Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.478
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(17)2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39273354

RESUMO

Pain is an unpleasant sensory and emotional experience. Adequate pain control is often challenging, particularly in patients with chronic pain. Despite advances in pain management, drug addiction, overtreatment, or substance use disorders are not rare. Hence the need for further studies in the field. The substantial progress made over the last decade has revealed genes, signalling pathways, molecules, and neuronal networks in pain control thus opening new clinical perspectives in pain management. In this respect, data on the epigenetic modulation of opioid and cannabinoid receptors, key actors in the modulation of pain, offered new perspectives to preserve the activity of opioid and endocannabinoid systems to increase the analgesic efficacy of opioid- and cannabinoid-based drugs. Similarly, upcoming data on cannabidiol (CBD), a non-psychoactive cannabinoid in the marijuana plant Cannabis sativa, suggests analgesic, anti-inflammatory, antioxidant, anticonvulsivant and ansiolitic effects and supports its potential application in clinical contexts such as cancer, neurodegeneration, and autoimmune diseases but also in health and fitness with potential use in athletes. Hence, in this review article, we summarize the emerging epigenetic modifications of opioid and cannabinoid receptors and focus on CBD as an emerging non-psychoactive cannabinoid in pain management in clinical practice, health, and fitness.


Assuntos
Analgésicos Opioides , Canabinoides , Receptores de Canabinoides , Humanos , Analgésicos Opioides/uso terapêutico , Analgésicos Opioides/farmacologia , Canabinoides/uso terapêutico , Canabinoides/farmacologia , Receptores de Canabinoides/metabolismo , Animais , Dor/tratamento farmacológico , Dor/metabolismo , Canabidiol/uso terapêutico , Canabidiol/farmacologia , Receptores Opioides/metabolismo , Epigênese Genética/efeitos dos fármacos , Manejo da Dor/métodos , Dor Crônica/tratamento farmacológico , Dor Crônica/metabolismo , Endocanabinoides/metabolismo
2.
J Chem Inf Model ; 64(20): 8034-8051, 2024 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-39137328

RESUMO

With nearly 700 structures solved and a growing number of customized structure prediction algorithms being developed at a fast pace, G protein-coupled receptors (GPCRs) are an optimal test case for validating new approaches for the prediction of receptor active state and ligand bioactive conformation complexes. In this study, we leveraged the availability of hundreds of peptide GPCRs in the active state and both classical homology and artificial intelligence (AI) based protein modeling combined with docking and AI-based peptide structure prediction approaches to predict the nociceptin/orphanin FQ-NOP receptor active state complex (N/OFQ-NOPa). The In Silico generated hypotheses were validated via the design, synthesis, and pharmacological characterization of novel linear N/OFQ(1-13)-NH2 analogues, leading to the discovery of a novel antagonist (3B; pKB = 6.63) bearing a single ring-constrained residue in place of the Gly2-Gly3 motif of the N/OFQ message sequence (FGGF). While the experimental validation was ongoing, the availability of the Cryo-EM structure of the predicted complex enabled us to unambiguously validate the generated hypotheses. To the best of our knowledge, this is the first example of a peptide-GPCR complex predicted with atomistic accuracy (full complex Cα RMSD < 1.0 Å) and of the N/OFQ message moiety being successfully modified with a rigid scaffold.


Assuntos
Simulação de Acoplamento Molecular , Peptídeos Opioides/química , Receptores Opioides/química , Receptores Opioides/metabolismo , Peptídeos/química , Inteligência Artificial , Receptor de Nociceptina , Humanos , Conformação Proteica , Ligação Proteica , Nociceptina , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Ligantes , Modelos Moleculares , Sequência de Aminoácidos
3.
Free Radic Biol Med ; 223: 398-412, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39122201

RESUMO

Enkephalins are reportedly correlated with heart function. However, their regulation in the heart remains unexplored. This study revealed a substantial increase in circulating levels of opioid growth factor (OGF) (also known as methionine enkephalin) and myocardial expression levels of both OGF and its receptor (OGFR) in subjects treated with doxorubicin (Dox). Silencing OGFR through gene knockout or using adeno-associated virus serotype 9 carrying small hairpin RNA effectively alleviated Dox-induced cardiotoxicity (DIC) in mice. Conversely, OGF supplementation exacerbated DIC manifestations, which could be abolished by administration of the OGFR antagonist naltrexone (NTX). Mechanistically, the previously characterized OGF/OGFR/P21 axis was identified to facilitate DIC-related cardiomyocyte apoptosis. Additionally, OGFR was observed to dissociate STAT1 from the promoters of ferritin genes (FTH and FTL), thereby repressing their transcription and exacerbating DIC-related cardiomyocyte ferroptosis. To circumvent the compromised therapeutic effects of Dox on tumors owing to OGFR blockade, SiO2-based modifiable lipid nanoparticles were developed for heart-targeted delivery of NTX. The pretreatment of tumor-bearing mice with the assembled NTX nanodrug successfully provided cardioprotection against Dox toxicity without affecting Dox therapy in tumors. Taken together, this study provides a novel understanding of Dox cardiotoxicity and sheds light on the development of cardioprotectants for patients with tumors receiving Dox treatment.


Assuntos
Cardiotoxicidade , Doxorrubicina , Miócitos Cardíacos , Animais , Doxorrubicina/efeitos adversos , Camundongos , Cardiotoxicidade/metabolismo , Cardiotoxicidade/prevenção & controle , Cardiotoxicidade/genética , Cardiotoxicidade/etiologia , Cardiotoxicidade/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Humanos , Apoptose/efeitos dos fármacos , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacologia , Receptores Opioides/metabolismo , Receptores Opioides/genética , Masculino , Transdução de Sinais/efeitos dos fármacos , Nanopartículas , Camundongos Endogâmicos C57BL
4.
Adv Neurobiol ; 35: 87-106, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38874719

RESUMO

Understanding the relationship between stress and breast cancer development is essential to preventing and alleviating the cancer. Recent research has shed light on the cognitive, physiological, cellular, and molecular underpinnings of how the endorphin pathway and stress pathway affect breast cancer. This chapter consists of two parts. Part 1 will discuss the role of endorphins in breast cancer development. This includes a discussion of three topics: (1) the neurophysiological effect of endorphins on breast tumor growth in vivo, along with further experiments that will deepen our knowledge of how ß-endorphin affects breast cancer; (2) how both the opioid receptor and somatostatin receptor classes alter intracellular signaling in breast cancer cells; and (3) genetic alleles in the opioid signaling pathway that are correlated with increased breast cancer risk. Part 2 will discuss the role of endorphins in recovery from breast cancer. This includes a discussion of three topics: (1) the relationship between breast cancer diagnosis and depression; (2) the effectiveness of cognitive behavioral therapy in reducing stress in breast cancer patients; and (3) the effect of psychotherapy and exercise on preserving telomere length in breast cancer patients.


Assuntos
Neoplasias da Mama , Estresse Psicológico , Animais , Feminino , Humanos , beta-Endorfina/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Terapia Cognitivo-Comportamental , Depressão/metabolismo , Endorfinas/metabolismo , Receptores Opioides/metabolismo , Transdução de Sinais , Estresse Psicológico/metabolismo
5.
Adv Neurobiol ; 35: 45-85, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38874718

RESUMO

It has become apparent that endogenous opioids act not only as neurotransmitters and neuromodulators, but have multiple functions in the body. Activation of the opioid system by opiate drugs is associated with a risk of cancer development through direct stimulation of tumor cell proliferation and through immunosuppression. In contrast, the endogenous peptide opioid [Met5]-enkephalin, now commonly referred to as Opioid Growth Factor (OGF), negatively regulates cell proliferation in a wide number of cells during development, homeostasis, and neoplasia. This action is mediated through the opioid growth factor receptor, originally designated the zeta (ζ) opioid receptor. Further, contrary to the traditional notion of opiates as immunosuppressive, endogenous OGF has been shown to possess a number of positive immunomodulatory properties and may provide a beneficial effect in cancer by augmenting the activity of cells involved in both innate and acquired immunity. Taken together, the evidence supports consideration of opioid peptides such as OGF as new strategies for cancer therapy.


Assuntos
Neoplasias , Receptores Opioides , Animais , Humanos , Proliferação de Células/efeitos dos fármacos , Encefalina Metionina/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Peptídeos Opioides/metabolismo , Receptores Opioides/metabolismo
6.
Cell Rep ; 43(6): 114343, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38865247

RESUMO

Activation of prepronociceptin (PNOC)-expressing neurons in the arcuate nucleus (ARC) promotes high-fat-diet (HFD)-induced hyperphagia. In turn, PNOCARC neurons can inhibit the anorexic response of proopiomelanocortin (POMC) neurons. Here, we validate the necessity of PNOCARC activity for HFD-induced inhibition of POMC neurons in mice and find that PNOCARC-neuron-dependent inhibition of POMC neurons is mediated by gamma-aminobutyric acid (GABA) release. When monitoring individual PNOCARC neuron activity via Ca2+ imaging, we find a subpopulation of PNOCARC neurons that is inhibited upon gastrointestinal calorie sensing and disinhibited upon HFD feeding. Combining retrograde rabies tracing and circuit mapping, we find that PNOC neurons from the bed nucleus of the stria terminalis (PNOCBNST) provide inhibitory input to PNOCARC neurons, and this inhibitory input is blunted upon HFD feeding. This work sheds light on how an increase in caloric content of the diet can rewire a neuronal circuit, paving the way to overconsumption and obesity development.


Assuntos
Dieta Hiperlipídica , Hiperfagia , Núcleos Septais , Animais , Hiperfagia/metabolismo , Camundongos , Núcleos Septais/metabolismo , Neurônios/metabolismo , Masculino , Ácido gama-Aminobutírico/metabolismo , Pró-Opiomelanocortina/metabolismo , Neurônios GABAérgicos/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Camundongos Endogâmicos C57BL , Precursores de Proteínas , Receptores Opioides
7.
Food Funct ; 15(9): 4773-4784, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38469873

RESUMO

Raphanus sativus L. cv. Sango, commonly known as red radish, is widely consumed around the world as a vegetable, but its benefit in pain relief is not sufficiently investigated. This study aimed to evaluate the antinociceptive effects of R. sativus and a possible mechanism of action. An aqueous extract of R. sativus sprouts (AERSS) was investigated by parenteral (10, 30, and 100 mg kg-1, i.p.) and enteral (500 mg kg-1, p.o.) administration in the neurogenic and inflammatory phases of the formalin test, where gastric damage was also evaluated as a possible adverse effect. Ketorolac (5 mg kg-1, i.p.) was used as the reference drug. Endogenous opioid and 5-HT1A serotonin receptors, as well as the cAMP/NO-cGMP pathways, were explored in the study of a possible mechanism of action by using their corresponding antagonists: naloxone, 1 mg kg-1, i.p., WAY100635, 1 mg kg-1, i.p., and enzymatic activators or inhibitors, respectively. Sulforaphane (SFN), a known bioactive metabolite, was analyzed using electroencephalography (EEG) to evidence its central involvement. A significant and dose-dependent antinociceptive activity was observed with the AERSS resembling the antinociceptive effect of the reference drug, with an equivalent significant response with a dose of 500 mg kg-1, p.o. without causing gastric damage. The participation of the endogenous opioid and 5-HT1A serotonin receptors at central and peripheral levels was also observed, with a differential participation of cAMP/NO-cGMP. SFN as one metabolite produced significant changes in the EEG analysis, reinforcing its effects on the CNS. Our preclinical evidence supports the benefits of consuming Raphanus sativus cv. Sango sprouts for pain relief.


Assuntos
Analgésicos , Isotiocianatos , Extratos Vegetais , Raphanus , Transdução de Sinais , Animais , Humanos , Masculino , Camundongos , Analgésicos/farmacologia , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Isotiocianatos/farmacologia , Dor/tratamento farmacológico , Extratos Vegetais/farmacologia , Raphanus/química , Receptor 5-HT1A de Serotonina/metabolismo , Receptores Opioides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfóxidos/farmacologia
8.
Behav Pharmacol ; 35(2-3): 114-121, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38451023

RESUMO

We hypothesized that opioid receptor antagonists would inhibit motivated behavior produced by a natural reward. To evaluate motivated responses to a natural reward, mice were given access to running wheels for 71.5 h in a multi-configuration testing apparatus. In addition to a running wheel activity, locomotor activity (outside of the wheel), food and water intake, and access to a food container were measured in the apparatus. Mice were also tested separately for novel-object exploration to investigate whether naloxone affects behavior unrelated to natural reward. In untreated mice wheel running increased from day 1 to day 3. The selective µ-opioid receptor antagonist ß-funaltrexamine (ß-FNA) (5 mg/kg) slightly decreased wheel running, but did not affect the increase in wheel running from day 1 to day 3. The non-selective opioid receptor antagonist naloxone produced a greater reduction in wheel running than ß-FNA and eliminated the increase in wheel running that occurred over time in the other groups. Analysis of food access, locomotor behavior, and behavior in the novel-object test suggested that the reduction in wheel running was selective for this highly reinforcing behavior. These results indicate that opioid receptor antagonism reduces responses to the natural rewarding effects of wheel running and that these effects involve multiple opioid receptors since the non-selective opioid receptor antagonist had greater effects than the selective µ-opioid receptor antagonist. It is possible that at the doses employed, other receptor systems than opioid receptors might be involved, at least in part, in the effect of naloxone and ß-FNA.


Assuntos
Atividade Motora , Antagonistas de Entorpecentes , Animais , Camundongos , Antagonistas de Entorpecentes/farmacologia , Motivação , Naloxona/farmacologia , Receptores Opioides
9.
Cells ; 13(3)2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38334624

RESUMO

The recent emphasis on circadian rhythmicity in critical skin cell functions related to homeostasis, regeneration and aging has shed light on the importance of the PER2 circadian clock gene as a vital antitumor gene. Furthermore, delta-opioid receptors (DOPrs) have been identified as playing a crucial role in skin differentiation, proliferation and migration, which are not only essential for wound healing but also contribute to cancer development. In this study, we propose a significant association between cutaneous opioid receptor (OPr) activity and circadian rhythmicity. To investigate this link, we conducted a 48 h circadian rhythm experiment, during which RNA samples were collected every 5 h. We discovered that the activation of DOPr by its endogenous agonist Met-Enkephalin in N/TERT-1 keratinocytes, synchronized by dexamethasone, resulted in a statistically significant 5.6 h delay in the expression of the core clock gene PER2. Confocal microscopy further confirmed the simultaneous nuclear localization of the DOPr-ß-arrestin-1 complex. Additionally, DOPr activation not only enhanced but also induced a phase shift in the rhythmic binding of ß-arrestin-1 to the PER2 promoter. Furthermore, we observed that ß-arrestin-1 regulates the transcription of its target genes, including PER2, by facilitating histone-4 acetylation. Through the ChIP assay, we determined that Met-Enkephalin enhances ß-arrestin-1 binding to acetylated H4 in the PER2 promoter. In summary, our findings suggest that DOPr activation leads to a phase shift in PER2 expression via ß-arrestin-1-facilitated chromatin remodeling. Consequently, these results indicate that DOPr, much like its role in wound healing, may also play a part in cancer development by influencing PER2.


Assuntos
Neoplasias , Receptores Opioides , Humanos , beta-Arrestinas , Receptores Opioides/genética , Queratinócitos , Ritmo Circadiano/fisiologia , beta-Arrestina 1 , Encefalina Metionina
10.
Pharmacol Rep ; 76(1): 112-126, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38236555

RESUMO

BACKGROUND: Colorectal cancer (CRC) is one of the leading causes of death globally. Multiple factors may contribute to the pathogenesis of CRC, including the abnormalities in the functioning of the endogenous opioid system (EOS) or adiponectin-related signaling. The aim of our study was to evaluate if differences in the expression of opioid receptors (ORs) influence the development of CRC and if modulation of adiponectin receptors using AdipoRon, a selective AdipoR1 receptor agonist, affects colorectal carcinogenesis. METHODS: Naltrexone, an opioid receptor antagonist, was injected intraperitoneally every second day for 2 weeks, at the dose of 1 mg/kg in healthy Balb/C mice to induce changes in ORs expression. CRC was induced by a single intraperitoneal injection of azoxymethane (AOM) and the addition of dextran sodium sulfate (DSS) into drinking water in three-week cycles. The development of CRC was assessed using macro- and microscopic scoring and molecular analysis (RT qPCR, ELISA) after 14 weeks. RESULTS: Naltrexone significantly increased the mRNA expression of Oprm1, Oprd1, and Oprk1 in the mouse colon and in the brain (non-significantly). The pretreatment of mice with naltrexone aggravated the course of CRC (as indicated by tumor area, colon thickness, and spleen weight). The level of circulatory adiponectin was lowered in mice with CRC and increased in the colon as compared with healthy mice. The ß-endorphin level was increased in the plasma of mice with CRC and decreased in the colon as compared to healthy mice. AdipoRon, AdipoR1 agonist, worsened the CRC development, and pretreatment with naltrexone enhanced this negative effect in mice. CRC did not affect the expression of the Adipor1 gene, but the Adipor1 level was increased in mice pretreated with naltrexone (AOM/DSS and healthy mice). AdipoRon did not influence the expression of opioid receptors at the mRNA level in the colon of mice with CRC. The mRNA expression of Ptgs2, Il6, Nos2, Il1b, Il18, Gsdmd, and Rela was increased in mice with CRC as compared to the healthy colon. AdipoRon significantly decreased mRNA expression of Ptgs2, Il6, Il1b, and Il18 as compared to CRC mice. CONCLUSION: EOS and adiponectin-related signaling may play a role in the pathogenesis of CRC and these systems may present some additivity during carcinogenesis.


Assuntos
Neoplasias Associadas a Colite , Colite , Neoplasias Colorretais , Camundongos , Animais , Interleucina-18 , Analgésicos Opioides/efeitos adversos , Interleucina-6 , Adipocinas , Naltrexona/farmacologia , Adiponectina/efeitos adversos , Ciclo-Oxigenase 2 , Carcinogênese , Azoximetano/toxicidade , Modelos Animais de Doenças , Receptores Opioides/genética , RNA Mensageiro , Sulfato de Dextrana , Neoplasias Colorretais/genética , Camundongos Endogâmicos C57BL , Colite/induzido quimicamente
11.
Behav Pharmacol ; 35(2-3): 66-78, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37578392

RESUMO

It has been demonstrated that the nucleus accumbens (NAc) plays an important role in modulation of nociception due to its extensive connections with different regions of the brain. In addition, this nucleus receives histaminergic projections from tuberomammillary nucleus. Considering the role of the central histaminergic system in nociception, the effect of histamine and its H 2 and H 3 receptors agonist and antagonist microinjections into the NAc on orofacial formalin nociception was investigated. In male Wistar rats, using stereotaxic surgery, two guide cannulas were bilaterally implanted into the right and left sides of the NAc. Diluted formalin solution (1.5%, 50 µl) injection into the vibrissa pad led to orofacial nociception. Immediately after injection, face rubbing was observed at 3-min blocks for 45 min. Orofacial formalin nociception was characterized by a biphasic nociceptive response (first phase: 0-3 min and second phase: 15-33 min). Microinjections of histamine (0.5 and 1 µg/site), dimaprit (1 µg/site, H 2 receptor agonist) and thioperamide (2 µg/site, H 3 receptor antagonist) attenuated both phases of formalin orofacial nociception. Prior microinjection of famotidine (2 µg/site) inhibited the antinociceptive effects of dimaprit (1 µg/site). Furthermore, comicroinjection of thioperamide (2 µg/site) and immepip (1 µg/site) prevented thioperamide (2 µg/site)-induced antinociception. Naloxone (2 µg/site) also prevented histamine, dimaprit- and thioperamide-induced antinociception. The results of this study demonstrate that at the level of the NAc, histamine and its H 2 and H 3 receptors are probably involved in the modulation of orofacial nociception with an opioid system-dependent mechanism.


Assuntos
Histamina , Receptores Opioides , Ratos , Animais , Masculino , Histamina/farmacologia , Ratos Wistar , Receptores Opioides/metabolismo , Núcleo Accumbens/metabolismo , Nociceptividade , Formaldeído/efeitos adversos , Dimaprit/efeitos adversos , Dor Facial/tratamento farmacológico , Receptores Histamínicos H2/metabolismo
12.
Psychopharmacology (Berl) ; 241(2): 291-304, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38049617

RESUMO

RATIONALE: While morphine has important therapeutic value it is also one of the most widely abused drugs in the world. As a newly discovered style of cell death, ferroptosis is involved in the occurrence and development of many diseases, however, the current understanding of the relationship between ferroptosis and morphine is still limited. OBJECTIVE: To clarify the role of opioid receptors in morphine-induced ferroptosis and to investigate the role of NRF2 in morphine-induced ferroptosis. METHODS: We first used different doses of morphine (0, 0.5, 1, and 1.5 mM) to investigate morphine-induced ferroptosis in SH-SY5Y cells, and we choose 1.5 mM morphine for subsequent experiments. We next inhibited opioid receptors and NRF2 separately and examined their influence on morphine-induced ferroptosis. Finally, we tested morphine-induced insufficient autophagy. RESULTS: Morphine triggered ferroptosis in a dose-dependent manner, which could be significantly rescued by the ferroptosis-specific inhibitor DFO. Moreover, GPX4 rather than xCT antiporter might be involved in morphine-induced ferroptosis. We also found naloxone could inhibit morphine-induced ferroptosis. Interestingly, our results demonstrated that NRF2 could promote rather than defend morphine-induced ferroptosis; this may be due to the increased p62-related insufficient autophagy. CONCLUSION: Morphine-induced ferroptosis is regulated by the opioid receptor and GPX4 rather than the xCT antiporter. NRF2-mediated ferroptosis in morphine-exposed cells may stem from increased p62-related insufficient autophagy.


Assuntos
Ferroptose , Neuroblastoma , Humanos , Antiporters , Autofagia , Morfina/farmacologia , Fator 2 Relacionado a NF-E2 , Receptores Opioides
13.
Reg Anesth Pain Med ; 49(3): 200-208, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-37353355

RESUMO

INTRODUCTION: Tramadol, a weak opioid anesthetic, is used for pain management in patients with cancer, but the effects of tramadol on cancer via µ-opioid receptor are still unknown. We assessed the effects of tramadol on pancreatic ductal adenocarcinoma using transgenic mice (LSL-KrasG12D/+; Trp53flox/flox; Pdx-1cre/+ ). METHODS: Six-week-old transgenic mice were orally administered 10 mg/kg/day tramadol (n=12), 10 mg/kg/day tramadol and 1 mg/kg/day naltrexone (n=9), or vehicle water (n=14) until the humane endpoint. Cancer-related pain and plasma cytokine levels were assessed by the mouse grimace scale and cytokine array, respectively. Tumor status was determined histopathologically. Tramadol's effects on proliferation and invasion in pancreatic ductal adenocarcinoma cell lines were studied in vitro. RESULTS: Tramadol with/without naltrexone improved mouse grimace scale scores while decreasing inflammatory cytokines such as tumor necrosis factor-α and interleukin-6. Proliferative Ki-67 and cyclins decreased by tramadol, while local M1-like tumor-associated macrophages increased by tramadol, which was blocked by naltrexone. Meanwhile, tramadol with/without naltrexone reduced juxta-tumoral cancer-associated fibroblasts and M2-like tumor-associated macrophages. Tumor-associated neutrophils, natural killers, and cytotoxic T cells were not altered. Tramadol decreased the proliferative and invasive potentials of pancreatic ductal adenocarcinoma cell lines via decreasing cyclins/cyclin-dependent kinases, which was partially reversed by naltrexone. CONCLUSIONS: These findings imply that tramadol might be a useful anesthetic for pancreatic ductal adenocarcinoma: inhibiting the proliferation and invasion along with increasing antitumor M1-like tumor-associated macrophages via the µ-opioid receptor, while improving cancer-associated pain possibly through the antitumor effects with the decrease of inflammatory cytokines.


Assuntos
Anestésicos , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Tramadol , Humanos , Camundongos , Animais , Tramadol/farmacologia , Tramadol/uso terapêutico , Naltrexona , Receptores Opioides , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Camundongos Transgênicos , Citocinas , Ciclinas
14.
Anesthesiology ; 140(4): 765-785, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38118180

RESUMO

BACKGROUND: The role of nerve growth factor (NGF)/tyrosine kinase A receptor (TrKA) signaling, which is activated in a variety of pain states, in regulating membrane-associated δ-opioid receptor (mDOR) expression is poorly understood. The hypothesis was that elevated NGF in bone cancer tumors could upregulate mDOR expression in spinal cord neurons and that mDOR agonism might alleviate bone cancer pain. METHODS: Bone cancer pain (BCP) was induced by inoculating Lewis lung carcinoma cells into the femoral marrow cavity of adult C57BL/6J mice of both sexes. Nociceptive behaviors were evaluated by the von Frey and Hargreaves tests. Protein expression in the spinal dorsal horn of animals was measured by biochemical analyses, and excitatory synaptic transmission was recorded in miniature excitatory synaptic currents. RESULTS: The authors found that mDOR expression was increased in BCP mice (BCP vs. sham, mean ± SD: 0.18 ± 0.01 g vs. mean ± SD: 0.13 ± 0.01 g, n = 4, P < 0.001) and that administration of the DOR agonist deltorphin 2 (Del2) increased nociceptive thresholds (Del2 vs. vehicle, median [25th, 75th percentiles]: 1.00 [0.60, 1.40] g vs. median [25th, 75th percentiles]: 0.40 [0.16, 0.45] g, n = 10, P = 0.001) and reduced miniature excitatory synaptic current frequency in lamina II outer neurons (Del2 vs. baseline, mean ± SD: 2.21 ± 0.81 Hz vs. mean ± SD: 2.43 ± 0.90 Hz, n = 12, P < 0.001). Additionally, NGF expression was increased in BCP mice (BCP vs. sham, mean ± SD: 0.36 ± 0.03 vs. mean ± SD: 0.16 ± 0.02, n = 4, P < 0.001), and elevated NGF was associated with enhanced mDOR expression via TrKA signaling. CONCLUSIONS: Activation of mDOR produces analgesia that is dependent on the upregulation of the NGF/TrKA pathway by increasing mDOR levels under conditions of BCP in mice.


Assuntos
Analgesia , Neoplasias Ósseas , Dor do Câncer , Ratos , Masculino , Feminino , Camundongos , Animais , Dor do Câncer/tratamento farmacológico , Receptores Proteína Tirosina Quinases , Ratos Sprague-Dawley , Fator de Crescimento Neural/metabolismo , Camundongos Endogâmicos C57BL , Dor , Neoplasias Ósseas/complicações , Corno Dorsal da Medula Espinal , Receptores Opioides
15.
Epigenetics ; 19(1): 2294515, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38118075

RESUMO

Chronic alcohol consumption may alter mRNA methylation and expression levels of genes related to addiction and reward in the brain, potentially contributing to alcohol tolerance and dependence. Neuron-like (SH-SY5Y) and non-neuronal (SW620) cells were utilized as models to examine chronic intermittent ethanol (CIE) exposure-induced global m6A RNA methylation changes, as well as m6A mRNA methylation changes around the stop codon of three opioid receptor genes (OPRM1, OPRD1, and OPRK1), which are known to regulate pain, reward, and addiction behaviours. CIE exposure for three weeks significantly increased global RNA methylation levels in both SH-SY5Y (t = 3.98, P = 0.007) and SW620 (t = 2.24, P = 0.067) cells. However, a 3-week CIE exposure resulted in hypomethylation around mRNA stop codon regions of OPRM1 and OPRD1 in both cell lines [OPRM1(SH-SY5Y): t = -5.05, P = 0.0005; OPRM1(SW620): t = -3.19, P = 0.013; OPRD1(SH-SY5Y): t = -13.43, P < 0.00001; OPRD1(SW620): t = -4.00, P = 0.003]. Additionally, mRNA expression levels of OPRM1, OPRD1, and OPRK1 were downregulated (corresponding to mRNA hypomethylation) in both SH-SY5Y and SW620 cells after a 3-week CIE exposure. The present study demonstrated that chronic ethanol exposure altered global RNA methylation levels, as well as mRNA methylation and expression levels of opioid receptor genes in both neuron-like and non-neuronal cells. Our findings suggest a potential epitranscriptomic mechanism by which chronic alcohol consumption remodels the expression of reward-related and alcohol responsive genes in the brain, thus increasing the risk of alcohol use disorder development.Abbreviations: OPRM1: the µ-opioid receptor; OPRD1: the δ-opioid receptor; OPRK1: the κ-opioid receptor; CIE: chronic intermittent ethanol exposure; CIE+WD: chronic intermittent ethanol exposure followed by a 24-hr withdrawal; SH-SY5Y: human neuroblastoma cell Line; SW620: human colon carcinoma cell line; RT-qPCR: reverse transcription followed by quantitative polymerase reaction; MazF-RT-qPCR: MazF digestion followed by RT-qPCR.


Assuntos
Neuroblastoma , Receptores Opioides , Humanos , Receptores Opioides/genética , Códon de Terminação , Etanol/farmacologia , RNA Mensageiro/genética , Metilação de DNA , Neuroblastoma/genética
16.
Int Immunopharmacol ; 124(Pt B): 110944, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37801968

RESUMO

Opioids are employed in the management of chemotherapy-induced neuropathic pain (CINP) when other pain management approaches have failed and proven ineffective. However, their use in CINP is generally considered as a second-line or adjunctive therapy owing to their central side effects and development of tolerance with their long-term usage. Targeting peripheral sites may offer several advantages over the conventional CNS-based approaches as peripheral targets modulate pain signals at their source, thereby relieving pain with higher specificity, efficacy and minimizing adverse effects associated with off-site CNS actions. Therefore, present study was designed with an aim to investigate the effect of loperamide, a peripherally acting mu-opioid receptor agonist, on paclitaxel-induced neuropathic pain in rats and elucidate its underlying mechanism. Loperamide treatment significantly attenuated mechanical, and cold hypersensitivity and produced significant place preference behaviour in neuropathic rats indicating its potential to treat both evoked and spontaneous pain. More importantly, loperamide treatment in naïve rats did not produce place preference to drug-paired chamber pointing towards its non-addictive analgesic potential. Further, molecular investigations revealed increased expression of ion channels such as TRPA1, TRPM8; voltage-gated sodium channels (VGSCs) and neuroinflammatory markers in the dorsal root ganglion (DRG) and lumbar (L4-L5) spinal cord of neuropathic rats, which was significantly downregulated upon loperamide treatment. These findings collectively suggest that activation of peripheral mu-opioid receptors contributes to the amelioration of both evoked and spontaneous pain in neuropathic rats by downregulating TRP channels and VGSCs along with suppression of oxido-nitrosative stress and neuroinflammatory cascade.


Assuntos
Antineoplásicos , Neuralgia , Ratos , Animais , Loperamida/uso terapêutico , Loperamida/farmacologia , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Analgésicos Opioides/uso terapêutico , Analgésicos Opioides/farmacologia , Antineoplásicos/efeitos adversos , Receptores Opioides
17.
Sci Rep ; 13(1): 18164, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37875567

RESUMO

Opioid addiction and the opioid overdose epidemic are becoming more serious, and the development of therapeutic agents is essential for the pharmacological treatment of substance use disorders. The κ-opioid receptor (KOP) is a member of the opioid receptor system that has been gaining attention as a promising molecular target for the treatment of numerous human disorders, including pain, depression, anxiety, and drug addiction. Here, we biologically and pharmacologically evaluated a novel azepane-derived ligand, NP-5497-KA, as a selective KOP agonist. NP-5497-KA had 1000-fold higher selectivity for the KOP over the µ-opioid receptor (MOP), which was higher than nalfurafine (KOP/MOP: 65-fold), and acted as a selective KOP full agonist in the 3',5'-cyclic adenosine monophosphate assay. The oral administration of NP-5497-KA (1-10 mg/kg) dose-dependently suppressed morphine-induced conditioned place preference in C57BL/6 J mice, and its effects were comparable to an intraperitoneal injection of nalfurafine (1-10 µg/kg). Nalfurafine (10 µg/kg) significantly inhibited rotarod performance, whereas NP-5497-KA (10 mg/kg) exerted no effect on rotarod performance. These results indicate that NP-5497-KA may be a novel option for the treatment of opioid use disorder with fewer side effects.


Assuntos
Morfina , Transtornos Relacionados ao Uso de Opioides , Camundongos , Animais , Humanos , Morfina/farmacologia , Camundongos Endogâmicos C57BL , Receptores Opioides , Receptores Opioides mu/agonistas , Receptores Opioides kappa/agonistas , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Recompensa , Analgésicos Opioides/farmacologia
18.
Nat Commun ; 14(1): 5632, 2023 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-37704594

RESUMO

With concurrent global epidemics of chronic pain and opioid use disorders, there is a critical need to identify, target and manipulate specific cell populations expressing the mu-opioid receptor (MOR). However, available tools and transgenic models for gaining long-term genetic access to MOR+ neural cell types and circuits involved in modulating pain, analgesia and addiction across species are limited. To address this, we developed a catalog of MOR promoter (MORp) based constructs packaged into adeno-associated viral vectors that drive transgene expression in MOR+ cells. MORp constructs designed from promoter regions upstream of the mouse Oprm1 gene (mMORp) were validated for transduction efficiency and selectivity in endogenous MOR+ neurons in the brain, spinal cord, and periphery of mice, with additional studies revealing robust expression in rats, shrews, and human induced pluripotent stem cell (iPSC)-derived nociceptors. The use of mMORp for in vivo fiber photometry, behavioral chemogenetics, and intersectional genetic strategies is also demonstrated. Lastly, a human designed MORp (hMORp) efficiently transduced macaque cortical OPRM1+ cells. Together, our MORp toolkit provides researchers cell type specific genetic access to target and functionally manipulate mu-opioidergic neurons across a range of vertebrate species and translational models for pain, addiction, and neuropsychiatric disorders.


Assuntos
Analgesia , Dor Crônica , Células-Tronco Pluripotentes Induzidas , Animais , Humanos , Camundongos , Ratos , Macaca , Receptores Opioides , Receptores Opioides mu/genética , Transgenes
19.
Int J Mol Sci ; 24(13)2023 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-37446064

RESUMO

This review article offers an outlook on the use of opioids as therapeutics for treating several diseases, including cancer and non-cancer pain, and focuses the analysis on the opportunity to target opioid receptors for treating opioid use disorder (OUD), drug withdrawal, and addiction. Unfortunately, as has been well established, the use of opioids presents a plethora of side effects, such as tolerance and physical and physiological dependence. Accordingly, considering the great pharmacological potential in targeting opioid receptors, the identification of opioid receptor ligands devoid of most of the adverse effects exhibited by current therapeutic agents is highly necessary. To this end, herein, we analyze some interesting molecules that could potentially be useful for treating OUD, with an in-depth analysis regarding in vivo studies and clinical trials.


Assuntos
Comportamento Aditivo , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Transtornos Relacionados ao Uso de Opioides , Síndrome de Abstinência a Substâncias , Humanos , Analgésicos Opioides/efeitos adversos , Receptores Opioides , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/tratamento farmacológico
20.
Life Sci ; 328: 121892, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37364634

RESUMO

The apelin receptor (APJ) and the opioid-related nociceptin receptor 1 (ORL1) are family A G protein-coupled receptors that participate in a variety of physiological processes. The distribution and function of APJ and ORL1 in the nervous system and peripheral tissues are similar; however, the detailed mechanism of how these two receptors modulate signaling and physiological effects remains unclear. Here, we examined whether APJ and ORL1 form dimers, and investigated signal transduction pathways. The endogenous co-expression of APJ and ORL1 in SH-SY5Y cells was confirmed by western blotting and RT-PCR. Bioluminescence and fluorescence resonance energy transfer assays, as well as a proximity ligation assay and co-immunoprecipitation experiments, demonstrated that APJ and ORL1 heterodimerize in HEK293 cells. We found that the APJ-ORL1 heterodimer is selectively activated by apelin-13, which causes the dimer to couple to Gαi proteins and reduce the recruitment of GRKs and ß-arrestins to the dimer. We showed that the APJ-ORL1 dimer exhibits biased signaling, in which G protein-dependent signaling pathways override ß-arrestin-dependent signaling pathways. Our results demonstrate that the structural interface of the APJ-ORL1 dimer switches from transmembrane domain TM1/TM2 in the inactive state to TM5 in the active state. We used mutational analysis and BRET assays to identify key residues in TM5 (APJ L2185.55, APJ I2245.61, and ORL1 L2295.52) responsible for the receptor-receptor interaction. These results provide important information on the APJ-ORL1 heterodimer and may assist the design of new drugs targeting biased signaling pathways for treatment of pain and cardiovascular and metabolic diseases.


Assuntos
Neuroblastoma , Humanos , Apelina/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides/genética , Receptores Opioides/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA