Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 527
Filtrar
1.
Pharm Res ; 40(8): 1885-1899, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37344602

RESUMO

BACKGROUND & PURPOSE: Heroin (diacetylmorphine; diamorphine) is a highly addictive opioid prodrug. Heroin prescription is possible in some countries for chronic, treatment-refractory opioid-dependent patients and as a potent analgesic for specific indications. We aimed to study the pharmacokinetic interactions of heroin and its main pharmacodynamically active metabolites, 6-monoacetylmorphine (6-MAM) and morphine, with the multidrug efflux transporters P-glycoprotein/ABCB1 and BCRP/ABCG2 using wild-type, Abcb1a/1b and Abcb1a/1b;Abcg2 knockout mice. METHODS & RESULTS: Upon subcutaneous (s.c.) heroin administration, its blood levels decreased quickly, making it challenging to detect heroin even shortly after dosing. 6-MAM was the predominant active metabolite present in blood and most tissues. At 10 and 30 min after heroin administration, 6-MAM and morphine brain accumulation were increased about 2-fold when mouse (m)Abcb1a/1b and mAbcg2 were ablated. Fifteen minutes after direct s.c. administration of an equimolar dose of 6-MAM, we observed good intrinsic brain penetration of 6-MAM in wild-type mice. Still, mAbcb1 limited brain accumulation of 6-MAM and morphine without affecting their blood exposure, and possibly mediated their direct intestinal excretion. A minor contribution of mAbcg2 to these effects could not be excluded. CONCLUSIONS: We show that mAbcb1a/1b can limit 6-MAM and morphine brain exposure. Pharmacodynamic behavioral/postural observations, while non-quantitative, supported moderately increased brain levels of 6-MAM and morphine in the knockout mouse strains. Variation in ABCB1 activity due to genetic polymorphisms or environmental factors (e.g., drug interactions) might affect 6-MAM/morphine exposure in individuals, but only to a limited extent.


Assuntos
Heroína , Morfina , Camundongos , Animais , Heroína/metabolismo , Heroína/farmacologia , Morfina/metabolismo , Analgésicos Opioides/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Neoplasias/metabolismo , Encéfalo/metabolismo , Derivados da Morfina/metabolismo , Derivados da Morfina/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Camundongos Knockout
2.
Transl Psychiatry ; 13(1): 120, 2023 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-37031205

RESUMO

Heroin is an opioid agonist commonly abused for its rewarding effects. Since its synthesis at the end of the nineteenth century, its popularity as a recreational drug has ebbed and flowed. In the last three decades, heroin use has increased again, and yet the pharmacology of heroin is still poorly understood. After entering the body, heroin is rapidly deacetylated to 6-monoacetylmorphine (6-MAM), which is then deacetylated to morphine. Thus, drug addiction literature has long settled on the notion that heroin is little more than a pro-drug. In contrast to these former views, we will argue for a more complex interplay among heroin and its active metabolites: 6-MAM, morphine, and morphine-6-glucuronide (M6G). In particular, we propose that the complex temporal pattern of heroin effects results from the sequential, only partially overlapping, actions not only of 6-MAM, morphine, and M6G, but also of heroin per se, which, therefore, should not be seen as a mere brain-delivery system for its active metabolites. We will first review the literature concerning the pharmacokinetics and pharmacodynamics of heroin and its metabolites, then examine their neural and behavioral effects, and finally discuss the possible implications of these data for a better understanding of opioid reward and heroin addiction. By so doing we hope to highlight research topics to be investigated by future clinical and pre-clinical studies.


Assuntos
Dependência de Heroína , Heroína , Humanos , Heroína/efeitos adversos , Analgésicos Opioides/efeitos adversos , Derivados da Morfina/metabolismo , Derivados da Morfina/farmacologia , Morfina/farmacologia
3.
Folia Neuropathol ; 61(1): 97-104, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37114965

RESUMO

INTRODUCTION: The aim of the study was to explore the analgesic mechanism of effects of intrathecally administered interferon a (IFN-a) on chronic constriction injury (CCI) model rats. MATERIAL AND METHODS: 24 rats were divided into 6 groups, with 4 rats in each group, including the negative control group (Group N, no operation or treatment), the sham operation group (Group S, only the left sciatic nerve of the rats was exposed without ligation, 0.9% NaCl was intrathecally administered), and four experimental groups (CCI model was established first and then different drugs were intrathecally administered respectively), including 0.9% NaCl (Group C), IFN-a (Group CI), morphine (Group CM), and IFN-a combined with morphine (Group CIM). The mRNA levels of G proteins in both the spinal cord and dorsal root ganglia (DRG), as well as the content of amino acid and chemokine (C-X-C motif) ligand 6 (CXCL-6) in the cerebrospinal fluid were measured and analysed in each group. RESULTS: Intrathecal administration of IFN-a increased the mechanical pain threshold in CCI rats (33.32 ±1.36 vs. 21.08 ±1.59, p < 0.001), achieving the effect comparable to that of morphine (33.32 ±1.36 vs. 32.44 ±3.18, p > 0.05), increased the mRNA expression level of Gi protein (0.62 ±0.04 vs. 0.49 ±0.05, p = 0.006), and decreased the mRNA expression level of Gs protein in the spinal cord (1.80 ±0.16 vs. 2.06 ±0.15, p = 0.035) and DRG (2.11 ±0.10 vs. 2.79 ±0.13, p < 0.001). The intrathecal administration of both IFN-a and morphine can reduce the glutamate content in the cerebrospinal fluid (261.55 ±38.12 vs. 347.70 ±40.69, p = 0.012), but without any statistically significant difference in the content of CXCL-6 across all groups ( p > 0.05). CONCLUSIONS: Intrathecal injection of IFN-a improved the mechanical pain threshold in CCI rats, so we inferred that intrathecal administration of IFN-a had analgesic effects on neuropathic pain, possibly related to the activation of G-proteincoupled µ receptors in the spinal cord and the inhibition of glutamate release.


Assuntos
Interferon-alfa , Limiar da Dor , Ratos , Animais , Ratos Sprague-Dawley , Interferon-alfa/farmacologia , Interferon-alfa/metabolismo , Constrição , Solução Salina/metabolismo , Solução Salina/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/farmacologia , RNA Mensageiro/metabolismo , Medula Espinal/metabolismo , Derivados da Morfina/metabolismo , Derivados da Morfina/farmacologia
4.
Int J Legal Med ; 137(4): 1039-1049, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36786894

RESUMO

The most common pulmonary findings in opiate-related fatalities are congestion and oedema, as well as acute and/or chronic alveolar haemorrhage, the cause of which is thought to be a damage to the capillary endothelium related to ischemia. Human vascular mesenchymal stromal cells (vMSCs) play a fundamental role in tissue regeneration and repair after endothelial cell injury, and they express opioid receptors. The aim of this study was to assess the effect of in vitro morphine exposure on the physiological activity and maintenance of human vMSCs. vMSCs were obtained from abdominal aorta fragments collected during surgery repair and were exposed to incremental doses (0.1 mM, 0.4 mM, 0.8 mM and 1 mM) of morphine sulphate for 7 days. The effect was investigated through cell viability assessment, proliferation assay, reactive oxygen species (ROS) detection assay, senescence-associated ß-galactosidase assay, senescent-related markers (p21WAF1/CIP1 and p16INK4) and the apoptosis-related marker caspase 3. Moreover, an ultrastructural analysis by transmission electron microscopy and in vitro vascular differentiation were evaluated. Results showed a decrease of the cellular metabolic activity, a pro-oxidant and pro-senescence effect, an increase in intracellular ROS and the activation of the apoptosis signalling, as well as ultrastructural modifications and impairment of vascular differentiation after morphine treatment of vMSC. Although confirmation studies are required on real fatal opiate intoxications, the approach based on morphological and immunofluorescence methodologies may have a high potential also as a useful tool or as a complementary method in forensic pathology. The application of these techniques in the future may lead to the identification of new markers and morphological parameters useful as complementary investigations for drug-related deaths.


Assuntos
Células-Tronco Mesenquimais , Alcaloides Opiáceos , Humanos , Senescência Celular/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Alcaloides Opiáceos/farmacologia , Células-Tronco Mesenquimais/metabolismo , Derivados da Morfina/farmacologia
5.
J Exp Anal Behav ; 119(2): 392-406, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36759567

RESUMO

Opioid overdose and opioid use disorder continue to be significant public health challenges despite the availability of effective medications and significant efforts at all levels of society. The emergence of highly potent and efficacious opioids such as fentanyl and its derivatives over the last decade has only exacerbated what was already a substantial problem. Behavioral pharmacology research has proven invaluable for understanding the effects of drugs as well as developing and evaluating pharmacotherapies for disorders involving the central nervous system, including substance abuse disorders. This paper describes a program of research characterizing a potent, selective, and long-lasting mu opioid receptor antagonist, methocinnamox, and evaluating its potential for treating opioid overdose and opioid use disorder. Studies in rodents and nonhuman primates demonstrate that methocinnamox prevents and reverses opioid-induced ventilatory depression and selectively blocks opioid self-administration. This work, taken together with rigorous in vitro and ex vivo studies investigating methocinnamox neuropharmacology, lays a solid foundation for the therapeutic utility of this potentially life-saving medication. Moreover, these studies demonstrate how rigorous behavioral pharmacological studies can be integrated in a broader drug discovery and development research program.


Assuntos
Overdose de Opiáceos , Transtornos Relacionados ao Uso de Opioides , Animais , Analgésicos Opioides/farmacologia , Overdose de Opiáceos/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Derivados da Morfina/farmacologia , Derivados da Morfina/uso terapêutico , Receptores Opioides mu/uso terapêutico
6.
J Theor Biol ; 562: 111435, 2023 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-36764443

RESUMO

Injection drug use is one of the most significant risk factors associated with contracting human immunodeficiency virus (HIV), and drug users infected with HIV suffer from a higher viral load and rapid disease progression. While replication of HIV may result in many mutant viruses that can escape recognition of the host's immune response, the presence of morphine (a drug of abuse) can decrease the viral mutation rate and cellular immune responses. This study develops a mathematical model to explore the effects of morphine-altered mutation and cellular immune response on the within-host dynamics of two HIV species, a wild-type and a mutant. Our model predicts that the morphine-altered mutation rate and cellular immune response allow the wild-type virus to outcompete the mutant virus, resulting in a higher set point viral load and lower CD4 count. We also compute the basic reproduction numbers and show that the dominant species is determined by morphine concentration, with the mutant dominating below and the wild-type dominating above a threshold. Furthermore, we identified three biologically relevant equilibria, infection-free, mutant-only, and coexistence, which are completely characterized by the fitness cost of mutation, mutant escape rate, and morphine concentration.


Assuntos
Infecções por HIV , HIV-1 , Humanos , HIV-1/fisiologia , Derivados da Morfina/farmacologia , Replicação Viral , Mutação
7.
Am J Vet Res ; 84(3)2023 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-36662607

RESUMO

OBJECTIVE: To investigate the cytotoxic effects of 2 different concentrations of buprenorphine and compare them with bupivacaine and morphine on healthy equine chondrocytes in vitro. SAMPLE: Primary cultured equine articular chondrocytes from 3 healthy adult horses. PROCEDURES: Chondrocytes were exposed for 0 and 2 hours to the following treatments: media (CON; negative control); bupivacaine at 2.2 mg/mL (BUPI; positive control); morphine at 2.85 mg/mL (MOR); buprenorphine at 0.12 mg/mL (HBUPRE); or buprenorphine at 0.05 mg/mL (LBUPRE). Chondrocyte viability was assessed using live/dead staining, water-soluble tetrazolium salt-8 (WST-8) cytotoxic assay, LDH assay, and flow cytometry. All continuous variables were evaluated with a mixed ANOVA with treatment, time, and their interactions as the fixed effects and each horse as the random effect. RESULTS: Buprenorphine showed a concentration-dependent chondrotoxic effect. The viability of chondrocytes was significantly decreased with exposure to HBUPRE and BUPI compared to CON, MOR, and LBUPRE. CLINICAL RELEVANCE: Negligible chondrotoxic effects were observed in healthy cultured equine chondrocytes exposed to 0.05 mg/mL of buprenorphine, whereas higher concentrations (0.12 mg/mL) showed a marked cytotoxic effect. Based on these results, low concentrations of buprenorphine appear to be safe for intra-articular administration. Further evaluation of this dose in vivo is needed before recommending its clinical use.


Assuntos
Antineoplásicos , Buprenorfina , Cartilagem Articular , Cavalos , Animais , Condrócitos , Anestésicos Locais/farmacologia , Buprenorfina/farmacologia , Bupivacaína/farmacologia , Antineoplásicos/farmacologia , Derivados da Morfina/farmacologia
8.
Br J Pharmacol ; 180(7): 843-861, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-34986502

RESUMO

BACKGROUND AND PURPOSE: In rodents, morphine antinociception is influenced by sex. However, conflicting results have been reported regarding the interaction between sex and morphine antinociceptive tolerance. Morphine is metabolised in the liver and brain into morphine-3-glucuronide (M3G). Sex differences in morphine metabolism and differential metabolic adaptations during tolerance development might contribute to behavioural discrepancies. This article investigates the differences in peripheral and central morphine metabolism after acute and chronic morphine treatment in male and female mice. EXPERIMENTAL APPROACH: Sex differences in morphine antinociception and tolerance were assessed using the tail-immersion test. After acute and chronic morphine treatment, morphine and M3G metabolic kinetics in the blood were evaluated using LC-MS/MS. They were also quantified in several CNS regions. Finally, the blood-brain barrier (BBB) permeability of M3G was assessed in male and female mice. KEY RESULTS: This study demonstrated that female mice showed weaker morphine antinociception and faster induction of tolerance than males. Additionally, female mice showed higher levels of M3G in the blood and in several pain-related CNS regions than male mice, whereas lower levels of morphine were observed in these regions. M3G brain/blood ratios after injection of M3G indicated no sex differences in M3G BBB permeability, and these ratios were lower than those obtained after injection of morphine. CONCLUSION: These differences are attributable mainly to morphine central metabolism, which differed between males and females in pain-related CNS regions, consistent with weaker morphine antinociceptive effects in females. However, the role of morphine metabolism in antinociceptive tolerance seemed limited. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.


Assuntos
Morfina , Espectrometria de Massas em Tandem , Camundongos , Masculino , Feminino , Animais , Cromatografia Líquida , Derivados da Morfina/farmacologia , Derivados da Morfina/uso terapêutico , Analgésicos Opioides/farmacologia , Dor/tratamento farmacológico
9.
Br J Pharmacol ; 180(10): 1362-1378, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36562107

RESUMO

BACKGROUND AND PURPOSE: Opioids are commonly used for the management of cancer-associated pain and chemotherapy-induced diarrhoea. The chemotherapeutic irinotecan (CPT-11) causes severe gastrointestinal (GI) toxicity due to deconjugation of inactive metabolite SN-38 glucuronide (SN-38G) by bacterial ß-glucuronidases to the active 7-ethyl-10-hydroxycamptothecin (SN-38). Opioids are known to cause gut microbial dysbiosis, this study evaluated whether CPT-11 anti-tumour efficacy and GI toxicity are exacerbated by opioid co-administration. EXPERIMENTAL APPROACH: Eight-week-old C57BL/6 male mice were co-administration with CPT-11 ± opioid. 16S rRNA sequencing was used for gut microbiome analysis. LC-MS analyses of plasma and intestinal extracts were performed to investigate the pharmacokinetic profile of CPT-11. Histological analysis and quantitative real-time polymerase chain reaction were used to determine the severity of intestinal tissue damage. Human liver microsome In vitro assay was performed to confirm the effects of opioids on CPT-11 metabolism. KEY RESULTS: Gut microbiome analysis showed that morphine treatment induced enrichment of ß-glucuronidase-producing bacteria in the intestines of CPT-11-treated mice, resulting in SN-38 accumulation and exacerbation of GI toxicity in the small intestine. Oral administration of both antibiotics and glucuronidase inhibitor protected mice against GI toxicity induced with CPT-11 and morphine co-administration, implicating a microbiome-dependent mechanism. Additionally, morphine and loperamide decreased the plasma concentration of SN-38 and compromised CPT-11 anti-tumour efficacy, this seemed to be microbiome independent. CONCLUSION AND IMPLICATIONS: Gut microbiota play a significant role in opioid and chemotherapeutic agent drug-drug interactions. Inhibition of gut microbial glucuronidase may also prevent adverse GI effects of CPT-11 in patients on opioids.


Assuntos
Antineoplásicos Fitogênicos , Neoplasias , Humanos , Camundongos , Masculino , Animais , Irinotecano , Analgésicos Opioides/farmacologia , Disbiose , Modelos Animais de Doenças , RNA Ribossômico 16S , Antineoplásicos Fitogênicos/toxicidade , Camundongos Endogâmicos C57BL , Camptotecina/toxicidade , Bactérias , Glucuronidase/metabolismo , Glucuronidase/farmacologia , Derivados da Morfina/farmacologia
10.
Int Wound J ; 20(6): 1954-1959, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36517963

RESUMO

In recent years, the use of topical morphine gel has increased in the palliative care setting to reduce pain in chronic wounds and fungating tumours. However, there is a paucity of evidence to support its effectiveness. Epidermolysis bullosa (EB) is a rare genetic skin fragility disorder characterised by painful chronic wounds. Adequate control of wound pain can be challenging in this patient group due to other complexities associated with the more severe sub-types of the disease. Topical morphine gel has been used as an adjunct therapy in a small number of EB patients in our tertiary centre in an attempt to improve pain control and quality of life. The purpose of this paper is to demonstrate the efficacy of topical morphine gel used in a variety of EB wounds as well as patient reported reduction in pain through a series of case studies. The case studies suggest a positive effect of topical morphine gel on painful wounds across a spectrum of EB subtypes.


Assuntos
Epidermólise Bolhosa , Qualidade de Vida , Humanos , Cicatrização , Epidermólise Bolhosa/complicações , Epidermólise Bolhosa/tratamento farmacológico , Dor/etiologia , Dor/complicações , Géis/uso terapêutico , Géis/farmacologia , Derivados da Morfina/farmacologia
11.
Arch Toxicol ; 97(2): 359-375, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36344690

RESUMO

Insights into the pathophysiology of many non-immune-mediated drug reactions referred to as toxicities, sensitivities, intolerances, or pseudoallergies have resulted from research identifying the mastocyte-related G-protein-coupled receptor (GPCR) member X2 (MRGPRX2), a human mast cell receptor mediating adverse reactions without the involvement of antibody priming. Opioid-induced degranulation of mast cells, particularly morphine, provoking release of histamine and other preformed mediators and causing hemodynamic and cutaneous changes seen as flushing, headache and wheal and flare reactions in the skin, is an example of results of MRGPRX2 activation. Opioids including morphine, codeine, dextromethorphan and metazocine as well as endogenous prodynorphin opioid peptides activate MRGPRX2 at concentrations causing mast cell degranulation. Unlike the canonical opioid receptors, MRGPRX2 shows stereochemical recognition preference for dextro rather than levo opioid enantiomers. Opioid analgesic drugs (OADs) display a range of histamine-releasing potencies from the strong releaser morphine to doubtful releasers like hydromorphone and the non-releaser fentanyl. Whether there is a correlation between histamine release by individual OADs, MRGPRX2 activation, and presence or absence of adverse cutaneous effects is not known. To investigate the question, ongoing research with recently pursued methodologies and strategies employing basophil and mast cell tests resulting from MRGPRX2 insights should help to elucidate whether or not an opioid's histamine-releasing potency, and its property of provoking an adverse reaction, are each a reflection of its activation of MRGPRX2.


Assuntos
Analgésicos Opioides , Hipersensibilidade , Humanos , Analgésicos Opioides/toxicidade , Histamina/farmacologia , Receptores de Neuropeptídeos , Receptores Acoplados a Proteínas G , Derivados da Morfina/farmacologia , Mastócitos , Degranulação Celular , Proteínas do Tecido Nervoso
12.
Front Neural Circuits ; 16: 977642, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36110920

RESUMO

Dysregulation of body weight maintenance and opioid dependence are often treated as independent disorders. Here, we assessed the effects of both acute and long-term administration of morphine with and without chemogenetic activation of agouti-related peptide (AGRP)-expressing neurons in the arcuate nucleus (ARCAGRP neurons) to elucidate whether morphine and neuronal activation affect feeding behavior and body weight. First, we characterized interactions of opioids and energy deficit in wild-type mice. We observed that opioid administration attenuated both fasting-induced refeeding and ghrelin-stimulated feeding. Moreover, antagonism of opioid receptors blocked fasting-induced refeeding behavior. Next, we interfaced chemogenetics with opioid dependence. For chemogenetic experiments of ARCAGRP neurons, we conducted a priori behavioral qualification and post-mortem FOS immunostaining verification of arcuate activation following ARCAGRP chemogenetic activation. We administered clozapine during short-term and long-term morphine administration paradigms to determine the effects of dependence on food intake and body weight. We found that morphine occluded feeding behavior characteristic of chemogenetic activation of ARCAGRP neurons. Notably, activation of ARCAGRP neurons attenuated opioid-induced weight loss but did not evoke weight gain during opioid dependence. Consistent with these findings, we observed that morphine administration did not block fasting-induced activation of the ARC. Together, these results highlight the strength of opioidergic effects on body weight maintenance and demonstrate the utility of ARCAGRP neuron manipulations as a lever to influence energy balance throughout the development of opioid dependence.


Assuntos
Clozapina , Transtornos Relacionados ao Uso de Opioides , Proteína Relacionada com Agouti/farmacologia , Analgésicos Opioides/farmacologia , Animais , Peso Corporal , Clozapina/farmacologia , Ingestão de Alimentos , Grelina/farmacologia , Camundongos , Derivados da Morfina/farmacologia , Neurônios/fisiologia , Receptores Opioides
13.
Cardiovasc J Afr ; 33(3): 153-156, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35333279

RESUMO

BACKGROUND: Open-heart surgery is associated with severe postoperative pain. Adequate analgesia after open-heart surgery improves patients' early postoperative recovery, extubation, ambulation and early discharge from hospital. Regional anaesthesia techniques are the new hope for adequate postoperative analgesia after cardiac surgery and are widely used for early pain management in the first six hours. METHODS: A total of 100 patients with the American Society of Anesthesiologists physical status classification I-III, aged 18 years and over, undergoing open-heart surgery with sternotomy for coronary artery bypass grafting or valve replacement under general anaesthesia, were included in this study. For postoperative analgesia, 50 patients with pectoral nerve (PECS II) block and 50 with parasternal (PS) block were consecutively enrolled in one of the groups at the end of the surgery and compared in terms of sedation scores, ventilation duration, pain scores at rest after extubation, block duration, total morphine consumption and complications. RESULTS: The block duration in the PS group was statistically significantly higher than in the PECS II group (p = 0.001, p < 0.05, respectively). The visual analogue scale scores at rest in the fourth and sixth hours were statistically significantly higher in the PECS II group than in the PS group ( p = 0.001, p = 0.001, p < 0.01). Cumulative morphine consumption in the PECS II group was statistically significantly higher than in the PS group in the fourth, sixth, 12th and 24th hours ( p = 0.001, p = 0.001, p = 0.001, p = 0.001, p = 0.001, p < 0.01, respectively). CONCLUSIONS: PS block provided longer block duration with lower postoperative pain and sedation scores than the PECS II block, with lower cumulative morphine consumption.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Bloqueio Nervoso , Nervos Torácicos , Adolescente , Adulto , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Humanos , Derivados da Morfina/farmacologia , Bloqueio Nervoso/efeitos adversos , Bloqueio Nervoso/métodos , Dor Pós-Operatória/diagnóstico , Dor Pós-Operatória/etiologia , Dor Pós-Operatória/prevenção & controle
14.
JBRA Assist Reprod ; 26(3): 444-449, 2022 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34995046

RESUMO

OBJECTIVE: Morphine is one of the major psychoactive chemicals in opium that can increase the production of free radicals and thus can negatively affect spermatogenesis. The purpose of this study was to demonstrate the effects of morphine consumption on sperm parameters, DNA integrity and apoptosis in men taking morphine. METHODS: In this case-control study, 30 man abusing morphine (cases) and 30 healthy men (controls) were compared in sperm parameters (count, motility and morphology) and sperm chromatin quality, with Aniline Blue (AB), Toluidine blue (TB) and Chromomycin A3 (CMA3) stains. The participants were matched for age, weight, amount and duration of cigarette smoking. RESULTS: In men with morphine dependency, sperm progressive and total motility (p=0.038 and p=0.000, respectively) showed a significant decrease compared to the control group. Concerning morphine abuse, although morphine can decrease the sperm chromatin condensation and increases the rate of sperm apoptosis, these differences were not statistically significant. CONCLUSIONS: According to our results morphine dependence can reduce male fertility by affecting sperm parameters.


Assuntos
Cromatina , Dependência de Morfina , Apoptose , Estudos de Casos e Controles , Humanos , Masculino , Derivados da Morfina/farmacologia , Sêmen , Motilidade dos Espermatozoides , Espermatozoides
15.
Life Sci ; 293: 120279, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35032552

RESUMO

BACKGROUND: Curcumin is one of the compounds present in plants of the genus Curcuma sp., being very used not only as condiment but also with medicinal purposes. As an analgesic, papers highlight the efficacy of curcumin in the treatment of various types of pain. AIMS: In this study we evaluated the peripheral antinociceptive effect of curcumin and by which mechanisms this effect is induced. MAIN METHODS: The mice paw pressure test was used on animals which had increased pain sensitivity by intraplantar injection of carrageenan. All the drugs were administered in the right hind paw. KEY FINDINGS: Curcumin was administered to the right hind paw animals induced antinociceptive effect. Non -selective antagonist of opioid receptors naloxone reverted the antinociceptive effect induced by curcumin. Selective antagonists for µ, δ and κ opioid receptors clocinnamox, naltrindole and nor- binaltorphimine, respectively, reverted the antinociceptive effect induced by curcumin. Bestatin, enkephalinases inhibitor that degrade peptides opioids, did not change the nociceptive response. Selective antagonists for CB1 and CB2 cannabinoid receptors, AM251 and AM630, respectively, reversed the antinociceptive effect induced by curcumin. The MAFP inhibitor of the enzyme FAAH which breaks down anandamide, JZL184, enzyme inhibitor MAGL which breaks down the 2-AG, as well as the VDM11 anandamide reuptake inhibitor potentiated the antinociceptive effect of curcumin. SIGNIFICANCE: These results suggest that curcumin possibly peripheral antinociception induced by opioid and cannabinoid systems activation and possibly for endocannabinoids and opioids release.


Assuntos
Analgésicos/uso terapêutico , Agonistas de Receptores de Canabinoides/uso terapêutico , Curcumina/uso terapêutico , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Receptores Opioides/metabolismo , Analgésicos/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Ácidos Araquidônicos/farmacologia , Ácidos Araquidônicos/uso terapêutico , Agonistas de Receptores de Canabinoides/farmacologia , Carragenina/toxicidade , Cinamatos/farmacologia , Curcumina/farmacologia , Relação Dose-Resposta a Droga , Endocanabinoides/farmacologia , Endocanabinoides/uso terapêutico , Hiperalgesia/induzido quimicamente , Masculino , Camundongos , Derivados da Morfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor/induzido quimicamente , Dor/tratamento farmacológico , Dor/metabolismo , Alcamidas Poli-Insaturadas/farmacologia , Alcamidas Poli-Insaturadas/uso terapêutico
16.
J Neurosci Res ; 100(1): 220-236, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-32954564

RESUMO

Tolerance and hyperalgesia associated with chronic exposure to morphine are major limitations in the clinical management of chronic pain. At a cellular level, neuronal signaling can in part account for these undesired side effects, but unknown mechanisms mediated by central nervous system glial cells are likely also involved. Here we applied data-independent acquisition mass spectrometry to perform a deep proteome and phosphoproteome analysis of how human astrocytes responds to opioid stimulation. We unveil time- and dose-dependent effects induced by morphine and its major active metabolites morphine-3-glucuronide (M3G) and morphine-6-glucuronide that converging on activation of mitogen-activated protein kinase and mammalian target of rapamycin signaling pathways. We also find that especially longer exposure to M3G leads to significant dysregulation of biological pathways linked to extracellular matrix organization, antigen presentation, cell adhesion, and glutamate homeostasis, which are crucial for neuron- and leukocyte-astrocyte interactions.


Assuntos
Astrócitos , Morfina , Astrócitos/metabolismo , Humanos , Morfina/farmacologia , Derivados da Morfina/metabolismo , Derivados da Morfina/farmacologia , Proteômica
17.
J Psychoactive Drugs ; 54(3): 258-268, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34355666

RESUMO

Prescription opioid misuse is an unintended consequence of acute pain management. Opioid-induced euphoria (OIE) with first therapeutic opioid exposure may influence opioid misuse. OIE is not assessed in clinical care and self-report measures of OIE have not been validated in adolescents. We (1) determined adolescents' ability to understand existing self-reported OIE measures, (2) revised measures for better understanding by this population, and (3) established initial content validity of revised measures with adolescents. Using runner's euphoria to simulate OIE in Study 1, 29 adolescents' (14 males) understanding of the Drug Effects Questionnaire (DEQ-5), the Addiction Resource Center Inventory Morphine Benzedrine Group scale (ARCI-MBG), and the ARCI Lysergic Acid Diethylamide scale (ARCI-LSD) were tested. In Study 2, 29 additional adolescents (9 males) participated in a modified Delphi study with focus groups to revise survey items to improve understanding by peers. In Study 1, runners understood <40% of ARCI-MBG and ARCI-LSD statements. In Study 2, all but 7 survey items were revised. Revised measures of OIE for adolescents may help define at-risk OIE phenotypes and validate risk assessments using survey methodology. Additional studies are needed to validate the revised OIE self-report measures with opioid-naive adolescents receiving opioids to treat acute pain.


Assuntos
Transtornos Relacionados ao Uso de Opioides , Uso Indevido de Medicamentos sob Prescrição , Analgésicos Opioides/uso terapêutico , Euforia , Humanos , Dietilamida do Ácido Lisérgico/farmacologia , Masculino , Derivados da Morfina/farmacologia , Transtornos Relacionados ao Uso de Opioides/epidemiologia
18.
Pharmacol Res Perspect ; 9(6): e00887, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34713624

RESUMO

Opioid overdose is a leading cause of death in the United States. The only treatment available currently is the competitive antagonist, naloxone (Narcan® ). Although naloxone is very effective and has saved many lives, as a competitive antagonist it has limitations. Due to the short half-life of naloxone, renarcotization can occur if the ingested opioid agonist remains in the body longer. Moreover, because antagonism by naloxone is surmountable, renarcotization can also occur in the presence of naloxone if a relatively larger dose of opioid agonist is taken. In such circumstances, a long-lasting, non-surmountable antagonist would offer an improvement in overdose treatment. Methocinnamox (MCAM) has been reported to have a long duration of antagonist action at mu opioid receptors in vivo. In HEK cells expressing the human mu opioid receptor, MCAM antagonism of mu agonist-inhibition of cAMP production was time-dependent, non-surmountable and non-reversible, consistent with (pseudo)-irreversible binding. In vivo, MCAM injected locally into the rat hindpaw antagonized mu agonist-mediated inhibition of thermal allodynia for up to 96 h. By contrast, antagonism by MCAM of delta or kappa agonists in HEK cells and in vivo was consistent with simple competitive antagonism. Surprisingly, MCAM also shifted the concentration-response curves of mu agonists in HEK cells in the absence of receptor reserve in a ligand-dependent manner. The shift in the [D-Ala2 ,N-MePhe4 ,Gly-ol5 ]-enkephalin (DAMGO) concentration-response curve by MCAM was insensitive to naloxone, suggesting that in addition to (pseudo)-irreversible orthosteric antagonism, MCAM acts allosterically to alter the affinity and/or intrinsic efficacy of mu agonists.


Assuntos
Cinamatos/farmacologia , Derivados da Morfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/antagonistas & inibidores , Regulação Alostérica/efeitos dos fármacos , Animais , AMP Cíclico/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Células HEK293 , Humanos , Ligantes , Masculino , Naloxona/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo , Fatores de Tempo
19.
Bioanalysis ; 13(18): 1415-1425, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34528849

RESUMO

Aim: Nicomorphine is rapidly metabolized mainly to the biologically active 6-nicotinoyl morphine and morphine. In sport, morphine and nicomorphine use is prohibited whereas codeine use is permitted. Accredited laboratories routinely test for morphine hence must be able to distinguish morphine, as a metabolite of a prohibited substance, from that whose use is permitted. Results: Here we show a relatively simple method to synthesize the nicomorphine metabolites, 3-nicotinoyl and 6-nicotinoyl morphine, and indicate how they may be used to identify nicomorphine administration. Conclusion: This approach should help confirm that it is not codeine, an allowable analgesic in sport, that has been administered.


Assuntos
Doping nos Esportes/prevenção & controle , Derivados da Morfina/uso terapêutico , Derivados da Morfina/farmacologia , Ácidos Nicotínicos/farmacologia , Ácidos Nicotínicos/uso terapêutico
20.
Eur J Pharmacol ; 901: 174089, 2021 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-33826922

RESUMO

The participation of the peripheral opioid and cannabinoid endogenous systems in modulating muscle pain and inflammation has not been fully explored. Thus, the aim of this study was to investigate the involvement of these endogenous systems during muscular-tissue hyperalgesia induced by inflammation. Hyperalgesia was induced by carrageenan injection into the tibialis anterior muscles of male Wistar rats. We padronized an available Randal-Sellito test adaptation to evaluate nociceptive behavior elicited by mechanical insult in muscles. Western blot analysis was performed to evaluate the expression levels of opioid and cannabinoid receptors in the dorsal root ganglia. The non-selective opioid peptide receptor antagonist (naloxone) and the selective mu opioid receptor MOP (clocinnamox) and kappa opioid receptor KOP (nor-binaltorphimine) antagonists were able to intensify carrageenan-induced muscular hyperalgesia. On the other hand, the selective delta opioid receptor (DOP) antagonist (naltrindole) did not present any effect on nociceptive behavior. Moreover, the selective inhibitor of aminopeptidases (Bestatin) provoked considerable dose-dependent analgesia when intramuscularly injected into the hyperalgesic muscle. The CB1 receptor antagonist (AM251), but not the CB2 receptor antagonist (AM630), intensified muscle hyperalgesia. All irreversible inhibitors of anandamide hydrolase (MAFP), the inhibitor for monoacylglycerol lipase (JZL184) and the anandamide reuptake inhibitor (VDM11) decreased carrageenan-induced hyperalgesia in muscular tissue. Lastly, MOP, KOP and CB1 expression levels in DRG were baseline even after muscular injection with carrageenan. The endogenous opioid and cannabinoid systems participate in peripheral muscle pain control through the activation of MOP, KOP and CB1 receptors.


Assuntos
Mialgia/tratamento farmacológico , Receptores de Canabinoides/fisiologia , Receptores Opioides/fisiologia , Animais , Ácidos Araquidônicos/antagonistas & inibidores , Carragenina , Cinamatos/farmacologia , Endocanabinoides/antagonistas & inibidores , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/psicologia , Masculino , Monoacilglicerol Lipases/antagonistas & inibidores , Derivados da Morfina/farmacologia , Mialgia/induzido quimicamente , Mialgia/psicologia , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Medição da Dor/efeitos dos fármacos , Alcamidas Poli-Insaturadas/antagonistas & inibidores , Ratos , Ratos Wistar , Receptores de Canabinoides/efeitos dos fármacos , Receptores Opioides/efeitos dos fármacos , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...