Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Drug Alcohol Depend ; 242: 109719, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36521236

ABSTRACT

Riluzole, approved to manage amyotrophic lateral sclerosis, is mechanistically unique among glutamate-based therapeutics because it reduces glutamate transmission through a dual mechanism (i.e., reduces glutamate release and enhances glutamate reuptake). The profile of riluzole is favorable for normalizing glutamatergic dysregulation that perpetuates methamphetamine (METH) dependence, but pharmacokinetic and metabolic liabilities hinder repurposing. To mitigate these limitations, we synthesized troriluzole (TRLZ), a third-generation prodrug of riluzole, and tested the hypothesis that TRLZ inhibits METH hyperlocomotion and conditioned place preference (CPP) and normalizes METH-induced changes in mesolimbic glutamate biomarkers. TRLZ (8, 16 mg/kg) reduced hyperlocomotion caused by METH (1 mg/kg) without affecting spontaneous activity. TRLZ (1, 4, 8, 16 mg/kg) administered during METH conditioning (0.5 mg/kg x 4 d) inhibited development of METH place preference, and TRLZ (16 mg/kg) administered after METH conditioning reduced expression of CPP. In rats with established METH place preference, TRLZ (16 mg/kg) accelerated extinction of CPP. In cellular studies, chronic METH enhanced mRNA levels of glutamate carboxypeptidase II (GCPII) in the ventral tegmental area (VTA) and prefrontal cortex (PFC). Repeated METH also caused enhancement of GCPII protein levels in the VTA that was prevented by TRLZ (16 mg/kg). TRLZ (16 mg/kg) administered during chronic METH did not affect brain or plasma levels of METH. These results indicate that TRLZ, already in clinical trials for cerebellar ataxia, reduces development, expression and maintenance of METH CPP. Moreover, normalization of METH-induced GCPII levels in mesolimbic substrates by TRLZ points toward studying GCPII as a therapeutic target of TRLZ.


Subject(s)
Amphetamine-Related Disorders , Central Nervous System Stimulants , Methamphetamine , Rats , Animals , Methamphetamine/pharmacology , Central Nervous System Stimulants/pharmacology , Glutamate Carboxypeptidase II/therapeutic use , Riluzole/therapeutic use , Amphetamine-Related Disorders/drug therapy , Glutamates/therapeutic use
2.
Drug Alcohol Depend ; 238: 109556, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35843139

ABSTRACT

Chemokine-opioid crosstalk is a physiological crossroads for influencing therapeutic and adverse effects of opioids. Activation of chemokine receptors, especially CCR2, CCR5 and CXCR4, reduces opioid-induced analgesia by desensitizing OPRM1 receptors. Chemokine receptor antagonists (CRAs) enhance opioid analgesia, but knowledge about how CRAs impact adverse opioid effects remains limited. We examined effects of RAP-103, a multi-CRA orally active peptide analog of "DAPTA", on opioid-derived dependence, reinforcement, and respiratory depression in male rats and on changes in chemokine and OPRM1 (µ opioid) receptor levels in mesolimbic substrates during opioid abstinence. In rats exposed to chronic morphine (75 mg pellet x 7 d), daily RAP-103 (1 mg/kg, IP) treatment reduced the severity of naloxone-precipitated withdrawal responses. For self-administration (SA) studies, RAP-103 (1 mg/kg, IP) reduced heroin acquisition (0.1 mg/kg/inf) and reinforcing efficacy (assessed by motivation on a progressive-ratio reinforcement schedule) but did not impact sucrose intake. RAP-103 (1-3 mg/kg, IP) also normalized the deficits in oxygen saturation and enhancement of respiratory rate caused by morphine (5 mg/kg, SC) exposure. Abstinence from chronic morphine elicited brain-region specific changes in chemokine receptor protein levels. CCR2 and CXCR4 were increased in the ventral tegmental area (VTA), whereas CCR2 and CCR5 were reduced in the nucleus accumbens (NAC). Effects of RAP-103 (1 mg/kg, IP) were focused in the NAC, where it normalized morphine-induced deficits in CCR2 and CCR5. These results identify CRAs as potential biphasic function opioid signaling modulators to enhance opioid analgesia and inhibit opioid-derived dependence and respiratory depression.


Subject(s)
Analgesics, Opioid , Respiratory Insufficiency , Analgesics, Opioid/pharmacology , Animals , Male , Morphine/pharmacology , Nucleus Accumbens , Peptides/metabolism , Peptides/pharmacology , Rats , Receptors, Chemokine/metabolism , Receptors, Opioid , Receptors, Opioid, mu , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy
3.
Drug Alcohol Depend ; 230: 109204, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34871976

ABSTRACT

Chemokine CXCR4 and CCR5 receptors are best known as HIV co-entry receptors, but evidence that CXCR4 or CCR5 blockade reduces rewarding and locomotor-stimulant effects of psychostimulants in rats suggests a role in psychostimulant use disorders. We investigated the impact of CXCR4 or CCR5 receptor antagonism on anxiety-related effects of the synthetic cathinone 3,4-methylenedioxypyrovalerone (MDPV) in the elevated zero-maze (EZM) assay. Rats exposed to a 4-day MDPV binge dosing paradigm and tested 24 or 72 h post-treatment spent more time in the open compartment at the 24-h time point but less time at the 72-h post-binge time point. Daily administration of AMD 3100, a CXCR4 antagonist (10 mg/kg), or maraviroc, a CCR5 antagonist (2.5 mg/kg), during MDPV treatment inhibited the MDPV-induced increase in time spent in the open compartment. Neither antagonist affected the MDPV-induced reduction in time spent in the open compartment at the 72-h post-binge time point. Cocaine, administered in the same paradigm as MDPV, did not increase time spent in the open compartment 24-h post-binge, suggesting specificity to MDPV. The present results identify a surprising anxiolytic-like effect of MDPV 24 h after cessation of repeated exposure that is sensitive to chemokine CXCR4 and CCR5 receptor activity.


Subject(s)
Anti-Anxiety Agents , Receptors, CCR5 , Alkaloids , Animals , Anti-Anxiety Agents/pharmacology , Benzodioxoles , CCR5 Receptor Antagonists/pharmacology , Chemokines , Pyrrolidines , Rats , Receptors, CXCR4 , Synthetic Cathinone
4.
Life Sci ; 285: 120014, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34619167

ABSTRACT

AIMS: We have shown that chemokines injected into the periaqueductal gray region of the brain blocks opioid-induced analgesia in the rat cold-water tail flick test (CWTF). The present experiments tested whether chemokine receptor antagonists (CRAs), in combination with sub-analgesic doses of morphine, would provide maximal analgesia in the CWTF test and the mouse formalin pain assay. The effect of CRAs on respiratory depression was also evaluated. MAIN METHODS: One, two or four CRAs (AMD3100/CXCR4, maraviroc/CCR5, RS504393/CCR2 orAZD8797/CX3CR1) were used in combination with sub-analgesic doses of morphine, all given systemically. Pain was assessed using the rat CWTF test or formalin injection into the paw of mice scored by licking. Respiration and oxygen saturation were measured in rats using a MouseOX® Plus - pulse oximeter. KEY FINDINGS: In the CWTF test, a sub-maximal dose of morphine in combination with maraviroc alone, maraviroc plus AMD3100, or with the four chemokine receptor antagonists, produced synergistic increases in antinociception. In the formalin test, the combination of four CRAs plus a sub-maximal dose of morphine resulted in increased antinociception in both male and female mice. AMD3100 had an additive effect with morphine in both sexes. Coadministration of CRAs with morphine did not potentiate the opioid respiratory depressive effect. SIGNIFICANCE: These results support the conclusion that combinations of CRAs can increase the potency of sub-analgesic doses of morphine analgesia without increasing respiratory depression. The results support an "opioid sparing" strategy for alleviation of pain using reduced doses of opioids in combination with CRAs to achieve maximal analgesia.


Subject(s)
Analgesia/methods , Analgesics, Opioid/pharmacology , Morphine/pharmacology , Nociception/drug effects , Nociceptive Pain/drug therapy , Receptors, Chemokine/antagonists & inhibitors , Animals , Benzylamines/administration & dosage , Benzylamines/pharmacology , Cyclams/administration & dosage , Cyclams/pharmacology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Male , Maraviroc/administration & dosage , Maraviroc/pharmacology , Morphine/administration & dosage , Morphine/adverse effects , Nociceptive Pain/physiopathology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Respiratory Insufficiency/chemically induced , Thiazoles/administration & dosage , Thiazoles/pharmacology
5.
Physiol Behav ; 239: 113499, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34146575

ABSTRACT

Mitragynine (MG), the most prevalent bioactive alkaloid in kratom, displays nanomolar affinity for µ, κ and δ opioid receptors and produces opioid-dependent antinociception and dependence in rats. Here, using a battery of behavioral assays, we investigated MG effects in planarians. Acute MG exposure (< 100 µM) did not affect planarian motility or environmental preference, but reduced motility was detected during abstinence from chronic MG (1, 10 µM). MG (10 µM) produced place conditioning effects that were reduced by naltrexone (10  µΜ). These results suggest that MG produces opioid-sensitive reinforcing effects in planarians and MG pharmacology is conserved across different species.


Subject(s)
Mitragyna , Planarians , Secologanin Tryptamine Alkaloids , Animals , Naltrexone/pharmacology , Rats , Secologanin Tryptamine Alkaloids/pharmacology
6.
Drug Alcohol Depend ; 220: 108511, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33465606

ABSTRACT

Efficacious pharmacotherapies for the treatment of substance use disorders need to be expanded and improved. Non-neuronal cells, particularly astrocytes and microglia, have emerged as therapeutic targets for the development of pharmacotherapies to treat dependence and relapse that accompanies chronic drug use. Cytokines and chemokines are neuroimmune factors expressed in neurons, astrocytes, and microglia that demonstrate promising clinical utility as therapeutic targets for substance use disorders. In this review, we describe a role for cytokines and chemokines in the rewarding and reinforcing effects of alcohol, opioids, and psychostimulants. We also discuss emerging cytokine- and chemokine-based therapeutic strategies that differ from conventional strategies directed toward transporters and receptors within the dopamine, glutamate, GABA, serotonin, and GABA systems.


Subject(s)
Chemokines/metabolism , Cytokines/metabolism , Substance-Related Disorders/metabolism , Analgesics, Opioid/metabolism , Animals , Astrocytes/drug effects , Dopamine , Glutamic Acid , Humans , Microglia/drug effects , Neurons , Serotonin
7.
Addict Behav ; 114: 106744, 2021 03.
Article in English | MEDLINE | ID: mdl-33291057

ABSTRACT

School-based drug prevention programs represent a widely endorsed public health goal, with an important aspect of knowledge-based curricula being education about the physiological effects of drugs. Nicotine is one of the world's most addictive substances and in this program we have used nicotine-induced mammalian-like behaviors in flatworms called planarians to successfully teach students (4th-12th grade; n = 1,616 students) about the physiological and addictive effects of nicotine. An initial study tested the change in knowledge about addictive substances in 6th-12th grade students after they completed a lab examining the effects of two concentrations of nicotine on the number of stereotypies (C-shaped spasms) planarians demonstrate in a 5-minute period of time. Lab discussion focused on developing and testing hypotheses, measurement reliability, and mechanisms of nicotine action. Surveys given pre- and post-lab experience showed that 6th grade students have significantly lower knowledge about nicotine than 7th-12th grade students (6th grade: 40.65 ± 0.78% correct, 7th-12th grade: 59.29 ± 1.71%, p < 0.001) pre-lab, but that students in all grades showed a significant increase in knowledge post-lab (p < 0.001). In 6th grade the lab was effective in improving knowledge about nicotine in urban, suburban and rural schools, p < 0.001, with students in suburban schools showing significantly greater knowledge both pre-test (urban: 37.62 ± 1.45%; suburban: 48.78 ± 1.62%; rural: 37.33 ± 0.99%; p < 0.001) and post-test (urban:60.60 ± 1.85%; suburban: 67.54 ± 1.82%; urban: 61.66 ± 1.18%; p < 0.001). A second study, modifying the lab so that the time spent observing the planarians is reduced to a 1-minute period, showed that students in both 4th and 5th grades had a significant increase in knowledge about the physiological and addictive effects of nicotine post-lab (p < 0.001).


Subject(s)
Nicotine , Schools , Animals , Health Knowledge, Attitudes, Practice , Humans , Reproducibility of Results , Rural Population , Students
8.
Br J Pharmacol ; 176(17): 3378-3389, 2019 09.
Article in English | MEDLINE | ID: mdl-31218677

ABSTRACT

BACKGROUND AND PURPOSE: Much of the opioid epidemic arose from abuse of prescription opioid drugs. This study sought to determine if the combination of a cannabinoid with an opioid could produce additive or synergistic effects on pain, allowing reduction in the opioid dose needed for maximal analgesia. EXPERIMENTAL APPROACH: Pain was assayed using the formalin test in mice and the carrageenan assay in rats. Morphine and two synthetic cannabinoids were tested: WIN55,212-2 (WIN), which binds to both CB1 and CB2 receptors, and possibly TRPV1 channels; and GP1a, which has activity at CB2 receptors and is reported to inhibit fatty acid amide hydrolase, thus raising levels of endogenous cannabinoids. KEY RESULTS: Morphine in combination with WIN in the formalin test gave synergistic analgesia. Studies with selective antagonists showed that WIN was acting through CB1 receptors. Morphine in combination with GP1a in the formalin test was sub-additive. In the carrageenan test, WIN had no added effect when combined with morphine, but GP1a with morphine showed enhanced analgesia. Both WIN and Gp1a used alone had analgesic activity in the formalin pain test, but not in the carrageenan pain test. CONCLUSIONS AND IMPLICATIONS: The ability of a cannabinoid to produce an additive or synergistic effect on analgesia when combined with morphine varies with the pain assay and may be mediated by CB1 or CB2 receptors. These results hold the promise of using cannabinoids to reduce the dose of opioids for analgesia in certain pain conditions.


Subject(s)
Analgesics, Opioid/pharmacology , Cannabinoids/pharmacology , Morphine/pharmacology , Pain/drug therapy , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Animals , Carrageenan , Dose-Response Relationship, Drug , Formaldehyde , Male , Mice , Pain/chemically induced , Pain/metabolism , Pain Management , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
10.
Drug Alcohol Depend ; 197: 22-27, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30754021

ABSTRACT

BACKGROUND AND PURPOSE: Purinergic P2X7 receptors are present on neurons, astrocytes and microglia and activated by extracellular ATP. Since P2X7 receptor activation releases endogenous substrates (e.g., pro-inflammatory cytokines, dopamine, and glutamate) that facilitate psychostimulant reward and reinforcement, we investigated the hypothesis that the synthetic cathinone 3,4-methylenedioxypyrovalerone (MDPV) produces rewarding effects that are dependent on active P2X7 receptors. METHODS: Reward function was measured in male mice using intracranial self-stimulation (ICSS). MDPV (0.1, 0.3, 0.5 mg/kg, SC) and a selective P2X7 antagonist (A438079) (5, 10, 50 mg/kg, IP) were tested alone and in combination. In separate mice, gene and protein expression of P2X7 and mitochondrial adenosine triphosphate (ATP) synthase (an enzyme that catalyzes synthesis of ATP, an endogenous ligand for P2X7 receptors) in the nucleus accumbens (NAcc) were quantified following MDPV exposure (0.1, 0.5, 5 mg/kg, SC). KEY RESULTS: MDPV (0.5 mg/kg, SC) facilitated ICSS as quantified by a significant reduction in brain reward threshold. A438079 (5, 10, 50 mg/kg, IP) did not affect ICSS by itself; however, for combined administration, A438079 (10 mg/kg, IP) inhibited facilitation of ICSS by MDPV (0.5 mg/kg, SC). At the cellular level, MDPV exposure increased gene and protein expression of P2X7 and ATP synthase in the NAcc. CONCLUSION AND IMPLICATION: We provide evidence that a psychostimulant drug produces reward enhancement that is influenced by P2X7 receptor activity and enhances P2X7 receptor expression in the brain reward circuit.


Subject(s)
Alkaloids/pharmacology , Benzodioxoles/pharmacology , Central Nervous System Stimulants/pharmacology , Nucleus Accumbens/drug effects , Pyrrolidines/pharmacology , Receptors, Purinergic P2X7/drug effects , Reward , Animals , Brain/drug effects , Male , Mice , Self Stimulation/drug effects , Synthetic Cathinone
11.
J Pharmacol Exp Ther ; 367(3): 433-441, 2018 12.
Article in English | MEDLINE | ID: mdl-30249618

ABSTRACT

Crossdesensitization between opioid and chemokine receptors and involvement of chemokines in pain modulation are well established. We investigated if coadministration of chemokine receptor antagonists (CRAs) with morphine would enhance the analgesic potency of morphine on incisional pain in rats. Animals underwent incisional surgery on the left hind paw and pain responses were evaluated using von Frey filaments at various time points postsurgery between 15 and 360 minutes and daily between 24 and 72 hours. Dose-response curves for morphine, maraviroc (a CCR5 antagonist), and AMD3100 (a CXCR4 antagonist) alone were established. While morphine significantly reduced pain in a time- and dose-dependent manner, maraviroc and AMD3100 had no effect by themselves. Coadministration of either maraviroc or AMD3100 with morphine significantly increased morphine's analgesic effect on incisional pain, shifting the dose-response curve to the left 2.3- and 1.8-fold, respectively. Coadministration of both CRAs with morphine significantly shifted further the morphine dose-response curve to the left 3.3-fold. The effect of treatments on mRNA levels in the draining popliteal lymph node for a panel of chemokines and cytokines showed that message for many of these mediators was upregulated by the incision, and the combination of morphine with the CRAs markedly downregulated them. The data show that combining morphine with CRAs potentiates morphine's analgesic effect on incisional pain. Thus, the same analgesic effect of morphine alone can be achieved with lower doses of morphine when combined with CRAs. Using morphine in lower doses could reduce unwanted side effects and possibly block development of tolerance and dependence.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Pain/drug therapy , Receptors, Chemokine/antagonists & inhibitors , Animals , Down-Regulation/drug effects , Drug Tolerance/physiology , Male , Pain/metabolism , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/metabolism
12.
Neuropsychopharmacology ; 43(5): 1001-1009, 2018 04.
Article in English | MEDLINE | ID: mdl-28741623

ABSTRACT

Hypothalamic hypocretin (orexin) peptides mediate arousal, attention, and reward processing. Fibers containing orexins project to brain structures that govern motivated behavior, including the ventral tegmental area (VTA). A number of psychiatric conditions, including attention deficit hyperactivity disorder (ADHD) and substance use disorders, are characterized by deficits in impulse control, however the relationship between orexin and impulsive behavior is incompletely characterized. The effects of systemic or centrally administered orexin receptor (OXR) antagonists on measures of impulsive-like behavior in rats were evaluated using the five-choice serial reaction time task (5-CSRTT) and delay discounting procedures. These paradigms were also used to test the capacity of OXR antagonists to attenuate acute cocaine-evoked impulsivity. Finally, immunohistochemistry and calcium imaging were used to assess potential cellular mechanisms by which OXR blockade may influence motor impulsivity. Suvorexant, a dual (OX1/2R) orexin receptor antagonist, reduced cocaine-evoked premature responses in 5-CSRTT when administered systemically or directly into VTA. Neither suvorexant nor OX1R- or OX2R-selective compounds (SB334867 or TCS-OX2-29, respectively) altered delay discounting. Finally, suvorexant did not alter Fos-immunoreactivity within tyrosine hydroxylase-immunolabeled neurons of VTA, but did attenuate cocaine- and orexin-induced increases in calcium transient amplitude within neurons of VTA. Results from the present studies suggest potential therapeutic utility of OXR antagonists in reducing psychostimulant-induced motor impulsivity. These findings also support the view that orexin transmission is closely involved in executive function in normal and pathological conditions.


Subject(s)
Azepines/pharmacology , Cocaine/antagonists & inhibitors , Cocaine/pharmacology , Impulsive Behavior/drug effects , Orexin Receptor Antagonists/pharmacology , Triazoles/pharmacology , Animals , Benzoxazoles/pharmacology , Choice Behavior , Delay Discounting/drug effects , Isoquinolines/pharmacology , Male , Microinjections , Naphthyridines , Orexins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Pyridines/pharmacology , Rats , Urea/analogs & derivatives , Urea/pharmacology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...