (1 mg/kg, s.c.) was injected while the recording was continued for an additional 40 minutes. Minocycline significantly increased the firing rates of neurons and significantly modified morphine inhibitory effects on NAc neurons. In subchronically minocycline-treated groups, the rats were given daily injections of minocycline (50 µg/5 µl, i.c.v) during the extinction period. Then, on the reinstatement day, NAc neurons were recorded for 10 minutes, the priming dose of morphine was administered and the recording was continued for 45 minutes. Our results showed the failure of minocycline to significantly modify the inhibitory effects of morphine. In conclusion, our findings indicated that minocycline modifies morphine-induced decreases in the firing rates of NAc neurons in the reinstatement phase.


Assuntos
Minociclina/farmacologia , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Animais , Antibacterianos/farmacologia , Condicionamento Clássico , Modelos Animais de Doenças , Comportamento de Procura de Droga/efeitos dos fármacos , Eletrodiagnóstico/métodos , Masculino , Dependência de Morfina/prevenção & controle , Ratos , Ratos Wistar
16.
Artigo em Inglês | MEDLINE | ID: mdl-29237389

RESUMO

BACKGROUND: Severe pain reduces quality of life of patients with various diseases, often because chronic morphine therapy results in reduced analgesic effectiveness, or tolerance, leading to escalating doses and distressing adverse effects. Nitric oxide (NO) plays a role in morphine tolerance and dependence. OBJECTIVE: Venlafaxine, an antidepressant, is known to modulate nitric oxide (NO) pathway in nervous tissues. In the present study, the effect of systemic venlafaxine (VLF) on the development of morphine tolerance and dependence, acute morphine-induced antinociception, and the probable involvement of the L-arginine/NO/cGMP pathway in these effects were investigated in mice. METHODS: Animals developed tolerance to the antinociceptive effect of morphine (50 mg/kg, s.c. daily) for 3 consecutive days. NO modulators like L-NAME (NO synthase inhibitor) and L-Arginine (L-Arg, substrate for NO synthase), sildenafil (cGMP-PDE inhibitor) alone or in combination with venlafaxine were used. RESULTS: The results showed that i.p. administration of VLF (5-40 mg/kg) produced antinociceptive effect in a dose-dependent way. Pretreatment with L-Arg (200 mg/kg, i.p.) reversed the antinociception and L-NAME (30 mg/kg, i.p.) and sildenafil (10 mg/kg, i.p.) potentiated the antinociceptive effect. Moreover, co-administration of VLF in non-effective dose (5 mg/kg) with morphine, potentiated acute morphine-induced analgesia (5 mg/kg, s.c.). This effect was antagonized by L-arginine (200 mg/kg, i.p.) and potentiated by L-NAME (30 mg/kg, i.p.) and sildenafil (10 mg/kg, i.p.). On the other hand, VLF was prevented the development of morphine antinociceptive tolerance and dependence. These effects were antagonized by L-arginine (200 mg/kg, i.p.) and potentiated by L-NAME (30 mg/kg, i.p.) and sildenafil (10 mg/kg, i.p.). CONCLUSION: Our data suggest that the combination of VLF with morphine may be a relevant therapeutic implication to manage pain even when tolerance to morphine exists. Moreover, our data demonstrates the involvement of L-Arg/NO/cGMP pathway in the prevention of morphine tolerance and dependence by venlafaxine.


Assuntos
Analgésicos Opioides/toxicidade , Arginina/metabolismo , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , GMP Cíclico/metabolismo , Tolerância a Medicamentos , Dependência de Morfina/prevenção & controle , Morfina/toxicidade , Óxido Nítrico/metabolismo , Dor Nociceptiva/prevenção & controle , Cloridrato de Venlafaxina/farmacologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Masculino , Camundongos , Dependência de Morfina/metabolismo , Dependência de Morfina/fisiopatologia , Dependência de Morfina/psicologia , Dor Nociceptiva/metabolismo , Dor Nociceptiva/fisiopatologia , Dor Nociceptiva/psicologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
17.
Mol Pharmacol ; 93(2): 49-62, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29192123

RESUMO

The CB2 cannabinoid agonist LY2828360 lacked both toxicity and efficacy in a clinical trial for osteoarthritis. Whether LY2828360 suppresses neuropathic pain has not been reported, and its signaling profile is unknown. In vitro, LY2828360 was a slowly acting but efficacious G protein-biased CB2 agonist, inhibiting cAMP accumulation and activating extracellular signal-regulated kinase 1/2 signaling while failing to recruit arrestin, activate inositol phosphate signaling, or internalize CB2 receptors. In wild-type (WT) mice, LY2828360 (3 mg/kg per day i.p. × 12 days) suppressed chemotherapy-induced neuropathic pain produced by paclitaxel without producing tolerance. Antiallodynic efficacy of LY2828360 was absent in CB2 knockout (KO) mice. Morphine (10 mg/kg per day i.p. × 12 days) tolerance developed in CB2KO mice but not in WT mice with a history of LY2828360 treatment (3 mg/kg per day i.p. × 12 days). LY2828360-induced antiallodynic efficacy was preserved in WT mice previously rendered tolerant to morphine (10 mg/kg per day i.p. × 12 days), but it was absent in morphine-tolerant CB2KO mice. Coadministration of LY2828360 (0.1 mg/kg per day i.p. × 12 days) with morphine (10 mg/kg per day × 12 days) blocked morphine tolerance in WT but not in CB2KO mice. WT mice that received LY2828360 coadministered with morphine exhibited a trend (P = 0.055) toward fewer naloxone-precipitated jumps compared with CB2KO mice. In conclusion, LY2828360 is a slowly signaling, G protein-biased CB2 agonist that attenuates chemotherapy-induced neuropathic pain without producing tolerance and may prolong effective opioid analgesia while reducing opioid dependence. LY2828360 may be useful as a first-line treatment in chemotherapy-induced neuropathic pain and may be highly efficacious in neuropathic pain states that are refractive to opioid analgesics.


Assuntos
Analgésicos Opioides/administração & dosagem , Agonistas de Receptores de Canabinoides/farmacologia , Tolerância a Medicamentos , Proteínas de Ligação ao GTP/metabolismo , Dependência de Morfina/prevenção & controle , Morfina/administração & dosagem , Neuralgia/prevenção & controle , Purinas/farmacologia , Receptor CB2 de Canabinoide/agonistas , Transdução de Sinais , Analgésicos Opioides/efeitos adversos , Animais , Antineoplásicos/administração & dosagem , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/efeitos adversos , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Paclitaxel/administração & dosagem , Purinas/administração & dosagem , Receptor CB2 de Canabinoide/metabolismo , Síndrome de Abstinência a Substâncias
18.
Drug Res (Stuttg) ; 68(1): 33-37, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28847022

RESUMO

BACKGROUND: Dependence and tolerance to morphine are major problems which limit its chronic clinical application. PURPOSE: This study was aimed to investigate the attenuation effect of Cerebrolysin, a mixture of potent growth factors (BDNF, GDNF, NGF, CNTF etc,), on the development of Morphine-induced dependence and tolerance. METHODS: Male Wistar rats were selected randomly and divided into different groups (n=8) including: a control group, groups received additive doses of morphine (5-25 mg/kg, ip, at an interval of 12 h until tolerance completion), and groups pretreated with Cerebrolysin (40, 80 and 160 mg/kg, ip, before morphine administration). Development of tolerance was assessed by tail-flick test and the attenuation effect of Cerebrolysin on morphine-induced dependence was evaluated after injection of naloxone (4 mg/kg, ip, 12 h after the morning dose of morphine). Seven distinct withdrawal signs including: jumping, rearing, genital grooming, abdominal writhing, wet dog shake and teeth grinding were recorded for 45 min and total withdrawal score (TWS) was calculated. RESULTS: Results showed that administration of Cerebrolysin could prolonged development (10 and 14 days in administration of 80 mg/kg and 160 mg/kg Cerebrolysin) and completion (4, 10 and 14 days in administration of 40, 80 and 160 mg/kg Cerebrolysin, respectively) of tolerance. Results also indicated that administration of Cerebrolysin (40, 80 and 160 mg/kg) could significantly decreased the TWS value (62±2, 77±4 and 85±6%, respectively). CONCLUSION: In conclusion, it was found that pretreatment with Cerebrolysin could attenuated morphine-induced tolerance and dependence.


Assuntos
Aminoácidos/uso terapêutico , Analgésicos Opioides/efeitos adversos , Dependência de Morfina/prevenção & controle , Morfina/efeitos adversos , Fármacos Neuroprotetores/uso terapêutico , Aminoácidos/farmacologia , Animais , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Humanos , Masculino , Dependência de Morfina/etiologia , Fármacos Neuroprotetores/farmacologia , Dor/tratamento farmacológico , Medição da Dor , Ratos , Ratos Wistar , Fatores de Tempo
19.
Neurosci Lett ; 659: 110-114, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-28867589

RESUMO

This study was designed to examine whether maternal swimming exercise during pregnancy would attenuate prenatally morphine-induced anxiety, depression and voluntary consumption of morphine in the pubertal male and female rat offspring. Pregnant rats during the development of morphine dependence were allowed to swim (30-45min/d, 3days per a week) on gestational days 11-18. Then, the pubertal male and female rat offspring were tested for the elevated plus-maze (EPM), sucrose preference test (SPT) and voluntary morphine consumption using a two-bottle choice (TBC) paradigm. The results showed that male and female rat offspring born of the swimmer morphine-dependent mothers exhibited an increase in EPM open arm time and entries, higher levels of sucrose preference than their sedentary control mothers. Voluntary consumption of morphine was less in the male and female rat offspring born of the swimmer morphine-dependent mothers as compared with their sedentary control mothers during three periods of the intake of drug. Thus, swimming exercise in pregnant morphine dependent mothers decreased anxiety, depressive-like behavior and also the voluntary morphine consumption in the pubertal male and female offspring, which may prevent prenatally morphine-induced behavioral sensitization in offspring.


Assuntos
Ansiedade/prevenção & controle , Depressão/prevenção & controle , Dependência de Morfina/prevenção & controle , Dependência de Morfina/terapia , Condicionamento Físico Animal/fisiologia , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Maturidade Sexual , Natação/fisiologia , Animais , Ansiedade/complicações , Depressão/complicações , Feminino , Preferências Alimentares , Masculino , Aprendizagem em Labirinto , Morfina/administração & dosagem , Dependência de Morfina/complicações , Gravidez , Ratos , Autoadministração
20.
Biomed Pharmacother ; 93: 406-411, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28654797

RESUMO

BACKGROUND: Tolerance to analgesic effects of opioids and dependence to them are main concerns in the treatment of chronic pain conditions, limiting clinical application of these drugs. This study aimed to evaluate the effect of simvastatin on the morphine-induced tolerance and dependence in mice. MATERIAL AND METHODS: For this purpose, mice were treated with either daily morphine (20 mg/kg, s.c.) alone, or in combination with simvastatin (2.5, 5 and 10mg/kg, i.p.), for 9 continuous days. Antinociceptive effect of morphine was assessed through measuring latency time withdrawal of paw exposed to thermal stimulus, in the hot plate test. Naloxone-precipitated morphine withdrawal (5mg/kg, i.p.), was used for dependence evaluation. Changes in brain gene expression levels of induced nitric oxide synthase (iNOS), astroglia marker, glial fibrillary acidic protein (GFAP), ionized calcium-binding protein (Iba1) a microglia activation marker, a pro-inflammatory mediator and tumor necrosis alpha (TNF-α) were measured after withdrawal by real-time polymerase chain reaction (RT-PCR). RESULTS: Behavioral tests indicated that latency time increased after morphine treatment in the hot plate test. However, this effect decreased on day 7, demonstrating tolerance to antinociceptive effect of morphine. Reduced anti-nociceptive effect of morphine was returned in animals treated with simvastatin (5 and 10mg/kg) in combination with morphine. Simvastatin (5 and 10mg/kg) attenuated morphine dependence as indicated by a less severe antagonist-precipitated withdrawal syndrome. Administration of naloxone was associated with the increased expression of TNF-α, GFAP, Iba1 and iNOS in the brain samples of morphine dependent mice, while the nine days treatment with both 5 and 10mg/kg simvastatin reduced such changes. CONCLUSION: The obtained results showed that the protective effects of simvastatin against both tolerance to nociceptive effects of morphine as well as withdrawal-induced behavioral profile are meaningful. Inhibition of glia activity as well as antioxidant effects of pharmaceutical simvastatin further proves its neuroprotective property.


Assuntos
Tolerância a Medicamentos , Dependência de Morfina/tratamento farmacológico , Dependência de Morfina/prevenção & controle , Sinvastatina/uso terapêutico , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Proteínas de Ligação ao Cálcio , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Proteínas dos Microfilamentos , Morfina/administração & dosagem , Morfina/farmacologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sinvastatina/farmacologia , Síndrome de Abstinência a Substâncias/complicações , Síndrome de Abstinência a Substâncias/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...