Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 301
Filtrar
1.
Eur J Pharmacol ; 966: 176333, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38278466

RESUMO

The µ-opioid receptor-biased agonist theory holds that Gio protein signaling mediates the analgesic effect of opioids and the related side effects via the ß-arrestin2 signaling pathway. A series of µ-opioid-biased agonists have been developed in accordance with this theory, and the FDA has approved TRV130 (as a representative of biased agonists) for marketing. However, several reports have raised the issue of opioid side effects associated with the use of agonists. In this study, five permeable peptides were designed to emulate 11 S/T phosphorylation sites at the µ-opioid receptor (MOR) carboxyl-terminal. In vitro experiments were performed to detect the activation level of G proteins from the cAMP inhibition assay and the ß-arrestin2 recruitment by the BRET assay. Designed peptides might effectively interfere with the activation of the Gio and ß-arrestin2 pathways when combined with morphine. The resulting morphine-induced tolerance, respiratory inhibition, and constipation in mice showed that the ß-arrestin2 pathway was responsible for morphine tolerance while the Gio signaling pathway was involved with respiratory depression and constipation and that these side effects were significantly related to phosphorylation sites S363 and T370. This study may provide new directions for the development of safer and more effective opioid analgesics, and the designed peptides may be an effective tool for exploring the mechanism by which µ-opioid receptors function, with the potential of reducing the side effects that are associated with clinical opioid treatment.


Assuntos
Analgésicos Opioides , Morfina , Camundongos , Animais , Morfina/efeitos adversos , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/metabolismo , Receptores Opioides mu/metabolismo , Transdução de Sinais , Constipação Intestinal/induzido quimicamente , Peptídeos/metabolismo , beta-Arrestina 2/metabolismo
2.
Xenobiotica ; 53(8-9): 536-546, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37850428

RESUMO

YR-1702, a hybrid µ/κ/δ receptor agonist, is modified from the traditional opioid analgesic dezocine. It had shown both excellent analgesic effect and lower addiction in phase I clinical trial in China, however, the metabolic pathway of YR-1702 in humans remains unelucidated.The goals of this study are to characterise the metabolism of YR-1702 in human liver microsomes (HLMs) and patients with chronic non-cancer pain by high performance liquid chromatography-coupled with quadrupole-time-of-flight mass spectrometry (HPLC-Q-TOF-MS/MS).The results showed that a total of twelve metabolites were identified in HLMs, in which 7, 6 and 5 metabolites were also found in human plasma, urine and feces, respectively. And the major metabolic pathways include mono-hydroxylation, di-hydroxylation, dehydrogenation and glucuronidation. The locations of hydroxylation and dehydrogenation were identified by the signature fragments of the metabolites.The relative contents of the metabolites in human plasma were also evaluated, in which the main metabolite M1 notably accounting for more than 14% of the total drug exposure. This study would contribute to the understanding of the in vivo metabolite profile of YR-1702 injection for future use.


Assuntos
Dor Crônica , Espectrometria de Massas em Tandem , Ratos , Animais , Humanos , Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas em Tandem/métodos , Ratos Sprague-Dawley , Analgésicos Opioides/análise , Analgésicos Opioides/metabolismo , Dor Crônica/metabolismo , Fezes/química , Microssomos Hepáticos/metabolismo
3.
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
4.
Neuroscience ; 519: 147-161, 2023 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-36997020

RESUMO

The analgesic effect of opioids decreases over time due to the development of analgesic tolerance. We have shown that inhibition of the platelet-derived growth factor beta (PDGFR-ß) signaling eliminates morphine analgesic tolerance in rats. Although the PDGFR-ß and its ligand, the platelet-derived growth factor type B (PDGF-B), are expressed in the substantia gelatinosa of the spinal cord (SG) and in the dorsal root ganglia (DRG), their precise distribution within different cell types of these structures is unknown. Additionally, the impact of a tolerance-mediating chronic morphine treatment, on the expression and distribution of PDGF-B and PDGFR-ß has not yet been studied. Using immunohistochemistry (IHC), we found that in the spinal cord, PDGFR-ß and PDGF-B were expressed in neurons and oligodendrocytes and co-localized with the mu-opioid receptor (MOPr) in opioid naïve rats. PDGF-B was also found in microglia and astrocytes. Both PDGFR-ß and PDGF-B were detected in DRG neurons but not in spinal primary afferent terminals. Chronic morphine exposure did not change the cellular distribution of PDGFR-ß or PDGF-B. However, PDGFR-ß expression was downregulated in the SG and upregulated in the DRG. Consistent with our previous finding that morphine caused tolerance by inducing PDGF-B release, PDGF-B was upregulated in the spinal cord. We also found that chronic morphine exposure caused a spinal proliferation of oligodendrocytes. The changes in PDGFR-ß and PDGF-B expression induced by chronic morphine treatment suggest potential mechanistic substrates underlying opioid tolerance.


Assuntos
Analgésicos Opioides , Morfina , Ratos , Masculino , Animais , Morfina/farmacologia , Analgésicos Opioides/farmacologia , Analgésicos Opioides/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Ratos Sprague-Dawley , Gânglios Espinais/metabolismo , Tolerância a Medicamentos/fisiologia , Medula Espinal/metabolismo
5.
Comb Chem High Throughput Screen ; 26(10): 1900-1906, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36372918

RESUMO

AIM: This study aimed to investigate how opioids affect phagocytosis and microglial nitrite and nitric oxide synthase (iNOS) production during inflammation. BACKGROUND: Opioids are a group of chemicals that are naturally found in the opium poppy plant and exert a variety of effects on the brain, including pain alleviation in some cases. They are commonly used in surgery and perioperative analgesia. However, research on the impact of opioids on microglial inflammatory factor production and phagocytosis is limited. OBJECTIVES: This study was designed to investigate the effects of opioids on inducible nitric oxide synthase (iNOS) activity and nitric oxide (NO) generation. Moreover, the influence of opioids on the engulfment of C8-B4 microglial cells after stimulation with LPS was also examined. METHODS: C8-B4 mouse microglial cells were exposed to various concentrations of opioids after stimulation with lipopolysaccharide (LPS) and interferon-γ (IFN-γ). Nitrite production was assayed. The iNOS and Cox-2 were determined by Western blotting, and fluorescent immunostaining was performed to assess the percentage of microglia that engulfed fluorescent microspheres in total microglia cultivating with opioids after being activated by LPS. RESULTS: After LPS and IFN-γ stimulation, microglia produced lower amounts of nitric oxide (NO) production with buprenorphine, salvinorin A, and naloxone (P<0.05). When combined with naloxone, no significant differences were found than buprenorphine. It was observed that buprenorphine and salvinorin A could suppress iNOS expression activated by LPS and IFN-γ. Phagocytosis was greatly increased after LPS stimulation, and a significant increase was observed after adding salvinorin A. CONCLUSION: Buprenorphine and salvinorin A were found to reduce NO production and iNOS induction in microglial cells activated by LPS and IFN-γ. Salvinorin A promoted the phagocytosis of microglia cells treated by LPS.


Assuntos
Buprenorfina , Microglia , Camundongos , Animais , Microglia/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo II/farmacologia , Nitritos/metabolismo , Nitritos/farmacologia , Analgésicos Opioides/farmacologia , Analgésicos Opioides/metabolismo , Lipopolissacarídeos/farmacologia , Óxido Nítrico , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase/farmacologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Interferon gama/metabolismo , Interferon gama/farmacologia , Naloxona/metabolismo , Naloxona/farmacologia , Fagocitose , Buprenorfina/metabolismo , Buprenorfina/farmacologia
6.
Mol Pharmacol ; 103(1): 1-8, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36310031

RESUMO

Opioid analgesics exert their therapeutic and adverse effects by activating µ opioid receptors (MOPR); however, functional responses to MOPR activation are modulated by distinct signal transduction complexes within the brain. The ventrolateral periaqueductal gray (vlPAG) plays a critical role in modulation of nociception and analgesia, but the exact intracellular pathways associated with opioid responses in this region are not fully understood. We previously showed that knockout of the signal transduction modulator Regulator of G protein Signaling z1 (RGSz1) enhanced analgesic responses to opioids, whereas it decreased the rewarding efficacy of morphine. Here, we applied viral mediated gene transfer methodology and delivered adeno-associated virus (AAV) expressing Cre recombinase to the vlPAG of RGSz1fl\fl mice to demonstrate that downregulation of RGSz1 in this region decreases sensitivity to morphine in the place preference paradigm, under pain-free as well as neuropathic pain states. We also used retrograde viral vectors along with flippase-dependent Cre vectors to conditionally downregulate RGSz1 in vlPAG projections to the ventral tegmental area (VTA) and show that downregulation of RGSz1 prevents the development of place conditioning to low morphine doses. Consistent with the role for RGSz1 as a negative modulator of MOPR activity, RGSz1KO enhances opioid-induced cAMP inhibition in periaqueductal gray (PAG) membranes. Furthermore, using a new generation of bioluminescence resonance energy transfer (BRET) sensors, we demonstrate that RGSz1 modulates Gαz but not other Gαi family subunits and selectively impedes MOPR-mediated Gαz signaling events invoked by morphine and other opioids. Our work highlights a regional and circuit-specific role of the G protein-signaling modulator RGSz1 in morphine reward, providing insights on midbrain intracellular pathways that control addiction-related behaviors. SIGNIFICANCE STATEMENT: This study used advanced genetic mouse models to highlight the role of the signal transduction modulator named RGSz1 in responses to clinically used opioid analgesics. We show that RGSz1 controls the rewarding efficacy of opioids by actions in ventrolateral periaqueductal gray projections to the ventral tegmental area, a key component of the midbrain dopamine pathway. These studies highlight novel mechanisms by which pain-modulating structures control the rewarding efficacy of opioids.


Assuntos
Analgésicos Opioides , Morfina , Camundongos , Animais , Morfina/farmacologia , Morfina/metabolismo , Analgésicos Opioides/farmacologia , Analgésicos Opioides/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Transdução de Sinais , Proteínas de Ligação ao GTP/metabolismo , Recompensa , Receptores Opioides mu/metabolismo
7.
J Leukoc Biol ; 112(5): 1317-1328, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36205434

RESUMO

HIV-associated neurocognitive impairment (HIV-NCI) is a debilitating comorbidity that reduces quality of life in 15-40% of people with HIV (PWH) taking antiretroviral therapy (ART). Opioid use has been shown to increase neurocognitive deficits in PWH. Monocyte-derived macrophages (MDMs) harbor HIV in the CNS even in PWH on ART. We hypothesized that morphine (MOR), a metabolite of heroin, further dysregulates functional processes in MDMs to increase neuropathogenesis. We found that, in uninfected and HIV-infected primary human MDMs, MOR activates these cells by increasing phagocytosis and up-regulating reactive oxygen species. Effects of MOR on phagocytosis were dependent on µ-opioid receptor activity and were mediated, in part, by inhibited lysosomal degradation of phagocytized substrates. All results persisted when cells were treated with both MOR and a commonly prescribed ART cocktail, suggesting minimal impact of ART during opioid exposure. We then performed mass spectrometry in HIV-infected MDMs treated with or without MOR to determine proteomic changes that suggest additional mechanisms by which opioids affect macrophage homeostasis. Using downstream pathway analyses, we found that MOR dysregulates ER quality control and extracellular matrix invasion. Our data indicate that MOR enhances inflammatory functions and impacts additional cellular processes in HIV-infected MDMs to potentially increases neuropathogenesis in PWH using opioids.


Assuntos
Infecções por HIV , Humanos , Infecções por HIV/metabolismo , Morfina/farmacologia , Morfina/metabolismo , Analgésicos Opioides/farmacologia , Analgésicos Opioides/metabolismo , Heroína/metabolismo , Heroína/farmacologia , Qualidade de Vida , Espécies Reativas de Oxigênio/metabolismo , Proteômica , Macrófagos/metabolismo , Receptores Opioides/metabolismo
8.
Int J Mol Sci ; 23(19)2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36232883

RESUMO

Pain is a worldwide public health problem and its treatment is still a challenge since clinically available drugs do not completely reverse chronic painful states or induce undesirable effects. Crotalphine is a 14 amino acids synthetic peptide that induces a potent and long-lasting analgesic effect on acute and chronic pain models, peripherally mediated by the endogenous release of dynorphin A and the desensitization of the transient receptor potential ankyrin 1 (TRPA1) receptor. However, the effects of crotalphine on the central nervous system (CNS) and the signaling pathway have not been investigated. Thus, the central effect of crotalphine was evaluated on the partial sciatic nerve ligation (PSNL)-induced chronic neuropathic pain model. Crotalphine (100 µg/kg, p.o.)-induced analgesia on the 14th day after surgery lasting up to 24 h after administration. This effect was prevented by intrathecal administration of CB1 (AM251) or CB2 (AM630) cannabinoid receptor antagonists. Besides that, crotalphine-induced analgesia was reversed by CTOP, nor-BNI, and naltrindole, antagonists of mu, kappa, and delta-opioid receptors, respectively, and also by the specific antibodies for ß-endorphin, dynorphin-A, and met-enkephalin. Likewise, the analgesic effect of crotalphine was blocked by the intrathecal administration of minocycline, an inhibitor of microglial activation and proliferation. Additionally, crotalphine decreased the PSNL-induced IL-6 release in the spinal cord. Importantly, in vitro, crotalphine inhibited LPS-induced CD86 expression and upregulated CD206 expression in BV-2 cells, demonstrating a polarization of microglial cells towards the M2 phenotype. These results demonstrated that crotalphine, besides activating opioid and cannabinoid analgesic systems, impairs central neuroinflammation, confirming the neuromodulatory mechanism involved in the crotalphine analgesic effect.


Assuntos
Analgesia , Canabinoides , Neuralgia , Aminoácidos/metabolismo , Analgésicos/metabolismo , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Analgésicos Opioides/metabolismo , Anquirinas/metabolismo , Antagonistas de Receptores de Canabinoides/uso terapêutico , Canabinoides/uso terapêutico , Dinorfinas/metabolismo , Encefalina Metionina/metabolismo , Humanos , Interleucina-6/metabolismo , Lipopolissacarídeos/metabolismo , Microglia/metabolismo , Minociclina/uso terapêutico , Neuralgia/metabolismo , Peptídeos , Fenótipo , Receptores Opioides/metabolismo , Medula Espinal , beta-Endorfina/metabolismo
9.
Phytochemistry ; 203: 113422, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36055422

RESUMO

Catharanthus roseus is a medicinal plant that produces an abundance of monoterpenoid indole alkaloids (MIAs), notably including the anticancer compounds vinblastine and vincristine. While the canonical pathway leading to these drugs has been resolved, the regulatory and catalytic mechanisms controlling many lateral branches of MIA biosynthesis remain largely unknown. Here, we describe an ethyl methanesulfonate (EMS) C. roseus mutant (M2-117523) that accumulates high levels of MIAs. The mutant exhibited stunted growth, partially chlorotic leaves, with deficiencies in chlorophyll biosynthesis, and a lesion-mimic phenotype. The lesions were sporadic and spontaneous, appearing after the first true bifoliate and continuing throughout development. The lesions are also the site of high concentrations of akuammicine, a minor constituent of wild type C. roseus leaves. In addition to akuammicine, the lesions were enriched in 25 other MIAs, resulting, in part, from a higher metabolic flux through the pathway. The unique metabolic shift was associated with significant upregulation of biosynthetic and regulatory genes involved in the MIA pathway, including the transcription factors WRKY1, CrMYC2, and ORCA2, and the biosynthetic genes STR, GO, and Redox1. Following the lesion-mimic mutant (LMM) phenotype, the accumulation of akuammicine is jasmonate (JA)-inducible, suggesting a role in plant defence response. Akuammicine is medicinally significant, as a weak opioid agonist, with a preference for the κ-opioid receptor, and a potential anti-diabetic. Further study of akuammicine biosynthesis and regulation can guide plant and heterologous engineering for medicinal uses.


Assuntos
Catharanthus , Alcaloides de Triptamina e Secologanina , Alcaloides , Analgésicos Opioides/metabolismo , Catharanthus/genética , Catharanthus/metabolismo , Clorofila/metabolismo , Metanossulfonato de Etila/metabolismo , Regulação da Expressão Gênica de Plantas , Indóis , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Receptores Opioides/genética , Receptores Opioides/metabolismo , Alcaloides de Triptamina e Secologanina/metabolismo , Alcaloides de Triptamina e Secologanina/farmacologia , Fatores de Transcrição/genética , Vimblastina , Vincristina
10.
Biochem Pharmacol ; 198: 114949, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35143755

RESUMO

Opioids, and numerous centrally active drugs, are metabolized by cytochrome P450 2D (CYP2D). There are sex and estrous cycle differences in brain oxycodone analgesia. Here we investigated the mechanism examining the selective role of CYP2D in the brain on sex, estrous cycle, and hormonal regulation. Propranolol, CYP2D-specific mechanism-based inhibitor, or vehicle was delivered into cerebral ventricles 24 h before administering oxycodone (or oxymorphone, negative control) orally to male and female (in estrus and diestrus) rats. Ovariectomized and sham-operated females received no treatment, estradiol, progesterone or vehicle. Analgesia was measured using tail-flick latency, and brain drug and metabolite concentrations were measured by microdialysis. Data were analyzed by two-way or mixed ANOVA. Following propranolol (versus vehicle) inhibition and oral oxycodone, there were greater increases in brain oxycodone concentrations and analgesia, and greater decreases in brain oxymorphone/oxycodone ratios (an in vivo phenotype of CYP2D in brain) in males and females in estrus, compared to females in diestrus; with no impact on plasma drug concentrations. There was no impact of propranolol pre-treatment, sex, or cycle after oral oxymorphone (non-CYP2D substrate) on brain oxymorphone concentrations or analgesia. There was no impact of propranolol pre-treatment following ovariectomy on brain oxycodone concentrations or analgesia, which was restored in ovariectomized females following estradiol, but not progesterone, treatment. Sex, cycle, and estradiol regulation of CYP2D in brain in turn altered brain oxycodone concentration and response, which may contribute to the large inter-individual variation in response to the numerous centrally acting CYP2D substrate drugs, including opioids.


Assuntos
Analgesia , Oxicodona , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Encéfalo/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Estradiol/metabolismo , Ciclo Estral/metabolismo , Feminino , Masculino , Oxicodona/farmacologia , Oximorfona/metabolismo , Dor/metabolismo , Progesterona/metabolismo , Propranolol/metabolismo , Ratos , Ratos Wistar
11.
Neurosci Lett ; 773: 136479, 2022 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-35085692

RESUMO

Opioid use and abuse remain a significant public health problem, particularly in the United States. Indeed, it is estimated that up to 10% of youths (age 12-18) have taken opioids illicitly. A growing body of evidence suggests that this level of widespread opioid exposure can have effects that extend to subsequent generations. Utilizing a well-established rodent model of preconception adolescent opioid exposure in females, we found decreased opioid self-administration coupled with increased cocaine self-administration in adult offspring. This bidirectional effect may be related to negative affect associated with opioid withdrawal, including enhanced stress reactivity. In this study, we tested the hypothesis that the adult offspring of females exposed to morphine during adolescence will demonstrate increased signs of opioid withdrawal when compared to offspring of saline controls. Females were administered increasing doses of morphine (5-25 mg/kg s.c.) or saline (1 ml/kg) from postnatal day 30 (PND30)-PND39. They were then maintained drug free for a minimum of 4 weeks and mated with drug-naïve males on or after PND70. As adults, their male and female offspring (referred to as Mor-F1 or Sal-F1) were administered morphine (10 mg/kg s.c.) twice a day for 5 days. They were then tested for spontaneous withdrawal behaviors for the next 4 days (∼PND70). Levels of corticotropin releasing hormone (Crh) and urocortin 3 (Ucn3) were examined in the amygdala at 48 h and 96 h of withdrawal. Circulating corticosterone was measured at 48 h. Results indicate that Mor-F1 males are heavier than Sal-F1 males with no baseline differences in females. However, Mor-F1 females did not gain weight at the same rate as Sal-F1 females during withdrawal. While there were no differences in somatic withdrawal signs, gene expression data revealed a sex-specific and time-dependent effect on Crh as well as increased Ucn3 and corticosterone in females at 48hrs withdrawal. Overall, these data point to differences in withdrawal and stress reactivity in Mor-F1 animals that may contribute to observed differences in addiction-like behaviors.


Assuntos
Morfina , Síndrome de Abstinência a Substâncias , Analgésicos Opioides/metabolismo , Animais , Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Morfina/efeitos adversos , Morfina/metabolismo , Entorpecentes , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Ratos Sprague-Dawley , Síndrome de Abstinência a Substâncias/metabolismo
12.
Mol Med Rep ; 25(2)2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34878160

RESUMO

The κ­opioid receptor (KOR) is one of the primary receptors of opioids and serves a vital role in the regulation of pain, anesthesia, addiction and other pathological and physiological processes. KOR is associated with several types of cancer and may influence cancer progression. It has been proposed that KOR may represent a new tumor molecular marker and provide a novel basis for molecular targeted therapies for cancer. However, the association between KOR and cancer remains to be explored comprehensively. The present review introduces KOR and its association with different types of cancer. Improved understanding of KOR may facilitate development of novel antitumor therapies.


Assuntos
Analgésicos Opioides/metabolismo , Hipóxia/metabolismo , Inflamação/metabolismo , Neoplasias/metabolismo , Neovascularização Patológica , Receptores Opioides kappa/fisiologia , Animais , Progressão da Doença , Humanos , Receptores Opioides kappa/química
13.
Int J Mol Sci ; 22(23)2021 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-34884858

RESUMO

The melanocortin system is a major regulator of stress responses in the skin and is responsible for the induction of melanin synthesis through activation of melanogenesis enzymes. The expression of both melanocortin system genes and melanogenesis enzyme genes is altered in psoriasis, and the focus here was on twelve genes related to the signal transduction between them. Additionally, five endogenous opioid system genes that are involved in cutaneous inflammation were examined. Quantitative real-time-PCR was utilized to measure mRNA expression in punch biopsies from lesional and non-lesional skin of psoriasis patients and from the skin of healthy control subjects. Most of the genes related to melanogenesis were down-regulated in patients (CREB1, MITF, LEF1, USF1, MAPK14, ICAM1, PIK3CB, RPS6KB1, KIT, and ATRN). Conversely, an up-regulation occurred in the case of opioids (PENK, PDYN, and PNOC). The suppression of genes related to melanogenesis is in agreement with the reported reduction in pigmentation signaling in psoriatic skin and potentially results from the pro-inflammatory environment. The increase in endogenous opioids can be associated with their involvement in inflammatory dysregulation in psoriasis.


Assuntos
Psoríase/genética , Psoríase/patologia , Pigmentação da Pele/genética , Adolescente , Adulto , Analgésicos Opioides/metabolismo , Biópsia , Estudos de Casos e Controles , Classe I de Fosfatidilinositol 3-Quinases/genética , Encefalinas/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Fator de Transcrição Associado à Microftalmia/genética , Precursores de Proteínas/genética , Receptores Opioides/genética , Pele/patologia , Adulto Jovem , Receptor de Nociceptina
14.
Vet Immunol Immunopathol ; 238: 110292, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34246811

RESUMO

Activated lymphocyte therapy is one of the immunotherapies for cancer patients that is expected to prolong life without any adverse effects and maintain satisfactory quality of life (QOL). However, the objective assessment and maintenance of a standardized evaluation of QOL are not easy. We aimed to evaluate activated autologous lymphocyte therapy for cancer dogs using the characteristics of the cultured cells and QOL as perceived by owners. In in vitro experiments, peripheral blood mononuclear cells (PBMCs) collected from healthy dogs were stimulated using anti-CD3 antibody and recombinant interleukin-2 under a closed system. The number of CD4+ and CD8+ T lymphocytes in the cultured cells was higher than that of PBMCs (P < 0.05). Natural killer activity, proenkephalin (known as the precursor of endogenous opioids) and interferon-γ mRNA in activated lymphocytes were significantly higher than in PBMCs (P < 0.05). Met-enkephalin was detected in activated lymphocytes. QOL of 58 dogs afflicted with common types of cancers in humans increased after every administration of activated lymphocyte therapy (P < 0.05). Overall, these results indicated that activated lymphocyte therapy could have beneficial effects on QOL in dogs with cancers. This was objectively evaluated and this improvement was related to presence of opioid-producing lymphocytes.


Assuntos
Analgésicos Opioides/metabolismo , Doenças do Cão/terapia , Imunoterapia Adotiva/veterinária , Neoplasias/veterinária , Linfócitos T/classificação , Linfócitos T/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Citotoxicidade Imunológica , Cães , Imunoterapia Adotiva/métodos , Neoplasias/patologia , Neoplasias/terapia
15.
Curr Top Med Chem ; 21(11): 964-975, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34061003

RESUMO

The peroxisome proliferator activated receptors (PPARs) are a superfamily of well-recognized ligand-binding nuclear receptors comprising three isoforms: PPARα, PPARγ, and PPARß/δ. In response to endogenous lipid messengers, PPARs trigger the transcription of genes related to a wider spectrum of physiological phenomena, including fatty acid oxidation, inflammation, adipogenesis, among many others. Thus, the importance of PPARs as putative protective therapy in health issues has increased the interest of studying these nuclear receptors, including the management of neurodegenerative disorders, multiple sclerosis, and likely addiction. In recent years, several pieces of evidence from animal models have demonstrated the promising role of PPARs as a critical element for interventions in addictive behaviors by reducing the reinforcing properties of addictive substances such as alcohol. However, there is a lack of data in the scope and has so far been unexplored the function of PPARs in additional drugs such as cannabis, opioids, methamphetamine, or cocaine. A similar scenario has been found for the management of binge-type eating disorders. Thus, here we review recent advances in understanding the relevance of the PPAR controlling addiction.


Assuntos
Comportamento Aditivo/tratamento farmacológico , Terapia de Alvo Molecular/métodos , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Preparações Farmacêuticas/metabolismo , Álcoois/metabolismo , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Cannabis/metabolismo , Cocaína/metabolismo , Cocaína/farmacologia , Humanos , Ligantes , Metanfetamina/metabolismo , Metanfetamina/farmacologia , Doenças Neurodegenerativas/tratamento farmacológico , Nicotina/metabolismo , Oxirredução , Isoformas de Proteínas , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo
16.
AAPS PharmSciTech ; 22(5): 186, 2021 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145510

RESUMO

The aim of this study was to investigate the effect of various parameters on the stability of butorphanol tartrate injection and to screen the optimal packaging material. The effect of the headspace oxygen levels, ampoule color, manufacturer, and size on the stability of butorphanol tartrate formulation were evaluated. The headspace oxygen levels controlled by nitrogen purging were found to be particularly effective in improving stability of the butorphanol formulation, especially below 2%. Although it is a photolabile drug, butorphanol tartrate was getting degraded at much higher extent in amber color ampoules in comparison to clear ampoules. The degradation by oxidation was found to be a free radical-mediated process catalyzed by the presence of iron ions leached from the amber ampoules. The ampoule manufacturers also had a significant effect on the stability of butorphanol. Two-milliliter ampoules provided a better stability of the butorphanol tartrate injection than 1mL ampoules as 2-mL ampoules had the lower headspace oxygen level at the same level of oxygen content. The oxidation mechanism of the butorphanol tartrate injection was investigated under various conditions, which include iron powder spiking, removal of excipients, exposure to oxygen/nitrogen, exposure to stainless steel and at different pH. Iron powder spiking, presence of citric acid, exposure to oxygen, exposure to stainless steel, and high pH accelerated the oxidative degradation. The effect of oxygen, iron ion and citric acid is in agreement with a metal-catalyzed oxidation mechanism called Udenfriend reaction. Based on the formulation test results, limiting headspace oxygen level, ampoule color, manufacturer, size, controlling iron ion contamination, and pH are recommended for formulation development. In conclusion, it can be suggested that this study can lead to a better understanding of the degradation mechanism of butorphanol tartrate; hence, it would contribute to the development of butorphanol tartrate injection with improved stability. Virous packaging materials have different effects on the stability of butorphanol tartrate injection, and the leached iron of packaging ampoules and stainless steel can trigger Udenfriend reaction with butorphanol tartrate and citric acid (CA), which lead to the oxydative degradation of butorphanol tartrate injection.


Assuntos
Analgésicos Opioides/química , Butorfanol/química , Contaminação de Medicamentos/prevenção & controle , Embalagem de Medicamentos/normas , Ferro/análise , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/metabolismo , Butorfanol/administração & dosagem , Butorfanol/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Embalagem de Medicamentos/métodos , Estabilidade de Medicamentos , Injeções Subcutâneas , Ferro/metabolismo , Oxirredução
17.
Pharmacogenomics ; 22(10): 591-602, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34100292

RESUMO

Aim: Methadone exhibits significant variability in clinical response. This study explores the genetic influence of variable methadone pharmacokinetics. Methods: This is a prospective study of methadone in children undergoing major surgery. CYP2B6 genotyping, plasma methadone and metabolite levels were obtained. Clinical outcomes include pain scores and postoperative nausea and vomiting (PONV). Results:CYP2B6 poor metabolizers (*6/*6) had >twofold lower methadone metabolism compared with normal/rapid metabolizers. The incidence of PONV was 4.7× greater with CYP2B6 rs1038376 variant. AG/GG variants of rs2279343 SNP had 2.86-fold higher incidence of PONV compared with the wild variant (AA). Nominal associations between rs10500282, rs11882424, rs4803419 and pain scores were observed. Conclusion: We have described novel associations between CYP2B6 genetic variants and perioperative methadone metabolism, and associations with pain scores and PONV.


Assuntos
Analgésicos Opioides/metabolismo , Citocromo P-450 CYP2B6/genética , Metadona/metabolismo , Assistência Perioperatória/métodos , Polimorfismo de Nucleotídeo Único/genética , Adolescente , Analgésicos Opioides/administração & dosagem , Criança , Feminino , Humanos , Masculino , Metadona/administração & dosagem , Dor Pós-Operatória/genética , Dor Pós-Operatória/metabolismo , Dor Pós-Operatória/prevenção & controle , Assistência Perioperatória/tendências , Estudos Prospectivos , Método Simples-Cego , Resultado do Tratamento
18.
Cell Mol Neurobiol ; 41(5): 977-993, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32424771

RESUMO

Mu opioid receptors (MOR-1) mediate the biological actions of clinically used opioids such as morphine, oxycodone, and fentanyl. The mu opioid receptor gene, OPRM1, undergoes extensive alternative splicing, generating multiple splice variants. One type of splice variants are truncated variants containing only six transmembrane domains (6TM) that mediate the analgesic action of novel opioid drugs such as 3'-iodobenzoylnaltrexamide (IBNtxA). Previously, we have shown that IBNtxA is a potent analgesic effective in a spectrum of pain models but lacks many side-effects associated with traditional opiates. In order to investigate the targets labeled by IBNtxA, we synthesized two arylazido analogs of IBNtxA that allow photolabeling of mouse mu opioid receptors (mMOR-1) in transfected cell lines and mMOR-1 protein complexes that may comprise the 6TM sites in mouse brain. We demonstrate that both allyl and alkyne arylazido derivatives of IBNtxA efficiently radio-photolabeled mMOR-1 in cell lines and MOR-1 protein complexes expressed either exogenously or endogenously, as well as found in mouse brain. In future, design and application of such radio-photolabeling ligands with a conjugated handle will provide useful tools for further isolating or purifying MOR-1 to investigate site specific ligand-protein contacts and its signaling complexes.


Assuntos
Analgésicos Opioides/metabolismo , Azidas/metabolismo , Encéfalo/metabolismo , Naltrexona/análogos & derivados , Marcadores de Fotoafinidade/metabolismo , Receptores Opioides/metabolismo , Analgésicos Opioides/síntese química , Animais , Azidas/síntese química , Encéfalo/efeitos dos fármacos , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Naltrexona/síntese química , Naltrexona/metabolismo , Marcadores de Fotoafinidade/síntese química , Ligação Proteica/fisiologia , Ensaio Radioligante/métodos
19.
Biochim Biophys Acta Rev Cancer ; 1875(1): 188460, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33184028

RESUMO

Colorectal cancer (CRC) is one of the most common cause of death among neoplasms around the world. The environmental factors, like diet and obesity, are crucial in CRC pathogenesis by creating cancer-favorable microenvironment and hormonal changes. Adiponectin, the adipose tissue-specific hormone, is generally considered to negatively correlate with CRC development. The interleukin 6 (IL-6) is one of the most important pro-inflammatory cytokine connected with CRC, which is strongly inflammation-associated. The opioids are variable group substantially correlated with cancers - the endogenous opioids affect immune system and cell cycle including proliferation and cell death whereas exogenous opioids are leading clinically used analgesics in terminal cancer patients. In this review we discuss the involvement of adiponectin, IL-6 and opioids in CRC pathogenesis, their link with obesity, possible cross-talk and potential novel therapeutic approach in CRC treatment.


Assuntos
Adiponectina/genética , Analgésicos Opioides/metabolismo , Neoplasias Colorretais/genética , Interleucina-6/genética , Carcinogênese/genética , Neoplasias Colorretais/patologia , Humanos , Transdução de Sinais/genética
20.
J Med Chem ; 63(24): 15709-15725, 2020 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-33271020

RESUMO

In a previously described chimeric peptide, we reported that the multifunctional opioid/neuropeptide FF (NPFF) receptor agonist 0 (BN-9) produced antinociception for 1.5 h after supraspinal administration. Herein, four cyclic disulfide analogs containing l- and/or d-type cysteine at positions 2 and 5 were synthesized. The cyclized analogs and their linear counterparts behaved as multifunctional agonists at both opioid and NPFF receptors in vitro and produced potent analgesia without tolerance development. In comparison to 0, cyclized peptide 6 exhibited sevenfold more potent µ-opioid receptor agonistic activity in vitro. Interestingly, the cyclized analog 6 possessed an improved stability in the brain and an increased blood-brain barrier permeability compared to the parent peptide 0 and produced more potent analgesia after supraspinal or subcutaneous administration with improved duration of action of 4 h. In addition, antinociceptive tolerance of analog 6 was greatly reduced after subcutaneous injection compared to fentanyl, as was the rewarding effect, withdrawal reaction, and gastrointestinal inhibition.


Assuntos
Analgésicos Opioides/química , Dissulfetos/química , Peptídeos Cíclicos/química , Receptores de Neuropeptídeos/agonistas , Receptores Opioides mu/agonistas , Sequência de Aminoácidos , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Cisteína/química , Modelos Animais de Doenças , Fentanila/farmacologia , Fentanila/uso terapêutico , Meia-Vida , Humanos , Masculino , Camundongos , Dor/tratamento farmacológico , Dor/patologia , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/uso terapêutico , Receptores de Neuropeptídeos/metabolismo , Receptores Opioides mu/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA