Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Publication year range
1.
J Transl Med ; 22(1): 630, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970055

ABSTRACT

Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a debilitating disease with a broad overlap of symptomatology with Post-COVID Syndrome (PCS). Despite the severity of symptoms and various neurological, cardiovascular, microvascular, and skeletal muscular findings, no biomarkers have been identified. The Transient receptor potential melastatin 3 (TRPM3) channel, involved in pain transduction, thermosensation, transmitter and neuropeptide release, mechanoregulation, vasorelaxation, and immune defense, shows altered function in ME/CFS. Dysfunction of TRPM3 in natural killer (NK) cells, characterized by reduced calcium flux, has been observed in ME/CFS and PCS patients, suggesting a role in ineffective pathogen clearance and potential virus persistence and autoimmunity development. TRPM3 dysfunction in NK cells can be improved by naltrexone in vitro and ex vivo, which may explain the moderate clinical efficacy of low-dose naltrexone (LDN) treatment. We propose that TRPM3 dysfunction may have a broader involvement in ME/CFS pathophysiology, affecting other organs. This paper discusses TRPM3's expression in various organs and its potential impact on ME/CFS symptoms, with a focus on small nerve fibers and the brain, where TRPM3 is involved in presynaptic GABA release.


Subject(s)
Fatigue Syndrome, Chronic , Naltrexone , TRPM Cation Channels , Humans , Fatigue Syndrome, Chronic/drug therapy , TRPM Cation Channels/metabolism , Naltrexone/therapeutic use , Naltrexone/pharmacology , Naltrexone/administration & dosage , Animals , Dose-Response Relationship, Drug , Treatment Outcome
2.
Semin Neurol ; 44(4): 441-451, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38848746

ABSTRACT

The rates of opioid use and opioid related deaths are escalating in the United States. Despite this, evidence-based treatments for Opioid Use Disorder are underutilized. There are three medications FDA approved for treatment of Opioid Use Disorder: Methadone, Buprenorphine, and Naltrexone. This article reviews the history, criteria, and mechanisms associated with Opioid Use Disorder. Pertinent pharmacology considerations, treatment strategies, efficacy, safety, and challenges of Methadone, Buprenorphine, and Naltrexone are outlined. Lastly, a practical decision making algorithm is discussed to address pertinent psychiatric and medical comorbidities when prescribing pharmacology for Opioid Use Disorder.


Subject(s)
Buprenorphine , Methadone , Naltrexone , Narcotic Antagonists , Opiate Substitution Treatment , Opioid-Related Disorders , Humans , Naltrexone/therapeutic use , Naltrexone/pharmacology , Buprenorphine/therapeutic use , Buprenorphine/pharmacology , Opiate Substitution Treatment/methods , Opioid-Related Disorders/drug therapy , Methadone/therapeutic use , Narcotic Antagonists/therapeutic use , Narcotic Antagonists/pharmacology , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/pharmacology
3.
Appetite ; 200: 107504, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38768926

ABSTRACT

The dynorphin peptides are the endogenous ligands for the kappa opioid receptor (KOR) and regulate food intake. Administration of dynorphin-A1-13 (DYN) in the paraventricular hypothalamic nucleus (PVN) increases palatable food intake, and this effect is blocked by co-administration of the orexin-A neuropeptide, which is co-released with DYN in PVN from neurons located in the lateral hypothalamus. While PVN administration of DYN increases palatable food intake, whether it increases food-seeking behaviors has yet to be examined. We tested the effects of DYN and norBNI (a KOR antagonist) on the seeking and consumption of sucrose using a progressive ratio (PR) and demand curve (DC) tasks. In PVN, DYN did not alter the sucrose breaking point (BP) in the PR task nor the elasticity or intensity of demand for sucrose in the DC task. Still, DYN reduced the delay in obtaining sucrose and increased licks during sucrose intake in the PR task, irrespective of the co-administration of orexin-A. In PVN, norBNI increased the delay in obtaining sucrose and reduced licks during sucrose intake in the PR task while increasing elasticity without altering intensity of demand in the DC task. However, subcutaneous norBNI reduced the BP for sucrose and increased the delay in obtaining sucrose in the PR task while reducing the elasticity of demand. Together, these data show different effects of systemic and PVN blockade of KOR on food-seeking, consummatory behaviors, and incentive motivation for sucrose and suggest that KOR activity in PVN is necessary but not sufficient to drive seeking behaviors for palatable food.


Subject(s)
Dynorphins , Motivation , Paraventricular Hypothalamic Nucleus , Receptors, Opioid, kappa , Receptors, Opioid, kappa/metabolism , Dynorphins/pharmacology , Dynorphins/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Animals , Male , Motivation/drug effects , Orexins , Rats , Rats, Sprague-Dawley , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Eating/drug effects , Eating/physiology , Eating/psychology , Sucrose , Feeding Behavior/drug effects , Feeding Behavior/psychology , Narcotic Antagonists/pharmacology
5.
Sci Rep ; 14(1): 48, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167957

ABSTRACT

Hepatic osteodystrophy, a prevalent manifestation of metabolic bone disease, can arise in the context of chronic liver disease. The THBS1-eNOS-NO signaling pathway plays a pivotal role in the maturation of osteoclast precursors. This study aimed to investigate the impact of Naltrexone (NTX) on bone loss by examining the THBS1-eNOS-NO signaling pathways in bile duct ligated (BDL) rats. Male Wistar rats were randomly divided into five groups (n = 10 per group): control, sham-operated + normal saline, BDL + normal saline, sham-operated + NTX (10 mg/kg), and BDL + NTX. Parameters related to liver injury were measured at the study's conclusion, and Masson-trichrome staining was employed to evaluate collagen deposition in liver tissue. Bone THBS-1 and endothelial nitric oxide synthase (eNOS) expression levels were measured using real-time PCR, while the level of bone nitric oxide (NO) was assessed through a colorimetric assay. NTX treatment significantly attenuated the BDL-induced increase in circulating levels of liver enzymes and bilirubin. THBS-1 expression levels, elevated after BDL, were significantly suppressed following NTX administration in the BDL + NTX group. Despite no alterations in eNOS expression between groups, the bone NO level, significantly decreased in the BDL group, was significantly reduced by NTX in the BDL + NTX group. This study partly provides insights into the possible molecular mechanisms in BDL-induced osteoporosis and highlights the modulating effect of NTX on these pathways. Further research is needed to establish the impact of NTX on histomorphometric indexes.


Subject(s)
Naltrexone , Nitric Oxide Synthase Type III , Rats , Male , Animals , Naltrexone/pharmacology , Nitric Oxide Synthase Type III/metabolism , Saline Solution , Rats, Wistar , Bile Ducts/surgery , Liver/metabolism , Ligation , Liver Cirrhosis/pathology
6.
J Med Chem ; 67(4): 3127-3143, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38306598

ABSTRACT

Disruptions in the toll-like receptor 4 (TLR4) signaling pathway are linked to chronic inflammation, neuropathic pain, and drug addiction. (+)-Naltrexone, an opioid-derived TLR4 antagonist with a (+)-isomer configuration, does not interact with classical opioid receptors and has moderate blood-brain barrier permeability. Herein, we developed a concise 10-step synthesis for (+)-naltrexone and explored its precursors, (+)-14-hydroxycodeinone (1) and (+)-14-hydroxymorphinone (3). These precursors exhibited TLR4 antagonistic activities 100 times stronger than (+)-naltrexone, particularly inhibiting the TLR4-TRIF pathway. In vivo studies showed that these precursors effectively reduced behavioral effects of morphine, like sensitization and conditioned place preference by suppressing microglial activation and TNF-α expression in the medial prefrontal cortex and ventral tegmental area. Additionally, 3 displayed a longer half-life and higher oral bioavailability than 1. Overall, this research optimized (+)-naltrexone synthesis and identified its precursors as potent TLR4 antagonists, offering potential treatments for morphine addiction.


Subject(s)
Morphine Dependence , Naltrexone , Rats , Animals , Humans , Naltrexone/pharmacology , Toll-Like Receptor 4 , Morphine Dependence/drug therapy , Rats, Sprague-Dawley , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Morphine/pharmacology , Analgesics, Opioid/therapeutic use
7.
Sci Rep ; 14(1): 14029, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38890353

ABSTRACT

Binge drinking (BD) contributes strongly to the harms of alcohol use disorder. Most rodent models do not result in binge-level blood alcohol concentrations (BACs), and to better understand individual and sex differences in neurobiological mechanisms related to BD, the use of outbred rat strains would be valuable. Here, we developed a novel BD model where after 3+ months of intermittent access to 20% alcohol Wistar rats drank, twice a week, with two 5-min intake (what we called Two-shot) separated by a 10-min break. Our findings showed during Two-Shot that most animals reached ≥ 80 mg% BAC levels (when briefly food-restricted). However, when increasing alcohol concentrations from 20 to 30%, 40%, or 50%, rats titrated to similar intake levels, suggesting rapid sensing of alcohol effects even when front-loading. Two-Shot drinking was reduced in both sexes by naltrexone (1 mg/kg), validating intake suppression by a clinical therapeutic agent for human problem drinking. Further, both propranolol (ß-adrenergic receptor antagonist) and prazosin (α1-adrenergic receptor antagonist) reduced female but not male BD at the lower dose. Thus, our results provide a novel model for BD in outbred rats and suggest that female binging is more sensitive to adrenergic modulation than males, perhaps providing a novel sex-related therapy.


Subject(s)
Binge Drinking , Disease Models, Animal , Rats, Wistar , Animals , Female , Binge Drinking/drug therapy , Male , Rats , Ethanol , Adrenergic Antagonists/pharmacology , Naltrexone/pharmacology , Propranolol/pharmacology , Sex Factors , Alcohol Drinking
8.
Transl Psychiatry ; 14(1): 90, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38346984

ABSTRACT

Recent studies have implicated the endogenous opioid system in the antidepressant actions of ketamine, but the underlying mechanisms remain unclear. We used a combination of pharmacological, behavioral, and molecular approaches in rats to test the contribution of the prefrontal endogenous opioid system to the antidepressant-like effects of a single dose of ketamine. Both the behavioral actions of ketamine and their molecular correlates in the medial prefrontal cortex (mPFC) are blocked by acute systemic administration of naltrexone, a competitive opioid receptor antagonist. Naltrexone delivered directly into the mPFC similarly disrupts the behavioral effects of ketamine. Ketamine treatment rapidly increases levels of ß-endorphin and the expression of the µ-opioid receptor gene (Oprm1) in the mPFC, and the expression of gene that encodes proopiomelanocortin, the precursor of ß-endorphin, in the hypothalamus, in vivo. Finally, neutralization of ß-endorphin in the mPFC using a specific antibody prior to ketamine treatment abolishes both behavioral and molecular effects. Together, these findings indicate that presence of ß-endorphin and activation of opioid receptors in the mPFC are required for the antidepressant-like actions of ketamine.


Subject(s)
Ketamine , Rats , Animals , Analgesics, Opioid/pharmacology , beta-Endorphin/metabolism , beta-Endorphin/pharmacology , Naltrexone/pharmacology , Naltrexone/metabolism , Antidepressive Agents , Prefrontal Cortex/metabolism
9.
Talanta ; 272: 125827, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38432124

ABSTRACT

Triple negative breast cancer (TNBC) is a very aggressive form of breast cancer, and the analgesic drug morphine has been shown to promote the proliferation of TNBC cells. This article investigates whether morphine causes activation of epidermal growth factor receptors (EGFR), the roles of µ-opioid and EGFR receptors on TNBC cell proliferation and migration. While examining the changes with molecular techniques, we also aimed to investigate the analysis ability of Raman spectroscopy and machine learning-based approach. Effects of morphine on the proliferation and migration of MDA.MB.231 cells were evaluated by MTT and scratch wound-healing tests, respectively. Morphine-induced phosphorylation of the EGFR was analyzed by western blotting in the presence and absence of µ-receptor antagonist naltrexone and the EGFR-tyrosine kinase inhibitor gefitinib. Morphine-induced EGFR phosphorylation and cell migration were significantly inhibited by pretreatments with both naltrexone and gefitinib; however, morphine-increased cell proliferation was inhibited only by naltrexone. While morphine-induced changes were observed in the Raman scatterings of the cells, the inhibitory effect of naltrexone was analyzed with similarity to the control group. Principal component analysis (PCA) of the Raman confirmed the epidermal growth factor (EGF)-like effect of morphine and was inhibited by naltrexone and partly by gefitinib pretreatments. Our in vitro results suggest that combining morphine with an EGFR inhibitor or a peripherally acting opioidergic receptor antagonist may be a good strategy for pain relief without triggering cancer proliferation and migration in TNBC patients. In addition, our results demonstrated the feasibility of the Raman spectroscopy and machine learning-based approach as an effective method to investigate the effects of agents in cancer cells without the need for complex and time-consuming sample preparation. The support vector machine (SVM) with linear kernel automatically classified the effects of drugs on cancer cells with ∼95% accuracy.


Subject(s)
ErbB Receptors , Triple Negative Breast Neoplasms , Humans , ErbB Receptors/metabolism , Triple Negative Breast Neoplasms/drug therapy , Gefitinib/pharmacology , Morphine/pharmacology , Spectrum Analysis, Raman , Naltrexone/pharmacology , Quinazolines/pharmacology , Cell Proliferation , EGF Family of Proteins/pharmacology , Cell Line, Tumor , Epidermal Growth Factor/pharmacology
10.
Drug Des Devel Ther ; 18: 1865-1874, 2024.
Article in English | MEDLINE | ID: mdl-38828019

ABSTRACT

Purpose: This study was designed to investigate the effects of preadministration of nalmefene before general anesthesia induction on sufentanil-induced cough (SIC) in patients undergoing breast surgery. Patients and Methods: A total of 105 patients scheduled for elective breast surgery under general anesthesia were selected and randomly assigned into three groups: normal saline (Group C), low-dose nalmefene 0.1 µg·kg-1 (Group LN), and high-dose nalmefene 0.25 µg·kg-1 (Group HN). Sufentanil 0.5 µg·kg-1 was injected intravenously within 2 s after 5 min of intervention. The count and severity of cough within 2 min after sufentanil injection, as well as the time to first cough, were recorded. In addition, we also collected intraoperative hemodynamic data, postoperative pain scores, the incidence of receiving rescue analgesics, and side effects up to 24 h after surgery. Results: Compared to Group C, the incidence of SIC was significantly lower in Group LN and HN (64.7% vs 30.3% and 14.7%, respectively; P < 0.001), but no significant difference was observed between the two groups (P=0.126). Compared to Group C, the risk factors decreased by 53.4% (95% confidence interval [CI] =0.181-0.735, P=0.008) in Group LN and by 75.9% (95% CI=0.432-0.898, P=0.001) in Group HN. Of the patients with SIC, less frequent SIC within 2 min after induction and a lower proportion of severe coughs were observed than Group C (P < 0.05), and no difference was detected between Group LN and HN. Additionally, the onset time to the first SIC did not differ significantly between the groups. Intraoperative hemodynamic data, postoperative pain scores, and side effects in the first 24 h did not differ among the groups. Conclusion: Preadministration of nalmefene prior to induction of general anesthesia effectively suppressed SIC in patients undergoing breast surgery, without affecting intraoperative hemodynamic fluctuation and postoperative pain intensity.


Subject(s)
Anesthesia, General , Cough , Naltrexone , Sufentanil , Humans , Sufentanil/administration & dosage , Sufentanil/adverse effects , Anesthesia, General/adverse effects , Cough/drug therapy , Cough/chemically induced , Female , Naltrexone/analogs & derivatives , Naltrexone/administration & dosage , Naltrexone/pharmacology , Double-Blind Method , Middle Aged , Adult , Breast/surgery , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Dose-Response Relationship, Drug
11.
Free Radic Biol Med ; 212: 94-116, 2024 02 20.
Article in English | MEDLINE | ID: mdl-38142953

ABSTRACT

Opioids regulate various physiological and pathophysiological functions, including cell proliferation, immune function, obesity, and neurodegenerative disorders. They have been used for centuries as a treatment for severe pain, binding to opioid receptors a specific G protein-coupled receptor. Common opioids, like ß-endorphin, [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO), and dynorphins, have analgesic effects. The use of a potent antagonist, like naltrexone hydrochloride, to block the effects of mu Opioid Receptor (µOR) may result in the withdrawal of physiological effects and could potentially impact immune responses in many diseases including respiratory disease. Asthma is a respiratory disease characterized by airway hyperresponsiveness, inflammation, bronchoconstriction, chest tightness, stress generation and release of various cytokines. Airway inflammation leads recruitment and activation of immune cells releasing mediators, including opioids, which may modulate inflammatory response by binding to their respective receptors. The study aims to explore the role of µOR antagonist (naltrexone) in regulating asthma pathophysiology, as the regulation of immune and inflammatory responses in asthma remains unclear. Balb/c mice were sensitized intranasally by 1% TDI and challenged with 2.5% TDI. Naltrexone hydrochloride (1 mg/kg body weight) was administered through intraperitoneal route 1 h before TDI induction. Blocking µOR by naltrexone exacerbates airway inflammation by recruiting inflammatory cells (lymphocytes and neutrophils), enhancing intracellular Reactive oxygen species in bronchoalveolar lavage fluid (BALF), and inflammatory mediator (histamine, Eosinophil peroxidase and neutrophil elastase) in lungs. Naltrexone administration modulated inflammatory cytokines (TNF-α, IL-4, IL-5, IL-6, IL-10, and IL-17A), and enhanced IgE and CRP levels. Naltrexone administration also increased the expression of NF-κB, and phosphorylated p-P38, p-Erk, p-JNK and NF-κB by inhibiting the µOR. Docking study revealed good binding affinity of naltrexone with µOR compared to δ and κ receptors. In future it might elucidate potential therapeutic against many respiratory pathological disorders. In conclusion, µOR blocking by naltrexone regulates and implicates inflammation, bronchoconstriction, and lung physiology.


Subject(s)
Asthma , Naltrexone , Animals , Mice , Naltrexone/pharmacology , Naltrexone/therapeutic use , NF-kappa B/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/therapeutic use , Disease Models, Animal , Asthma/drug therapy , Inflammation/pathology , Lung/metabolism , Cytokines/metabolism , Oxidative Stress , Mice, Inbred BALB C , Ovalbumin
12.
Pharmacol Rep ; 76(1): 112-126, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38236555

ABSTRACT

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.


Subject(s)
Colitis-Associated Neoplasms , Colitis , Colorectal Neoplasms , Mice , Animals , Interleukin-18 , Analgesics, Opioid/adverse effects , Interleukin-6 , Adipokines , Naltrexone/pharmacology , Adiponectin/adverse effects , Cyclooxygenase 2 , Carcinogenesis , Azoxymethane/toxicity , Disease Models, Animal , Receptors, Opioid/genetics , RNA, Messenger , Dextran Sulfate , Colorectal Neoplasms/genetics , Mice, Inbred C57BL , Colitis/chemically induced
13.
J Clin Pharmacol ; 64(7): 828-839, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38436495

ABSTRACT

An open-label, randomized, crossover study in healthy volunteers compared the reversal of remifentanil-induced respiratory depression by intranasal (IN) naloxone hydrochloride (4 mg) to IN nalmefene (2.7 mg) (NCT04828005). Subjects were administered a hypercapnic gas mixture which produces an elevation in minute ventilation (MV), a result of the ventilatory response to hypercapnia. Subjects breathed a hypercapnic gas mixture through a tight-fitting mask for an initial period of 46 min prior to a series of mask "holidays" introduced to reduce subject discomfort and encourage study completion. Ten minutes after initiating the hypercapnic gas mixture, a remifentanil bolus was administered, and an infusion continued for the study duration. Subjects were administered either naloxone or nalmefene 15 min after initiating the remifentanil infusion and MV monitored for 21 min followed by a mask holiday. Both nalmefene and naloxone produced a time-dependent reversal of remifentanil-induced reductions in MV measured 2.5-20 min post administration. At the primary endpoint (5 min post administration), nalmefene increases in MV (5.75 L/min) were nearly twice that produced by naloxone (3.01 L/min) (P < .0009); the point estimate favors nalmefene, demonstrating non-inferiority and superiority. In this model of opioid-induced respiratory depression, nalmefene has a more rapid onset of action than naloxone, which required 20 min to achieve a comparable reversal of respiratory depression. Both nalmefene and naloxone were well tolerated by healthy volunteers. This rapid onset of action may prove particularly valuable in an era when over 90% of fatalities are linked to synthetic opioid overdose.


Subject(s)
Administration, Intranasal , Analgesics, Opioid , Cross-Over Studies , Healthy Volunteers , Naloxone , Naltrexone , Narcotic Antagonists , Remifentanil , Respiratory Insufficiency , Humans , Naloxone/administration & dosage , Naloxone/pharmacology , Male , Adult , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Naltrexone/administration & dosage , Respiratory Insufficiency/chemically induced , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Female , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Remifentanil/administration & dosage , Remifentanil/pharmacology , Young Adult , Middle Aged
14.
Psychopharmacol Bull ; 54(3): 97-99, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38993658

ABSTRACT

NTX is FDA-approved for opiate and alcohol use disorders as anti-craving agent. It has been used successfully off-label in other psychiatric indications. Here, we shed some light on these while examining the extant evidence.


Subject(s)
Naltrexone , Narcotic Antagonists , Animals , Humans , Alcoholism/drug therapy , Craving/drug effects , Naltrexone/administration & dosage , Naltrexone/pharmacology , Naltrexone/therapeutic use , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Off-Label Use , Opioid-Related Disorders/drug therapy , Psychopharmacology
15.
Exp Biol Med (Maywood) ; 249: 10175, 2024.
Article in English | MEDLINE | ID: mdl-38756167

ABSTRACT

Diabetes mellitus is a prevalent disease that is often accompanied by ocular surface abnormalities including delayed epithelial wound healing and decreased corneal sensitivity. The impact of diabetes on the lacrimal functional unit (LFU) and the structures responsible for maintaining tear homeostasis, is not completely known. It has been shown that the Opioid Growth Factor Receptor (OGFr), and its ligand, Opioid Growth Factor (OGF), is dysregulated in the ocular surface of diabetic rats leading to overproduction of the inhibitory growth peptide OGF. The opioid antagonist naltrexone hydrochloride (NTX) blocks the OGF-OGFr pathway, and complete blockade following systemic or topical treatment with NTX restores the rate of re-epithelialization of corneal epithelial wounds, normalizes corneal sensitivity, and reverses dry eye in diabetic animal models. These effects occur rapidly and within days of initiating treatment. The present study was designed to understand mechanisms related to the fast reversal (<5 days) of dry eye by NTX in type 1 diabetes (T1D) by investigating dysregulation of the LFU. The approach involved examination of the morphology of the LFU before and after NTX treatment. Male and female adult Sprague-Dawley rats were rendered hyperglycemic with streptozotocin, and after 6 weeks rats were considered to be a T1D model. Rats received topical NTX twice daily to one eye for 10 days. During the period of treatment, tear production and corneal sensitivity were recorded. On day 11, animals were euthanized and orbital tissues including conjunctiva, eyelids, and lacrimal glands, were removed and processed for histologic examination including immunohistochemistry. Male and female T1D rats had significantly decreased tear production and corneal insensitivity, significantly decreased number and size of lacrimal gland acini, decreased expression of aquaporin-5 (AQP5) protein and decreased goblet cell size. Thus, 10 days of NTX treatment restored tear production and corneal sensitivity to normal values, increased AQP5 expression, and restored the surface area of goblet cells to normal. NTX had no effect on the number of lacrimal gland acini or the number of conjunctival goblet cells. In summary, blockade of the OGF-OGFr pathway with NTX reversed corneal and lacrimal gland complications and restored some components of tear homeostasis confirming the efficacy of topical NTX as a treatment for ocular defects in diabetes.


Subject(s)
Aquaporin 5 , Diabetes Mellitus, Experimental , Lacrimal Apparatus , Naltrexone , Tears , Animals , Male , Rats , Administration, Topical , Aquaporin 5/metabolism , Diabetes Mellitus, Experimental/complications , Dry Eye Syndromes/drug therapy , Dry Eye Syndromes/pathology , Dry Eye Syndromes/metabolism , Lacrimal Apparatus/metabolism , Lacrimal Apparatus/drug effects , Lacrimal Apparatus/pathology , Naltrexone/pharmacology , Rats, Sprague-Dawley , Tears/metabolism , Tears/drug effects
16.
Basic Clin Pharmacol Toxicol ; 134(6): 792-804, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38584299

ABSTRACT

Understanding the function of the kappa opioid receptor (KOP) is crucial for the development of novel therapeutic interventions that target KOP for the treatment of pain, stress-related disorders and other indications. Activation of KOP produces diuretic effects in rodents and man. Sex is a vital factor to consider when assessing drug response in pre-clinical and clinical studies. In this study, the diuretic effect of the KOP agonist, U50488 (1-10 mg/kg), was investigated in both adult female and male Wistar rats that were either normally hydrated or water-loaded. The KOP antagonist norbinaltorphimine (norBNI, 10 mg/kg) was administered 24 h prior to U50488 to confirm the involvement of KOP. U50488 elicited a significant diuretic response at doses ≥ 3 mg/kg in both female and male rats independent of hydration status. U50488 diuretic effects were inhibited by norBNI pre-administration. Water-loading reduced data variability for urine volume in males, but not in females, compared with normally hydrated rats. Sex differences were also evident in U50488 eliciting a significant increase in sodium and potassium ion excretion only in males. This may suggest different mechanisms of U50488 diuretic action in males where renal excretion mechanisms are directly affected more than in females.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer , Diuresis , Rats, Wistar , Receptors, Opioid, kappa , Animals , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Male , Female , Diuresis/drug effects , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Rats , Sex Factors , Diuretics/pharmacology , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Sodium/urine , Sodium/metabolism , Organism Hydration Status/drug effects , Potassium/urine , Potassium/metabolism , Dose-Response Relationship, Drug , Narcotic Antagonists/pharmacology
17.
Pharmacol Biochem Behav ; 242: 173822, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996927

ABSTRACT

The volatile compound 2,4,5-trimethylthiazoline (TMT, a synthetic predator scent) triggers fear, anxiety, and defensive responses in rodents that can outlast the encounter. The receptor systems underlying the development and persistence of TMT-induced behavioral changes remain poorly characterized, especially in females. Kappa opioid receptors regulate threat generalization and fear conditioning and alter basal anxiety, but their role in unconditioned fear responses in females has not been examined. Here, we investigated the effects of the long-lasting kappa opioid receptor antagonist, nor-binalthorphinmine dihydrochloride (nor-BNI; 10 mg/kg), on TMT-induced freezing and conditioned place aversion in female mice. We also measured anxiety-like behavior in the elevated plus maze three days after TMT and freezing behavior when returned to the TMT-paired context ten days after the single exposure. We found that 35µl of 10 % TMT elicited a robust freezing response during a five-minute exposure in female mice. TMT evoked persistent fear as measured by conditioned place aversion, reduced entries into the open arm of the elevated plus maze, and increased general freezing behavior long after TMT exposure. In line with the known role of kappa-opioid receptors in threat generalization, we found that kappa-opioid receptor antagonism increased basal freezing but reduced freezing during TMT presentation. Together, these findings indicate that a single exposure to TMT causes long-lasting changes in fear-related behavioral responses in female mice and highlights the modulatory role of kappa-opioid receptor signaling on fear-related behavioral patterns in females.


Subject(s)
Behavior, Animal , Fear , Odorants , Receptors, Opioid, kappa , Thiazoles , Animals , Female , Receptors, Opioid, kappa/metabolism , Mice , Thiazoles/pharmacology , Fear/drug effects , Behavior, Animal/drug effects , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Signal Transduction/drug effects , Anxiety/psychology , Anxiety/metabolism , Mice, Inbred C57BL , Narcotic Antagonists/pharmacology
18.
Neuropsychopharmacology ; 49(10): 1550-1558, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38528134

ABSTRACT

Although the kappa-opioid receptor (KOR) and its endogenous ligand, dynorphin, are believed to be involved in ethanol drinking, evidence on the direction of their effects has been mixed. The nucleus accumbens (NAc) shell densely expresses KORs, but previous studies have not found KOR activation to influence ethanol drinking. Using microinjections into the NAc shell of male and female Long-Evans rats that drank under the intermittent-access procedure, we found that the KOR agonist, U50,488, had no effect on ethanol drinking when injected into the middle NAc shell, but that it promoted intake in males and high-drinking females in the caudal NAc shell and high-drinking females in the rostral shell, and decreased intake in males and low-drinking females in the rostral shell. Conversely, injection of the KOR antagonist, nor-binaltorphimine, stimulated ethanol drinking in low-drinking females when injected into the rostral NAc shell and decreased drinking in high-drinking females when injected into the caudal NAc shell. These effects of KOR activity were substance-specific, as U50,488 did not affect sucrose intake. Using quantitative real-time PCR, we found that baseline gene expression of the KOR was higher in the rostral compared to caudal NAc shell, but that this was upregulated in the rostral shell with a history of ethanol drinking. Our findings have important clinical implications, demonstrating that KOR stimulation in the NAc shell can affect ethanol drinking, but that this depends on NAc subregion, subject sex, and ethanol intake level, and suggesting that this may be due to differences in KOR expression.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer , Alcohol Drinking , Ethanol , Naltrexone , Nucleus Accumbens , Rats, Long-Evans , Receptors, Opioid, kappa , Animals , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/agonists , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Male , Female , Rats , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Alcohol Drinking/metabolism , Ethanol/pharmacology , Ethanol/administration & dosage , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacology , Microinjections/methods , Central Nervous System Depressants/pharmacology , Central Nervous System Depressants/administration & dosage
19.
Neuropharmacology ; 258: 110093, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39067665

ABSTRACT

Agonists at µ opioid receptors relieve acute pain, however, their long-term use is limited by side effects, which may involve ß-arrestin2. Agonists biased against ß-arrestin2 recruitment may be advantageous. However, the classification of bias may be compromised by assays utilising overexpressed µ receptors which overestimate efficacy for G-protein activation. There is a need for re-evaluation with restricted receptor availability to determine accurate agonist efficacies. We depleted µ receptor availability in PathHunter CHO cells using the irreversible antagonist, ß-funaltrexamine (ß-FNA), and compared efficacies and apparent potencies of twelve agonists, including several previously reported as biased, in ß-arrestin2 recruitment and cAMP assays. With full receptor availability all agonists had partial efficacy for stimulating ß-arrestin2 recruitment relative to DAMGO, while only TRV130 and buprenorphine were partial agonists as inhibitors of cAMP accumulation. Limiting receptor availability by prior exposure to ß-FNA (100 nM) revealed morphine, oxycodone, PZM21, herkinorin, U47700, tianeptine and U47931e are also partial agonists in the cAMP assay. The efficacies of all agonists, except SR-17018, correlated between ß-arrestin2 recruitment and cAMP assays, with depleted receptor availability in the latter. Furthermore, naloxone and cyprodime exhibited non-competitive antagonism of SR-17018 in the ß-arrestin2 recruitment assay. Limited antagonism by naloxone was also non-competitive in the cAMP assay, while cyprodime was competitive. Furthermore, SR-17018 only negligibly diminished ß-arrestin2 recruitment stimulated by DAMGO (1 µM), whereas fentanyl, morphine and TRV130 all exhibited the anticipated competitive inhibition. The data suggest that SR-17018 achieves bias against ß-arrestin2 recruitment through interactions with µ receptors outside the orthosteric agonist site. This article is part of the Special Issue on "Ligand Bias".


Subject(s)
Analgesics, Opioid , Cricetulus , Cyclic AMP , Receptors, Opioid, mu , Animals , CHO Cells , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/agonists , Analgesics, Opioid/pharmacology , Cyclic AMP/metabolism , Narcotic Antagonists/pharmacology , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Cricetinae , Humans , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , beta-Arrestins/metabolism , Dose-Response Relationship, Drug , beta-Arrestin 2/metabolism , Spiro Compounds , Thiophenes
20.
Drug Alcohol Depend ; 255: 110967, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38150894

ABSTRACT

BACKGROUND: Opioid withdrawal symptoms (OWS) are highly aversive and prompt unprescribed opioid use, which increases morbidity, mortality, and, among individuals being treated for opioid use disorder (OUD), recurrence. OWS are driven by sympathetic nervous system (SNS) hyperactivity that occurs when blood opioid levels wane. We tested whether brief inhalation of xenon gas, which inhibits SNS activity and is used clinically for anesthesia and diagnostic imaging, attenuates naltrexone-precipitated withdrawal-like signs in morphine-dependent mice. METHODS: Adult CD-1 mice were implanted with morphine sulfate-loaded (60 mg/ml) minipumps and maintained for 6 days to establish morphine dependence. On day 7, mice were given subcutaneous naltrexone (0.3 mg/kg) and placed in a sealed exposure chamber containing either 21% oxygen/balance nitrogen (controls) or 21% oxygen/added xenon peaking at 30%/balance nitrogen. After 10 minutes, mice were transferred to observation chambers and videorecorded for 45 minutes. Videos were scored in a blind manner for morphine withdrawal behaviors. Data were analyzed using 2-way ANOVAs testing for treatment and sex effects. RESULTS AND CONCLUSIONS: Xenon-exposed mice exhibited fewer jumps (P = 0.010) and jumping suppression was detectible within the first 10-minute video segment, but no sex differences were detected. Brief inhalation of low concentration xenon rapidly and substantially attenuated naltrexone-precipitated jumping in morphine-dependent mice, suggesting that it can inhibit OWS. If xenon effects translate to humans with OUD, xenon inhalation may be effective for reducing OWS, unprescribed opioid use, and for easing OUD treatment initiation, which could help lower excess morbidity and mortality associated with OUD.


Subject(s)
Morphine Dependence , Opioid-Related Disorders , Substance Withdrawal Syndrome , Humans , Adult , Mice , Animals , Naltrexone/pharmacology , Naltrexone/therapeutic use , Analgesics, Opioid/therapeutic use , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Morphine/pharmacology , Morphine/therapeutic use , Narcotics/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Opioid-Related Disorders/drug therapy , Nitrogen/therapeutic use , Oxygen/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL