Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 381
Filter
1.
J Toxicol Sci ; 49(10): 435-446, 2024.
Article in English | MEDLINE | ID: mdl-39358233

ABSTRACT

BACKGROUND: Neuroinflammation plays a critical role in various neurological disorders. Oxycodone has anti-inflammatory properties. The purpose of this work was to look into the effect of oxycodone in controlling lipopolysaccharide (LPS)-induced neuroinflammation in microglia. METHODS: LPS-induced HMC3 cells were subjected to oxycodone (2.5, 5, 10 and 20 µg/mL). The mRNA and protein expressions were examined by qRT-PCR and western blotting. TNF-α, IL-1ß, IL-6, and IL-8 levels were assessed by ELISA. MTT assay was adopted to measure cell viability. The interactions between CREB, miR-181c and PDCD4 were analyzed by dual-luciferase reporter assay, ChIP and/or RIP assays. RESULTS: Oxycodone treatment alleviated LPS-induced inflammation in HMC3 cells and increased p-CREB level, but reduced PDCD4 and iNOS levels in LPS-treated cells. Mechanistically, oxycodone mitigated LPS-induced neuroinflammation by upregulating miR-181c. In addition, CREB promoted miR-181c expression by directly binding to the MIR181C promoter, and miR-181c inhibited PDCD4 expression by directly binding to PDCD4 3'UTR. As expected, oxycodone alleviated LPS-induced neuroinflammation by regulating the CREB/miR-181c/PDCD4 axis. CONCLUSION: Oxycodone attenuated LPS-induced neuroinflammation in microglia by regulating the CREB/miR-181c/PDCD4 axis. These findings proved that oxycodone is a potential drug for treating neuroinflammation and elucidate the mechanisms involved.


Subject(s)
Apoptosis Regulatory Proteins , Cyclic AMP Response Element-Binding Protein , Lipopolysaccharides , MicroRNAs , Microglia , Neuroinflammatory Diseases , Oxycodone , RNA-Binding Proteins , MicroRNAs/genetics , MicroRNAs/metabolism , Oxycodone/pharmacology , Oxycodone/adverse effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Humans , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Microglia/drug effects , Microglia/metabolism , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/genetics , Anti-Inflammatory Agents/pharmacology , Cell Line , Inflammation/chemically induced , Inflammation/genetics , Signal Transduction/drug effects
2.
Addict Biol ; 29(8): e13430, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39121884

ABSTRACT

Approximately 50 million Americans suffer from chronic pain, and nearly a quarter of chronic pain patients have reported misusing opioid prescriptions. Repeated drug seeking is associated with reactivation of an ensemble of neurons sparsely scattered throughout the dorsomedial prefrontal cortex (dmPFC). Prior research has demonstrated that chronic pain increases intrinsic excitability of dmPFC neurons, which may increase the likelihood of reactivation during drug seeking. We tested the hypothesis that chronic pain would increase oxycodone-seeking behaviour and that the pain state would differentially increase intrinsic excitability in dmPFC drug-seeking ensemble neurons. TetTag mice self-administered intravenous oxycodone. After 7 days of forced abstinence, a drug-seeking session was performed, and the ensemble was tagged. Mice received spared nerve injury (SNI) to induce chronic pain during the period between the first and second seeking session. Following the second seeking session, we performed electrophysiology on individual neurons within the dmPFC to assess intrinsic excitability of the drug-seeking ensemble and non-ensemble neurons. SNI had no impact on sucrose seeking or intrinsic excitability of dmPFC neurons from these mice. In females, SNI increased oxycodone seeking and intrinsic excitability of non-ensemble neurons. In males, SNI had no impact on oxycodone seeking or neuron excitability. Data from females are consistent with clinical reports that chronic pain can promote drug craving and relapse and support the hypothesis that chronic pain itself may lead to neuroadaptations which promote opioid seeking.


Subject(s)
Analgesics, Opioid , Drug-Seeking Behavior , Neuralgia , Neurons , Oxycodone , Prefrontal Cortex , Animals , Oxycodone/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Drug-Seeking Behavior/drug effects , Mice , Neuralgia/physiopathology , Neurons/drug effects , Male , Female , Analgesics, Opioid/pharmacology , Self Administration , Chronic Pain/physiopathology , Sex Factors
3.
Clinics (Sao Paulo) ; 79: 100458, 2024.
Article in English | MEDLINE | ID: mdl-39084065

ABSTRACT

BACKGROUND: The influences of Oxycodone (OXY) combined with Paclitaxel (PTX) on breast cancer cells are unclear. The present study aimed to examine the effects of OXY combined with PTX on the proliferation, apoptosis, and migration of human breast cancer SKBR3 cells and the underlying mechanism. METHODS: The proliferation, apoptosis and invasion of SKBR3 cells were assessed by CCK-8, colony formation assay, flowcytometric, Transwell assay and scratch assays, respectively. In addition, Western blotting was used to detect the expression of related proteins in these cells. The autophagic bodies were observed under a transmission electron microscope. RESULTS: OXY (0.25, 0.5 and 1 mM) significantly inhibited the viability, colony-forming, migration, and invasion of SKBR3 cells as compared to the control group. Furthermore, OXY (0.25, 0.5 and 1 mM) markedly induced the apoptosis of SKBR3 cells and the levels of apoptosis-related proteins. In addition, OXY (0.25, 0.5 and 1 mM) and PTX inhibited the proliferation of SKBR3 cells synergistically as compared to PTX group in vitro. Moreover, OXY (0.25, 0.5 and 1 mM) significantly elevated the PTX-induced apoptosis in SKBR3 cells via downregulating the expression of N-cadherin, Becline-1 LC3-Ⅱ, p-Akt and p-mTOR and upregulating E-cadherin expression. Compared with the control group, OXY (1 mM) treatment induced autophagy in SKBR3 cells. CONCLUSIONS: The present study indicates that OXY can enhance the antitumor effect of PTX on breast cancer in vitro. Hence, the combination of OXY with PTX may serve as a potential strategy for the treatment of breast cancer.


Subject(s)
Apoptosis , Breast Neoplasms , Cell Movement , Cell Proliferation , Oxycodone , Paclitaxel , Humans , Paclitaxel/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Cell Line, Tumor , Oxycodone/pharmacology , Cell Movement/drug effects , Drug Synergism , Cell Survival/drug effects , Autophagy/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Reproducibility of Results , Blotting, Western
4.
Clin Exp Pharmacol Physiol ; 51(9): e13910, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39073215

ABSTRACT

Myocardial injury and cardiovascular dysfunction are the most common complications of sepsis, and effective therapeutic candidate is still lacking. This study aims to investigate the protective effect of oxycodone in myocardial injury of lipopolysaccharide-induced sepsis and its related signalling pathways. Wild-type and nuclear factor erythroid 2-related factor 2 (Nrf2)-knockout mice, as well as H9c2 cardiomyocytes cultures treated with lipopolysaccharide (LPS) were used as models of septic myocardial injury. H9c2 cardiomyocytes culture showed that oxycodone protected cells from pyroptosis induced by LPS. Mice model confirmed that oxycodone pretreatment significantly attenuated myocardial pathological damage and improved cardiac function demonstrated by increased ejection fraction (EF) and fractional shortening (FS), as well as decreased cardiac troponin I (cTnI) and creatine kinase isoenzymes MB (CK-MB). Oxycodone also reduced the levels of inflammatory factors and oxidative stress damage induced by LPS, which involves pyroptosis-related proteins including: Nod-like receptor protein 3 (NLRP3), Caspase 1, Apoptosis-associated speck-like protein contain a CARD (ASC), and Gasdermin D (GSDMD). These changes were mediated by Nrf2 and heme oxygenase-1 (HO-1) because Nrf2-knockout mice or Nrf2 knockdown in H9c2 cells significantly reversed the beneficial effect of oxycodone on oxidative stress, inflammatory responses and NLRP3-mediated pyroptosis. Our findings yielded that oxycodone therapy reduces LPS-induced myocardial injury by suppressing NLRP3-mediated pyroptosis via the Nrf2/HO-1 signalling pathway in vivo and in vitro.


Subject(s)
Heme Oxygenase-1 , Inflammation , Lipopolysaccharides , Myocytes, Cardiac , NF-E2-Related Factor 2 , Oxycodone , Pyroptosis , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Pyroptosis/drug effects , Lipopolysaccharides/toxicity , Signal Transduction/drug effects , Mice , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Heme Oxygenase-1/metabolism , Oxycodone/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Male , Cell Line , Rats , Oxidation-Reduction/drug effects , Mice, Knockout , Mice, Inbred C57BL , Oxidative Stress/drug effects
5.
CNS Neurosci Ther ; 30(7): e14830, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39046182

ABSTRACT

N6-methyladenosine (m6A) methylation is a vital epigenetic mechanism associated with drug addiction. However, the relationship between m6A modification and oxycodone rewarding is less well explored. Based on an open field test, the present study evaluated oxycodone rewarding using chromatin immunoprecipitation PCR, immunofluorescence, and RNA sequencing. A marked increase in METTL14 protein and a decrease in PP1α protein due to oxycodone abundance in the striatal neurons were observed in a dose- and time-dependent manner. Oxycodone markedly increased LSD1 expression, and decreased H3K4me1 expression in the striatum. In the open field test, intra-striatal injection of METTL14 siRNA, HOTAIR siRNA, or LSD1 shRNA blocked oxycodone-induced increase in locomotor activity. The downregulation of PP1α was also inhibited after treatment with METTL14/HOTAIR siRNA and LSD1 shRNA. Enhanced binding of LSD1 with CoRest and of CoRest with the PP1α gene induced by oxycodone was also reversed by LSD1 shRNA. In addition, H3K4me1 demethylation was also blocked by the treatment. In summary, the investigation confirmed that METTL14-mediated upregulation of HOTAIR resulted in the repression of PP1α, which in turn facilitated the recruitment of LSD1, thus catalyzing H3K4me1 demethylation and promoting oxycodone addiction.


Subject(s)
Methyltransferases , Oxycodone , RNA, Long Noncoding , Animals , Male , Mice , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Demethylation , Histone Demethylases/metabolism , Histone Demethylases/genetics , Histones/metabolism , Lysine/analogs & derivatives , Methyltransferases/metabolism , Methyltransferases/genetics , Mice, Inbred C57BL , Oxycodone/pharmacology , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/genetics , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Up-Regulation
6.
Behav Pharmacol ; 35(5): 269-279, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38847447

ABSTRACT

Excessive prescribing and misuse of prescription opioids, such as oxycodone, significantly contributed to the current opioid crisis. Although oxycodone is typically consumed orally by humans, parenteral routes of administration have primarily been used in preclinical models of oxycodone dependence. To address this issue, more recent studies have used oral self-administration procedures to study oxycodone seeking and withdrawal in rodents. Behavioral differences, however, following oral oxycodone intake versus parenteral oxycodone administration remain unclear. Thus, the goal of the current studies was to compare anxiety- and withdrawal-like behaviors using established opioid dependence models of either home cage oral intake of oxycodone (0.5 mg/ml) or repeated subcutaneous (s.c.) injections of oxycodone (10 mg/kg) in male and female mice. Here, mice received 10 days of oral or s.c. oxycodone administration, and following 72 h of forced abstinence, anxiety- and withdrawal-like behaviors were measured using elevated zero maze, open field, and naloxone-induced precipitated withdrawal procedures. Global withdrawal scores were increased to a similar degree following oral and s.c. oxycodone use, while both routes of oxycodone administration had minimal effects on anxiety-like behaviors. When examining individual withdrawal-like behaviors, mice receiving s.c. oxycodone exhibited more paw tremors and jumps during naloxone-induced precipitated withdrawal compared with oral oxycodone mice. These results indicate that both models of oxycodone administration are sufficient to elevate global withdrawal scores, but, when compared with oral consumption, s.c. oxycodone injections yielded more pronounced effects on some withdrawal-like behaviors.


Subject(s)
Analgesics, Opioid , Anxiety , Mice, Inbred C57BL , Oxycodone , Substance Withdrawal Syndrome , Animals , Oxycodone/pharmacology , Oxycodone/administration & dosage , Male , Female , Administration, Oral , Injections, Subcutaneous , Mice , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage , Opioid-Related Disorders , Naloxone/pharmacology , Naloxone/administration & dosage , Behavior, Animal/drug effects , Narcotic Antagonists/pharmacology , Narcotic Antagonists/administration & dosage
7.
Calcif Tissue Int ; 115(2): 174-184, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38856730

ABSTRACT

Patients with chronic kidney disease (CKD) report high pain levels, but reduced renal clearance eliminates many analgesic options; therefore, 30-50% of CKD patients have chronic opioid prescriptions. Opioid use in CKD is associated with higher fracture rates. Opioids may directly alter bone turnover directly through effects on bone cells and indirectly via increasing inflammation. We hypothesized that continuous opioid exposure would exacerbate the high bone turnover state of CKD and be associated with elevated measures of inflammation. Male C57Bl/6J mice after 8 weeks of adenine-induced CKD (AD) and non-AD controls (CON) had 14-day osmotic pumps (0.25-µL/hr release) containing either saline or 50-mg/mL oxycodone (OXY) surgically implanted in the subscapular region. After 2 weeks, all AD mice had elevated blood urea nitrogen, parathyroid hormone, and serum markers of bone turnover compared to controls with no effect of OXY. Immunohistochemical staining of the distal femur showed increased numbers of osteocytes positive for the mu opioid and for toll-like receptor 4 (TLR4) due to OXY. Osteocyte protein expression of tumor necrosis factor-α (TNF-α) and RANKL were higher due to both AD and OXY so that AD + OXY mice had the highest values. Trabecular osteoclast-covered surfaces were also significantly higher due to both AD and OXY, resulting in AD + OXY mice having 4.5-fold higher osteoclast-covered surfaces than untreated CON. These data demonstrate that opioids are associated with a pro-inflammatory state in osteocytes which increases the pro-resorptive state of CKD.


Subject(s)
Adenine , Analgesics, Opioid , Disease Models, Animal , Mice, Inbred C57BL , Osteoclasts , Renal Insufficiency, Chronic , Animals , Adenine/pharmacology , Adenine/adverse effects , Male , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Analgesics, Opioid/adverse effects , Mice , Inflammation , Bone Remodeling/drug effects , Oxycodone/pharmacology , Bone and Bones/metabolism , Bone and Bones/drug effects
8.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38928357

ABSTRACT

Cannabidiol (CBD), a phytocannabinoid, appeared to satisfy several criteria for a safe approach to preventing drug-taking behavior, including opioids. However, most successful preclinical and clinical results come from studies in adult males. We examined whether systemic injections of CBD (10 mg/kg, i.p.) during extinction of oxycodone (OXY, 3 mg/kg, i.p.) induced conditioned place preference (CPP) could attenuate the reinstatement of CPP brought about by OXY (1.5 mg/kg, i.p.) priming in adolescent rats of both sexes, and whether this effect is sex dependent. Accordingly, a priming dose of OXY produced reinstatement of the previously extinguished CPP in males and females. In both sexes, this effect was linked to locomotor sensitization that was blunted by CBD pretreatments. However, CBD was able to prevent the reinstatement of OXY-induced CPP only in adolescent males and this outcome was associated with an increased cannabinoid 1 receptor (CB1R) and a decreased mu opioid receptor (MOR) expression in the prefrontal cortex (PFC). The reinstatement of CCP in females was associated with a decreased MOR expression, but no changes were detected in CB1R in the hippocampus (HIP). Moreover, CBD administration during extinction significantly potentialized the reduced MOR expression in the PFC of males and showed a tendency to potentiate the reduced MOR in the HIP of females. Additionally, CBD reversed OXY-induced deficits of recognition memory only in males. These results suggest that CBD could reduce reinstatement to OXY seeking after a period of abstinence in adolescent male but not female rats. However, more investigation is required.


Subject(s)
Cannabidiol , Oxycodone , Receptor, Cannabinoid, CB1 , Receptors, Opioid, mu , Animals , Cannabidiol/pharmacology , Male , Female , Oxycodone/pharmacology , Rats , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid, mu/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Analgesics, Opioid/pharmacology , Conditioning, Psychological/drug effects
9.
Biomed Pharmacother ; 176: 116931, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38870630

ABSTRACT

The lysine-specific demethylase 1 (KDM1A) is reported to be a regulator in learning and memory. However, the effect of KDM1A in oxycodone rewarding memory has yet to be studied. In our study, rewarding memory was assessed by using conditioned place preference (CPP) in male mice. Next generation sequencing and chromatin immunoprecipitation-PCR were used to explore the molecular mechanisms. Oxycodone significantly decreased PP1α mRNA and protein levels in hippocampal neurons. Oxycodone significantly increased KDM1A and H3K4me1 levels, while significantly decreased H3K4me2 levels in a time- and dose-dependent manner. Behavioral data demonstrated that intraperitoneal injection of ORY-1001 (KDM1A inhibitor) or intra-hippocampal injection of KDM1A siRNA/shRNA blocked the acquisition and expression of oxycodone CPP and facilitated the extinction of oxycodone CPP. The decrease of PP1α was markedly blocked by the injection of ORY-1001 or KDM1A siRNA/shRNA. Oxycodone-induced enhanced binding of CoRest with KDM1A and binding of CoRest with the PP1α promoter was blocked by ORY-1001. The level of H3K4me2 demethylation was also decreased by the treatment. The results suggest that oxycodone-induced upregulation of KDM1A via demethylation of H3K4me2 promotes the binding of CoRest with the PP1α promoter, and the subsequent decrease in PP1α expression in hippocampal neurons may contribute to oxycodone reward.


Subject(s)
Epigenesis, Genetic , Histone Demethylases , Oxycodone , Animals , Male , Epigenesis, Genetic/drug effects , Mice , Oxycodone/pharmacology , Histone Demethylases/metabolism , Histone Demethylases/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Reward , Conditioning, Psychological/drug effects , Mice, Inbred C57BL , Histones/metabolism , Neurons/drug effects , Neurons/metabolism , Memory/drug effects
10.
Science ; 384(6700): eadn0886, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843332

ABSTRACT

In addition to their intrinsic rewarding properties, opioids can also evoke aversive reactions that protect against misuse. Cellular mechanisms that govern the interplay between opioid reward and aversion are poorly understood. We used whole-brain activity mapping in mice to show that neurons in the dorsal peduncular nucleus (DPn) are highly responsive to the opioid oxycodone. Connectomic profiling revealed that DPn neurons innervate the parabrachial nucleus (PBn). Spatial and single-nuclei transcriptomics resolved a population of PBn-projecting pyramidal neurons in the DPn that express µ-opioid receptors (µORs). Disrupting µOR signaling in the DPn switched oxycodone from rewarding to aversive and exacerbated the severity of opioid withdrawal. These findings identify the DPn as a key substrate for the abuse liability of opioids.


Subject(s)
Analgesics, Opioid , Avoidance Learning , Opioid-Related Disorders , Oxycodone , Parabrachial Nucleus , Prefrontal Cortex , Receptors, Opioid, mu , Reward , Animals , Male , Mice , Analgesics, Opioid/pharmacology , Connectome , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Opioid-Related Disorders/metabolism , Oxycodone/pharmacology , Parabrachial Nucleus/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Pyramidal Cells/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/genetics , Substance Withdrawal Syndrome/metabolism , Transcriptome
11.
Pharmacol Biochem Behav ; 242: 173795, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38834159

ABSTRACT

Opioid use disorder (OUD) is a serious health problem that may lead to physical dependence, in addition to affective disorders. Preclinical models are essential for studying the neurobiology of and developing pharmacotherapies to treat these problems. Historically, chronic morphine injections have most often been used to produce opioid-dependent animals, and withdrawal signs indicative of dependence were precipitated by administering an opioid antagonist. In the present studies, we have developed and validated a model of dependence on oxycodone (a widely prescribed opioid) during spontaneous withdrawal in male and female C57BL/6J mice. Dependence was induced by chronically administering oxycodone through osmotic minipumps at different doses for 7 days. Somatic withdrawal signs were measured after 3, 6, 24, and 48 h following minipump removal. Additionally, sensitivity to mechanical, thermal, and cold stimuli, along with anxiety-like behavior, were also measured. Our results indicated that spontaneous withdrawal following discontinuation of oxycodone produced an increase in total withdrawal signs after 60 and 120 mg/kg/day regimens of oxycodone administration. These signs were reversed by the administration of clinically approved medications for OUD. In general, both female and male mice showed similar profiles of somatic signs of spontaneous withdrawal. Spontaneous withdrawal also resulted in mechanical and cold hypersensitivity lasting for 24 and 14 days, respectively, and produced anxiety-like behaviors after 2 and 3 weeks following oxycodone removal. These results help validate a new model of oxycodone dependence, including the temporally distinct emergence of somatic, hyperalgesic, and anxiety-like behaviors, potentially useful for mechanistic and translational studies of opioid dependence.


Subject(s)
Analgesics, Opioid , Disease Models, Animal , Mice, Inbred C57BL , Oxycodone , Substance Withdrawal Syndrome , Animals , Oxycodone/administration & dosage , Oxycodone/pharmacology , Female , Male , Mice , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Analgesics, Opioid/adverse effects , Opioid-Related Disorders/drug therapy , Anxiety/chemically induced , Anxiety/drug therapy
12.
J Psychopharmacol ; 38(7): 647-660, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38888086

ABSTRACT

BACKGROUND: The orexin (OX) system has received increasing interest as a potential target for treating substance use disorder. OX transmission in the posterior paraventricular nucleus of the thalamus (pPVT), an area activated by highly salient stimuli that are both reinforcing and aversive, mediates cue- and stress-induced reinstatement of reward-seeking behavior. Oral administration of suvorexant (SUV), a dual OX receptor (OXR) antagonist (DORA), selectively reduced conditioned reinstatement of oxycodone-seeking behavior and stress-induced reinstatement of alcohol-seeking behavior in dependent rats. AIMS: This study tested whether OXR blockade in the pPVT with SUV reduces oxycodone or sweetened condensed milk (SCM) seeking elicited by conditioned cues or stress. METHODS: Male Wistar rats were trained to self-administer oxycodone (0.15 mg/kg, i.v., 8 h/day) or SCM (0.1 ml, 2:1 dilution [v/v], 30 min/day). After extinction, we tested the ability of intra-pPVT SUV (15 µg/0.5 µl) to prevent reinstatement of oxycodone or SCM seeking elicited by conditioned cues or footshock stress. RESULTS: The rats acquired oxycodone and SCM self-administration, and oxycodone intake correlated with signs of physical opioid withdrawal, confirming dependence. Following extinction, the presentation of conditioned cues or footshock elicited reinstatement of oxycodone- and SCM-seeking behavior. Intra-pPVT SUV blocked stress-induced reinstatement of oxycodone seeking but not conditioned reinstatement of oxycodone or SCM seeking or stress-induced reinstatement of SCM seeking. CONCLUSIONS: The results indicate that OXR signaling in the pPVT is critical for stress-induced reinstatement of oxycodone seeking, further corroborating OXRs as treatment targets for opioid use disorder.


Subject(s)
Drug-Seeking Behavior , Orexin Receptor Antagonists , Orexins , Oxycodone , Rats, Wistar , Self Administration , Stress, Psychological , Animals , Oxycodone/pharmacology , Oxycodone/administration & dosage , Male , Rats , Drug-Seeking Behavior/drug effects , Stress, Psychological/metabolism , Orexins/metabolism , Orexin Receptor Antagonists/pharmacology , Orexin Receptor Antagonists/administration & dosage , Cues , Signal Transduction/drug effects , Extinction, Psychological/drug effects , Orexin Receptors/metabolism , Orexin Receptors/drug effects , Reward , Opioid-Related Disorders/metabolism , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/metabolism
13.
Psychopharmacology (Berl) ; 241(7): 1477-1490, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38710856

ABSTRACT

RATIONALE: Medications are urgently needed to treat symptoms of drug withdrawal and mitigate dysphoria and psychiatric comorbidities that drive opioid abuse and relapse. ITI-333 is a novel molecule in development for treatment of substance use disorders, psychiatric comorbidities, and pain. OBJECTIVE: Characterize the preclinical profile of ITI-333 using pharmacological, behavioral, and physiological assays. METHODS: Cell-based assays were used to measure receptor binding and intrinsic efficacy of ITI-333; animal models were employed to assess effects on opioid reinstatement, precipitated oxycodone withdrawal, and drug abuse liability. RESULTS: In vitro, ITI-333 is a potent 5-HT2A receptor antagonist (Ki = 8 nM) and a biased, partial agonist at µ-opioid (MOP) receptors (Ki = 11 nM; lacking ß-arrestin agonism) with lesser antagonist activity at adrenergic α1A (Ki = 28 nM) and dopamine D1 (Ki = 50 nM) receptors. In vivo, ITI-333 blocks 5-HT2A receptor-mediated head twitch and MOP receptor-mediated effects on motor hyperactivity in mice. ITI-333 alone is a naloxone-sensitive analgesic (mice) which suppresses somatic signs of naloxone-precipitated oxycodone withdrawal (mice) and heroin cue-induced reinstatement responding without apparent tolerance or physical dependence after chronic dosing (rats). ITI-333 did not acutely impair gastrointestinal or pulmonary function (rats) and was not intravenously self-administered by heroin-maintained rats or rhesus monkeys. CONCLUSIONS: ITI-333 acts as a potent 5-HT2A receptor antagonist, as well a biased MOP receptor partial agonist with low intrinsic efficacy. ITI-333 mitigates opioid withdrawal/reinstatement, supporting its potential utility as a treatment for OUD.


Subject(s)
Substance Withdrawal Syndrome , Animals , Mice , Male , Substance Withdrawal Syndrome/drug therapy , Rats , Humans , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/administration & dosage , Substance-Related Disorders/drug therapy , Opioid-Related Disorders/drug therapy , Dose-Response Relationship, Drug , Oxycodone/pharmacology , Oxycodone/administration & dosage , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage , Self Administration , Cricetulus , CHO Cells
14.
Neuropharmacology ; 254: 109972, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38710443

ABSTRACT

Opioid use disorder (OUD) is a chronic condition associated with long-lasting molecular and behavioral changes. Animals with prolonged access to opioids develop behaviors similar to human OUD. Identifying associated molecular changes can provide insight to underpinnings that lead to or maintain OUD. In pilot studies, we identified several miRNA targets that are altered by the administration of oxycodone. We selected mir182 for follow up as it was recently shown to be dysregulated in plasma of men administered oxycodone. In addition, mir182 is increased in reward-related brain regions of male rats following exposure to various addictive substances. The present study utilizes a long-access oxycodone self-administration paradigm to examine changes in mir182 and its mRNA targets associated with neuroplasticity, which may be involved in the maintenance of OUD-like phenotype in rats. Male rats were trained to self-administer oxycodone (0.1 mg/kg/infusion, i. v.) for 6 h daily sessions for 12 days. Each animal had a yoked saline control that received matched saline infusions. Animals were then tested on a progressive ratio schedule to measure motivation to obtain a single infusion of oxycodone. Drug seeking was measured following 28 days of forced abstinence using a 90-min cued/test. RTqPCR was utilized to measure mir182 and mRNA targets related to neuroplasticity (wnt3, plppr4, pou3f3, tle4, cacna2d, and bdnf) from the nucleus accumbens. Data revealed that animals responded on a continuum for oxycodone. When divided into two groups termed high- and low responders, animals diverged during self-administration acquisition and maintained differences in behavior and gene expression throughout the study. mir182 was upregulated in the nucleus accumbens of both high and low responders and negatively correlated with tle4, which showed a strong negative correlation with reinstatement behavior. mRNA target levels were correlated with behaviors associated with increased severity of OUD behavior in male rats.


Subject(s)
MicroRNAs , Neuronal Plasticity , Oxycodone , Self Administration , Animals , Male , Oxycodone/administration & dosage , Oxycodone/pharmacology , Neuronal Plasticity/drug effects , Rats , MicroRNAs/metabolism , MicroRNAs/genetics , Individuality , Rats, Sprague-Dawley , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Opioid-Related Disorders/genetics , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics
15.
Psychopharmacology (Berl) ; 241(8): 1705-1717, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38676755

ABSTRACT

RATIONALE/OBJECTIVES: Targeting cannabinoid receptor type 1 (CB1R) has shown promise for treating opioid withdrawal symptoms. This study aimed to investigate the efficacy of a specific CB1R negative allosteric modulator (NAM), Org27569, in reducing both naloxone-precipitated and protracted withdrawal symptoms in oxycodone-dependent mice. METHODS: Mice received escalating doses of oxycodone (9-33 mg/kg IP) or saline twice daily for 9 days, followed by a final dose of oxycodone (33 mg/kg) or saline in the morning of day 9. In one cohort, the impact of Org27569 (3, 10, and 30 mg/kg) on naloxone (10 mg/kg IP) precipitated withdrawal symptoms was assessed. In another cohort, Org27569 (3 mg/kg) effects on the acquisition of conditioned place aversion to naloxone (0.6 mg/kg) precipitated opioid withdrawal, on behaviour following a 7-9-day abstinence period, and on naloxone (0.6 mg/kg) precipitated withdrawal-induced escape behaviour in a novel assay were assessed. RESULTS: Although Org27569 decreased opioid withdrawal-induced jumping at doses of 10 and 30 mg/kg, these effects were confounded by reduced locomotion. At all doses tested, Org27569 had a modest inhibitory effect on gastrointestinal motility. At the lower dose of 3 mg/kg, which was not confounded by locomotor effects, Org27569 did not impact naloxone-precipitated withdrawal-induced jumping, acquisition of oxycodone withdrawal-induced conditioned place aversion, or naloxone-precipitated withdrawal-induced escape behaviour in a novel assay. A clear protracted opioid withdrawal phenotype was not observed in assays of anxiety-like or social behaviour. CONCLUSIONS: Org27569 effects on negative affective-like symptoms were confounded by locomotor effects and effects on gastrointestinal motility were not opioid withdrawal specific. Further studies are needed in a model that produces a more pronounced protracted withdrawal syndrome.


Subject(s)
Dose-Response Relationship, Drug , Naloxone , Oxycodone , Receptor, Cannabinoid, CB1 , Substance Withdrawal Syndrome , Animals , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/drug therapy , Oxycodone/pharmacology , Oxycodone/administration & dosage , Mice , Male , Naloxone/pharmacology , Naloxone/administration & dosage , Receptor, Cannabinoid, CB1/metabolism , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage , Behavior, Animal/drug effects , Narcotic Antagonists/pharmacology , Narcotic Antagonists/administration & dosage , Allosteric Regulation/drug effects , Mice, Inbred C57BL , Indoles , Thiophenes
16.
Genes Brain Behav ; 23(2): e12894, 2024 04.
Article in English | MEDLINE | ID: mdl-38597363

ABSTRACT

Opioid use disorder (OUD) is an ongoing public health concern in the United States, and relatively little work has addressed how genetic background contributes to OUD. Understanding the genetic contributions to oxycodone-induced analgesia could provide insight into the early stages of OUD development. Here, we present findings from a behavioral phenotyping protocol using several inbred strains from the Hybrid Rat Diversity Panel. Our behavioral protocol included a modified "up-down" von Frey procedure to measure inherent strain differences in the sensitivity to a mechanical stimulus on the hindpaw. We also performed the tail immersion assay, which measures the latency to display tail withdrawal in response to a hot water bath. Initial withdrawal thresholds were taken in drug-naïve animals to record baseline thermal sensitivity across the strains. Oxycodone-induced analgesia was measured after administration of oxycodone over the course of 2 h. Both mechanical and thermal sensitivity are shaped by genetic factors and display moderate heritability (h2 = 0.23-0.40). All strains displayed oxycodone-induced analgesia that peaked at 15-30 min and returned to baseline by 2 h. There were significant differences between the strains in the magnitude and duration of their analgesic response to oxycodone, although the heritability estimates were quite modest (h2 = 0.10-0.15). These data demonstrate that genetic background confers differences in mechanical sensitivity, thermal sensitivity, and oxycodone-induced analgesia.


Subject(s)
Analgesia , Opioid-Related Disorders , Rats , Animals , Oxycodone/pharmacology , Analgesics, Opioid/pharmacology
17.
J Appl Physiol (1985) ; 136(5): 1097-1104, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38511209

ABSTRACT

When contracting muscles are freely perfused, the acid-sensing ion channel 3 (ASIC3) on group IV afferents plays a minor role in evoking the exercise pressor reflex. We recently showed in isolated dorsal root ganglion neurons innervating the gastrocnemius muscles that two mu opioid receptor agonists, namely endomorphin 2 and oxycodone, potentiated the sustained inward ASIC3 current evoked by acidic solutions. This in vitro finding prompted us to determine whether endomorphin 2 and oxycodone, when infused into the arterial supply of freely perfused contracting hindlimb muscles, potentiated the exercise pressor reflex. We found that infusion of endomorphin 2 and naloxone in decerebrated rats potentiated the pressor responses to contraction of the triceps surae muscles. The endomorphin 2-induced potentiation of the pressor responses to contraction was prevented by infusion of APETx2, an ASIC3 antagonist. Specifically, the peak pressor response to contraction averaged 19.3 ± 5.6 mmHg for control (n = 10), 27.2 ± 8.1 mmHg after naloxone and endomorphin 2 infusion (n = 10), and 20 ± 8 mmHg after APETx2 and endomorphin 2 infusion (n = 10). Infusion of endomorphin 2 and naloxone did not potentiate the pressor responses to contraction in ASIC3 knockout rats (n = 6). Partly similar findings were observed when oxycodone was substituted for endomorphin 2. Oxycodone infusion significantly increased the exercise pressor reflex over its control level, but subsequent APETx2 infusion failed to restore the increase to its control level (n = 9). The peak pressor response averaged 23.1 ± 8.6 mmHg for control (n = 9), 33.2 ± 11 mmHg after naloxone and oxycodone were infused (n = 9), and 27 ± 8.6 mmHg after APETx2 and oxycodone were infused (n = 9). Our data suggest that after opioid receptor blockade, ASIC3 stimulation by the endogenous mu opioid, endomorphin 2, potentiated the exercise pressor reflex.NEW & NOTEWORTHY This paper provides the first in vivo evidence that endomorphin 2, an endogenous opioid peptide, can paradoxically increase the magnitude of the exercise pressor reflex by an ASIC3-dependent mechanism even when the contracting muscles are freely perfused.


Subject(s)
Acid Sensing Ion Channels , Muscle Contraction , Muscle, Skeletal , Naloxone , Oligopeptides , Receptors, Opioid, mu , Reflex , Animals , Male , Rats , Acid Sensing Ion Channels/metabolism , Analgesics, Opioid/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oligopeptides/pharmacology , Oxycodone/pharmacology , Oxycodone/administration & dosage , Physical Conditioning, Animal/physiology , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Reflex/drug effects , Reflex/physiology
18.
Neuroscience ; 539: 76-85, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38211933

ABSTRACT

The use of mu-opioid receptor (MOP-r) agonists such as oxycodone together with cocaine is prevalent, and deaths attributed to using these combinations have increased. RATIONALE: It is unknown if functional single nucleotide polymorphisms (SNPs), such as the OPRM1 (MOP-r gene) SNP A118G, can predispose individuals to more dual opioid and psychostimulant intake. The dual self-administration (SA) of MOP-r agonists and cocaine has not been thoroughly examined, especially with regard to neurobiological changes. OBJECTIVES: We examined oxycodone SA and subsequent dual oxycodone and cocaine SA in male and female A112G (A/G and G/G, heterozygote and homozygote, respectively) mice, models of human A118G carriers, versus wild-type (A/A) mice. METHODS: Adult male and female A/G, G/G and A/A mice self-administered oxycodone (0.25 mg/kg/infusion, 4hr/session, FR 1.) for 10 consecutive days (sessions 1-10). Mice then self-administered cocaine (2 hr) following oxycodone SA (4 hr, as above) in each session for a further 10 consecutive days (sessions 11-20). Message RNA transcripts of 24 reward-related genes were examined in the dorsal striatum. RESULTS: Male and female A/G and G/G mice had greater oxycodone SA than A/A mice did in the initial 10 days and in the last 10 sessions. Further, A/G and G/G mice showed greater cocaine intake than A/A mice. Dorsal striatal mRNA levels of Pdyn, Fkbp5, Oprk1, and Oprm1 were altered following oxycodone and cocaine SA. CONCLUSIONS: These studies demonstrated that this functional genetic variation in Oprm1 affected dual opioid and cocaine SA and altered specific gene expression in the striatum.


Subject(s)
Cocaine , Oxycodone , Adult , Male , Female , Humans , Mice , Animals , Oxycodone/pharmacology , Analgesics, Opioid , Polymorphism, Single Nucleotide , Cocaine/pharmacology , Receptors, Opioid , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism
19.
Brain Behav Immun ; 117: 100-111, 2024 03.
Article in English | MEDLINE | ID: mdl-38199516

ABSTRACT

Oxycodone is the most prescribed opioid for pain management and has been available in clinics for almost a century, but effects of chronic oxycodone have been studied less than morphine in preclinical and clinical studies. Newly developed depression has been coupled with chronic oxycodone use in a few clinical studies, but no preclinical studies have investigated the pathogenesis of oxycodone-induced depression. Gut microbiome changes following oxycodone use is an understudied area, and interleukin-17A (IL-17A) is linked to both the development of mood disorders and regulation of gut microbiome. The present study investigated effects of chronic oxycodone exposure on mood-related behaviors (depression and anxiety), pain hypersensitivity, physical dependence, immune markers, and the gut microbiome and tested the hypothesis that blocking IL-17A with a systemically administered monoclonal antibody reduces oxycodone-derived effects. Oxycodone (using an incremental dosing regimen) or saline was injected twice a day for 12 days. IL-17A Ab (200 µg/100 µl) or saline was administered every 3rd day during the 12-day interval. Chronic oxycodone induced a depression-like effect, but not anxiogenic- or anxiolytic-like effects; promoted hyperalgesia; increased IL-17A and IL-6 levels in the ventral tegmental area (VTA); and induced physical dependence. IL-17A Ab co-administration with oxycodone prevented the depression-like effect and hyperalgesia, reduced naloxone-precipitated withdrawal signs, and normalized the increase in cytokine levels. Chronic oxycodone exposure did not affect gut microbiome and integrity. Our results identify a role for IL-17A in oxycodone-related behavioral and neuroimmune effects and show that IL-17A Ab has potential therapeutic value in blocking these effects. Given that humanized IL-17A Ab is approved for treatment of psoriasis and psoriatic arthritis, our findings point toward studying it for use in the treatment of oxycodone use disorder.


Subject(s)
Oxycodone , Substance-Related Disorders , Rats , Animals , Oxycodone/pharmacology , Ventral Tegmental Area , Interleukin-17/metabolism , Interleukin-6/pharmacology , Depression/drug therapy , Hyperalgesia/drug therapy
20.
Psychopharmacology (Berl) ; 241(2): 305-314, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37870564

ABSTRACT

RATIONALE: Combinations of mu and kappa-opioid receptor (KOR) agonists have been proposed as analgesic formulations with reduced abuse potential. The feasibility of this approach has been increased by the development of KOR agonists with biased signaling profiles that produce KOR-typical antinociception with fewer KOR-typical side effects. OBJECTIVE: The present study determined if the biased KOR agonists, nalfurafine and triazole 1.1, could reduce choice for oxycodone in rhesus monkeys as effectively as the typical KOR agonist, salvinorin A. METHODS: Adult male rhesus monkeys (N = 5) responded under a concurrent schedule of food delivery and intravenous cocaine injections (0.018 mg/kg/injection). Once trained, cocaine (0.018 mg/kg/injection) or oxycodone (0.0056 mg/kg/injection) was tested alone or in combination with contingent injections of salvinorin A (0.1-3.2 µg/kg/injection), nalfurafine (0.0032-0.1 µg/kg/injection), triazole 1.1 (3.2-100.0 µg/kg/injection), or vehicle. In each condition, the cocaine or oxycodone dose, as well as the food amount, was held constant across choice components, while the dose of the KOR agonist was increased across choice components. RESULTS: Cocaine and oxycodone were chosen over food on more than 80% of trials when administered alone or contingently with vehicle. When KOR agonists were administered contingently with either cocaine or oxycodone, drug choice decreased in a dose-dependent manner. Salvinorin A and triazole 1.1 decreased drug-reinforcer choice without altering total trials completed (i.e., choice allocation shifted to food), while nalfurafine dose dependently decreased total trials completed. CONCLUSIONS: These results demonstrate that salvinorin A and triazole 1.1, but not nalfurafine, selectively reduce cocaine and oxycodone self-administration independent of nonspecific effects on behavior, suggesting that G-protein bias does not appear to be a moderating factor in this outcome. Triazole 1.1 represents an important prototypical compound for developing novel KOR agonists as deterrents for prescription opioid abuse.


Subject(s)
Cocaine , Diterpenes, Clerodane , Morphinans , Oxycodone , Spiro Compounds , Animals , Male , Oxycodone/pharmacology , Analgesics, Opioid/pharmacology , Macaca mulatta , Pharmaceutical Preparations , Self Administration , Cocaine/pharmacology , Triazoles , Receptors, Opioid, kappa/agonists , Dose-Response Relationship, Drug
SELECTION OF CITATIONS
SEARCH DETAIL