Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Drug Chem Toxicol ; 45(1): 250-261, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31656103

RESUMO

Long-term morphine use for therapeutic approaches may lead to serious side effects. Several studies have suggested opioid antagonist and antioxidant therapy for reducing adverse effects of morphine. Cinnamaldehyde has a potent anti-oxidant property. In this study, separate and combined effects of cinnamaldehyde and naloxone (an opioid receptor antagonist) on behavioral changes and cerebellar histological and biochemical outcomes were investigated after long-term morphine administration. Seventy-eight rats were divided into two major morphine-treated and morphine-untreated groups. Morphine-treated group was subdivided into seven subgroups for receiving vehicle, normal saline, cinnamaldehyde (1.25, 5, and 20 mg/kg), naloxone, and cinnamaldehyde plus naloxone before morphine. Morphine-untreated group was subdivided into six subgroups and treated with vehicle, cinnamaldehyde (1.25, 5, and 20 mg/kg), naloxone, and their combination. Chemical compounds were administered for 28 consecutive days. Behavioral tests including footprint, rotarod, and beam balance tests were employed. Histopathological and biochemical alterations of cerebellum were determined. Body and cerebellum weights, stride width, time spent on the rotarod, Purkinje cell number, thickness of molecular and granular layers, superoxide dismutase (SOD), and total antioxidant capacity (TAC) decreased as a result of administrating morphine. Morphine increased beam transverse time, malondealdehyde (MDA), tumor necrosis factor-α (TNF-α), and caspase-3 levels. Histopathological changes such as cellular vacuolation and loss were also produced as a result of treatment with morphine. Cinnamaldehyde, naloxone, and their combination treatments improved all the above-mentioned alterations induced by morphine. We concluded that cinnamaldehyde produced a neuroprotective effect through anti-oxidant, anti-inflammatory, apoptotic, and probably naloxone-sensitive opioid receptor interaction mechanisms.


Assuntos
Morfina , Naloxona , Acroleína/análogos & derivados , Animais , Cerebelo , Morfina/toxicidade , Naloxona/toxicidade , Antagonistas de Entorpecentes/toxicidade , Ratos
2.
Psychopharmacology (Berl) ; 237(2): 375-384, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31712968

RESUMO

RATIONALE: Acute naloxone-precipitated morphine withdrawal (MWD) produces a conditioned place aversion (CPA) in rats even after one or two exposures to high-dose (20 mg/kg, sc) morphine followed 24-h later by naloxone (1 mg/kg, sc). However, the somatic withdrawal reactions produced by acute naloxone-precipitated MWD in rats have not been investigated. A recently discovered fatty acid amide, N-oleoylglycine (OlGly), which has been suggested to act as a fatty acid amide hydrolase (FAAH) inhibitor and as a peroxisome proliferator-activated receptor alpha (PPARα) agonist, was previously shown to interfere with a naloxone-precipitated MWD-induced CPA in rats. OBJECTIVES: The aims of these studies were to examine the somatic withdrawal responses produced by acute naloxone-precipitated MWD and determine whether OlGly can also interfere with these responses. RESULTS: Here, we report that following two exposures to morphine (20 mg/kg, sc) each followed by naloxone (1 mg/kg, sc) 24 h later, rats display nausea-like somatic reactions of lying flattened on belly, abdominal contractions and diarrhea, and display increased mouthing movements and loss of body weight. OlGly (5 mg/kg, ip) interfered with naloxone-precipitated MWD-induced abdominal contractions, lying on belly, diarrhea and mouthing movements in male Sprague-Dawley rats, by both a cannabinoid 1 (CB1) and a PPARα mechanism of action. Since these withdrawal reactions are symptomatic of nausea, we evaluated the potential of OlGly to interfere with lithium chloride (LiCl)-induced and MWD-induced conditioned gaping in rats, a selective measure of nausea; the suppression of MWD-induced gaping reactions by OlGly was both CB1 and PPARα mediated. CONCLUSION: These results suggest that the aversive effects of acute naloxone-precipitated MWD reflect nausea, which is suppressed by OlGly.


Assuntos
Glicina/análogos & derivados , Morfina/efeitos adversos , Naloxona/toxicidade , Antagonistas de Entorpecentes/toxicidade , Náusea/tratamento farmacológico , Ácidos Oleicos/uso terapêutico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Animais , Feminino , Glicina/farmacologia , Glicina/uso terapêutico , Masculino , Sintomas Inexplicáveis , Dependência de Morfina/tratamento farmacológico , Dependência de Morfina/fisiopatologia , Náusea/induzido quimicamente , Náusea/fisiopatologia , Ácidos Oleicos/farmacologia , Ratos , Ratos Sprague-Dawley , Musaranhos , Síndrome de Abstinência a Substâncias/etiologia , Síndrome de Abstinência a Substâncias/fisiopatologia
3.
Nat Neurosci ; 22(7): 1053-1056, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31209376

RESUMO

The lateral habenula encodes aversive stimuli contributing to negative emotional states during drug withdrawal. Here we report that morphine withdrawal in mice leads to microglia adaptations and diminishes glutamatergic transmission onto raphe-projecting lateral habenula neurons. Chemogenetic inhibition of this circuit promotes morphine withdrawal-like social deficits. Morphine withdrawal-driven synaptic plasticity and reduced sociability require tumor necrosis factor-α (TNF-α) release and neuronal TNF receptor 1 activation. Hence, habenular cytokines control synaptic and behavioral adaptations during drug withdrawal.


Assuntos
Citocinas/fisiologia , Habenula/fisiologia , Morfina/efeitos adversos , Comportamento Social , Síndrome de Abstinência a Substâncias/fisiopatologia , Transmissão Sináptica/fisiologia , Adaptação Psicológica , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/fisiologia , Naloxona/toxicidade , Plasticidade Neuronal , Distribuição Aleatória , Receptores de Glutamato/análise , Receptores de N-Metil-D-Aspartato/análise , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Síndrome de Abstinência a Substâncias/psicologia , Fator de Necrose Tumoral alfa/fisiologia
4.
Psychopharmacology (Berl) ; 236(9): 2623-2633, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30993360

RESUMO

RATIONALE: Oleoyl glycine (OlGly), a recently discovered fatty acid amide that is structurally similar to N- acylethanolamines, which include the endocannabinoid, anandamide (AEA), as well as endogenous peroxisome proliferator-activated receptor alpha (PPARα) agonists oleoylethanolamide (OEA) and palmitoylethanolamide (PEA), has been shown to interfere with nicotine reward and dependence in mice. OBJECTIVES AND METHODS: Behavioral and molecular techniques were used to investigate the ability of OlGly to interfere with the affective properties of morphine and morphine withdrawal (MWD) in male Sprague-Dawley rats. RESULTS: Synthetic OlGly (1-30 mg/kg, intraperitoneal [ip]) produced neither a place preference nor aversion on its own; however, at doses of 1 and 5 mg/kg, ip, it blocked the aversive effects of MWD in a place aversion paradigm. This effect was reversed by the cannabinoid 1 (CB1) receptor antagonist, AM251 (1 mg/kg, ip), but not the PPARα antagonist, MK886 (1 mg/kg, ip). OlGly (5 or 30 mg/kg, ip) did not interfere with a morphine-induced place preference or reinstatement of a previously extinguished morphine-induced place preference. Ex vivo analysis of tissue (nucleus accumbens, amygdala, prefrontal cortex, and interoceptive insular cortex) collected from rats experiencing naloxone-precipitated MWD revealed that OlGly was selectively elevated in the nucleus accumbens. MWD did not modify levels of the endocannabinoids 2-AG and AEA, nor those of the PPARα ligands, OEA and PEA, in any region evaluated. CONCLUSION: Here, we show that OlGly interferes with the aversive properties of acute naloxone-precipitated morphine withdrawal in rats. These results suggest that OlGly may reduce the impact of MWD and may possess efficacy in treating opiate withdrawal.


Assuntos
Analgésicos Opioides/efeitos adversos , Glicina/análogos & derivados , Morfina/efeitos adversos , Naloxona/toxicidade , Ácidos Oleicos/administração & dosagem , Recompensa , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Relação Dose-Resposta a Droga , Glicina/administração & dosagem , Glicina/metabolismo , Masculino , Camundongos , Antagonistas de Entorpecentes/toxicidade , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Ácidos Oleicos/metabolismo , Ratos , Ratos Sprague-Dawley , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/psicologia
5.
Cancer Chemother Pharmacol ; 79(2): 431-434, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28011980

RESUMO

PURPOSE: Irinotecan-induced gut toxicity is mediated in part by Toll-Like receptor 4 (TLR4) signalling. The primary purpose of this preclinical study was to determine whether blocking TLR4 signalling by administering (-)-naloxone, a TLR4 antagonist, would improve irinotecan-induced gut toxicity. Our secondary aim was to determine the impact of (-)-naloxone on tumour growth. METHODS: Female Dark Agouti (DA) tumour-bearing rats were randomly assigned to four treatments (n = 6 in each); control, (-)-naloxone (100 mg/kg oral gavage at -2, 24, 48, and 72 h), irinotecan (175 mg/kg intraperitoneal at 0 h), and (-)-naloxone and irinotecan. Body weight and tumour growth were measured daily, and diarrhoea incidence and severity were recorded 4× per day up to 72 h post-treatment. RESULTS: At 72 h, all rats that received irinotecan lost weight compared to controls (p = 0.03). In addition, rats that received (-)-naloxone and irinotecan lost significantly more weight compared to controls (p < 0.005) than irinotecan only compared to controls (p = 0.001). (-)-Naloxone did not attenuate irinotecan-induced severe diarrhoea at 48 and 72 h. Finally, (-)-naloxone caused increased tumour growth compared to control at 72 h (p < 0.05) and significantly reduced the efficacy of irinotecan (p = 0.001). CONCLUSIONS: (-)-Naloxone in our preclinical model was unable to block irinotecan-induced gut toxicity and decreased the efficacy of irinotecan. As (-)-naloxone-oxycodone combination is used for cancer pain, this may present a potential safety concern for patients receiving (-)-naloxone-oxycodone and irinotecan concurrently and requires further investigation.


Assuntos
Camptotecina/análogos & derivados , Naloxona/toxicidade , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Camptotecina/toxicidade , Dor do Câncer/tratamento farmacológico , Diarreia/induzido quimicamente , Diarreia/prevenção & controle , Feminino , Irinotecano , Naloxona/farmacologia , Ratos , Receptor 4 Toll-Like/fisiologia
6.
Brain Res ; 1510: 22-37, 2013 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-23399681

RESUMO

Peroxizome proliferator-activated receptor gamma (PPARγ) is highly expressed in the central nervous system where it modulates numerous gene transcriptions. Nitric oxide synthase (NOS) expression could be modified by simulation of PPARγ which in turn activates nitric oxide (NO)/soluble guanylyl-cyclase (sGC)/cyclic guanosine mono phosphate (cGMP) pathway. It is well known that NO/cGMP pathway possesses pivotal role in the development of opioid dependence and this study is aimed to investigate the effect of PPARγ stimulation on opioid dependence in mice as well as human glioblastoma cell line. Pioglitazone potentiated naloxone-induced withdrawal syndrome in morphine dependent mice in vivo. While selective inhibition of PPARγ, neuronal NOS or GC could reverse the pioglitazone-induced potentiation of morphine withdrawal signs; sildenafil, a phosphodiesterase-5 inhibitor amplified its effect. We also showed that nitrite levels in the hippocampus were significantly elevated in pioglitazone-treated morphine dependent mice. In the human glioblastoma (U87) cell line, rendered dependent to morphine, cAMP levels did not show any alteration after chronic pioglitazone administration while cGMP measurement revealed a significant rise. We were unable to show a significant alteration in neuronal NOS mRNA expressions by pioglitazone in mice hippocampus or U87 cells. Our results suggest that pioglitazone has the ability to enhance morphine-dependence and to augment morphine withdrawal signs. The possible pathway underlying this effect is through activation of NO/GC/cGMP pathway.


Assuntos
GMP Cíclico/metabolismo , Hipoglicemiantes/efeitos adversos , Dependência de Morfina/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tiazolidinedionas/efeitos adversos , Animais , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Glioblastoma/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Dependência de Morfina/complicações , Dependência de Morfina/patologia , Naloxona/toxicidade , Antagonistas de Entorpecentes/toxicidade , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Pioglitazona , RNA Mensageiro , Síndrome de Abstinência a Substâncias/etiologia , Síndrome de Abstinência a Substâncias/metabolismo , Transfecção
7.
J Neurosci ; 25(6): 1366-74, 2005 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-15703390

RESUMO

Compulsive drug-seeking behavior and its renewal in former drug addicts is promoted by several situations, among which reactivation of drug withdrawal memories plays a crucial role. A neural hypothesis is that such memories reactivate the circuits involved in withdrawal itself and promote a motivational state leading to drug seeking or taking. To test this hypothesis, we have analyzed the neural circuits and cell populations recruited when opiate-dependent rats are reexposed to stimuli previously paired with withdrawal (memory retrieval) and compared them with those underlying acute withdrawal during conditioning (memory formation). Using in situ hybridization for c-fos expression, we report here that reexposure to a withdrawal-paired environment induced conditioned c-fos responses in a specific limbic circuit, which can be partially dissociated from the structures involved in acute withdrawal. At the amygdala level, c-fos responses were doubly dissociated between the central and basolateral (BLA) nuclei, when comparing the two situations. Detailed phenotypical analyses in the amygdala and ventral tegmental area (VTA) show that specific subpopulations in the BLA are differentially involved in the formation and retrieval of withdrawal memories, and strikingly that a population of VTA dopamine neurons is activated in both situations. Together, this indicates that withdrawal memories can drive activity changes in specific neuronal populations of interconnected limbic areas known to be involved in aversive motivational processes. This first study on the neural substrates of withdrawal memories strongly supports an incentive-motivational view of withdrawal in opiate addiction that could be crucial in compulsive drug seeking and relapse.


Assuntos
Comportamento Apetitivo/fisiologia , Aprendizagem da Esquiva/fisiologia , Sistema Límbico/fisiopatologia , Memória/fisiologia , Morfina/toxicidade , Rede Nervosa/fisiologia , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Tonsila do Cerebelo/fisiopatologia , Animais , Condicionamento Clássico , Preparações de Ação Retardada , Dopamina/fisiologia , Implantes de Medicamento , Genes fos , Hibridização In Situ , Masculino , Modelos Neurológicos , Morfina/administração & dosagem , Motivação , Naloxona/administração & dosagem , Naloxona/toxicidade , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neurônios/fisiologia , Transtornos Relacionados ao Uso de Opioides/psicologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , Ratos , Ratos Sprague-Dawley , Recidiva , Síndrome de Abstinência a Substâncias/psicologia , Tegmento Mesencefálico/fisiopatologia , Tirosina 3-Mono-Oxigenase/genética , Ácido gama-Aminobutírico/fisiologia
8.
J Neurosci ; 17(21): 8520-7, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9334424

RESUMO

Chronic opiate administration upregulates the cAMP pathway in the locus coeruleus (LC). This adaptation is thought to increase the electrical excitability of LC neurons and contribute to the dramatic increase in LC firing induced by opioid receptor antagonists in opiate-dependent animals. The goal of the present study was to evaluate directly a role of the cAMP pathway in opiate withdrawal behaviors by studying, in vivo, whether withdrawal is influenced by intra-LC infusion of compounds known to activate or inhibit protein kinase A (PKA). Infusions into amygdala or periaqueductal gray (PAG) were studied for comparison. In one series of experiments the effect of intra-LC, intra-amygdala, or intra-PAG infusions of the PKA inhibitor Rp-cAMPS on naloxone-precipitated withdrawal from morphine was examined. Intra-LC infusions of Rp-cAMPS significantly attenuated several prominent behavioral signs of morphine withdrawal. Intra-PAG infusions of Rp-cAMPS also significantly attenuated opiate withdrawal behaviors, although different behaviors were affected. In contrast, intra-amygdala infusions of Rp-cAMPS were without significant effect. In a second series of experiments the effect of intra-LC or intra-PAG infusions of the PKA activator Sp-cAMPS on behavior in nondependent drug-naive animals was determined. Sp-cAMPS infusions into either brain region induced a quasi-withdrawal syndrome, but the observed behaviors differed between the two groups. Analysis of the phosphorylation state of tyrosine hydroxylase, a well characterized substrate for PKA, confirmed the ability of Rp-cAMPS and Sp-cAMPS to inhibit and activate, respectively, PKA activity in vivo. Together, these data provide direct evidence for involvement of the cAMP-PKA system in the LC, as well as in the PAG, in opiate withdrawal and withdrawal-related behaviors.


Assuntos
Comportamento Animal/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , AMP Cíclico/análogos & derivados , Inibidores Enzimáticos/farmacologia , Locus Cerúleo/efeitos dos fármacos , Dependência de Morfina/complicações , Naloxona/toxicidade , Antagonistas de Entorpecentes/toxicidade , Proteínas do Tecido Nervoso/antagonistas & inibidores , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/fisiopatologia , Tionucleotídeos/farmacologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Animais , AMP Cíclico/farmacologia , AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Inibidores Enzimáticos/administração & dosagem , Infusões Parenterais , Locus Cerúleo/fisiologia , Masculino , Atividade Motora/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Substância Cinzenta Periaquedutal/fisiologia , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Comportamento Estereotipado/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/etiologia , Tirosina 3-Mono-Oxigenase/metabolismo
9.
Reprod Toxicol ; 9(2): 169-74, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7795327

RESUMO

Three groups of ovariectomized Holstein heifers were used in this study. Group I heifers (n = 6) were pretreated with saline (SAL), followed 3 h later by three injections of naloxone hydrochloride (NLX) given 1 h apart. In the same sequence, group II heifers (n = 3) received lipopolysaccharide (LPS), followed by the opioid antagonist NLX. Group III heifers (n = 3) received LPS followed by SAL. Concentrations of cortisol and progesterone increased (P < 0.05) following LPS injections in both groups II and III, whereas luteinizing hormone (LH) concentrations were suppressed (P < 0.05). Administration of NLX to heifers pretreated with LPS elicited significant increases (P < 0.05) in LH concentrations, whereas SAL infusion had no effect. These results indicate that the inhibitory actions of opiate mu-delta receptors, and possibly other POMC gene products, were at least partially involved in LPS-induced suppression of the gonadotropic hormone. These results are discussed, including the possibility that feedback suppression by LPS-triggered adrenal gland steroids may have interfered with complete restoration of LH secretion by the opioid antagonist.


Assuntos
Endotoxinas/toxicidade , Lipopolissacarídeos/toxicidade , Hormônio Luteinizante/metabolismo , Naloxona/toxicidade , Peptídeos Opioides/fisiologia , Animais , Bovinos , Endotoxinas/administração & dosagem , Escherichia coli , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hidrocortisona/sangue , Lipopolissacarídeos/administração & dosagem , Hormônio Luteinizante/antagonistas & inibidores , Hormônio Luteinizante/sangue , Naloxona/administração & dosagem , Peptídeos Opioides/antagonistas & inibidores , Peptídeos Opioides/genética , Ovariectomia , Progesterona/sangue , Distribuição Aleatória , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Solução Salina Hipertônica/administração & dosagem
10.
Life Sci ; 48(21): 2035-42, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1851916

RESUMO

We studied the effect of parenteral morphine and naloxone administration on intestinal mucosal Prostaglandin E2 (PGE2) and 3',5' cyclic adenosine monophosphate (cAMP) levels and on indomethacin-induced intestinal ulceration in the rat. Compared to the control group, morphine significantly decreased whereas naloxone markedly increased both PGE2 and cAMP mucosal levels, respectively. Morphine or naloxone alone did not cause mucosal injury. However, when given with indomethacin, morphine significantly potentiated the ulcerogenic effect of indomethacin while naloxone exerted a protective effect. These results suggest that opioid peptides may play a role in modulation of intestinal mucosal PGE2 and cAMP levels. In addition, enhancement of indomethacin-induced ulcer formation by morphine and amelioration by naloxone might be in part mediated through their effect on mucosal PGE2 and cAMP levels.


Assuntos
AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Indometacina , Intestino Delgado/efeitos dos fármacos , Entorpecentes/toxicidade , Úlcera/induzido quimicamente , Animais , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Masculino , Morfina/toxicidade , Naloxona/toxicidade , Ratos , Ratos Endogâmicos , Úlcera/tratamento farmacológico , Úlcera/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA