Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
2.
Anesth Analg ; 132(2): 406-419, 2021 02 01.
Article En | MEDLINE | ID: mdl-33332902

A serious adverse effect of prescription opioid analgesics is addiction, both to these analgesics and to illicit drugs like heroin that also activate the µ-opioid receptor (MOR). Opioid use disorder (OUD) and opioid overdose deaths represent a current American health crisis, and the prescription of opioid analgesics has contributed significantly to this crisis. While prescription opioids are highly effective analgesics, there currently exists no facile way to use them for extended periods without the risk of addiction. If addiction caused by MOR-targeting analgesics could be blocked by blending in a new "antiaddiction" ingredient that does not diminish analgesia and does not introduce its own therapeutically limiting side effects, then continued clinical use of prescription opioids for treating pain could be maintained (or even enhanced) instead of curtailed. In this narrative review, we contextualize this hypothesis, first with a brief overview of the current American opioid addiction crisis. The neurobiology of 2 key receptors in OUD development, MOR and the κ-opioid receptor (KOR), is then discussed to highlight the neuroanatomical features and circuitry in which signal transduction from these receptors lie in opposition-creating opportunities for pharmacological intervention in curtailing the addictive potential of MOR agonism. Prior findings with mixed MOR/KOR agonists are considered before exploring new potential avenues such as biased KOR agonists. New preclinical data are highlighted, demonstrating that the G protein-biased KOR agonist nalfurafine reduces the rewarding properties of MOR-targeting analgesics and enhances MOR-targeting analgesic-induced antinociception. Finally, we discuss the recent discovery that a regulator of G protein signaling (namely, RGS12) is a key component of signaling bias at KOR, presenting another drug discovery target toward identifying a single agent or adjuvant to be added to traditional opioid analgesics that could reduce or eliminate the addictive potential of the latter drug.


Drug Design , Narcotic Antagonists/pharmacology , Nociception/drug effects , Nociceptive Pain/drug therapy , Opioid-Related Disorders/prevention & control , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Animals , Humans , Molecular Structure , Narcotic Antagonists/adverse effects , Narcotic Antagonists/chemistry , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Nociceptive Pain/psychology , Opioid-Related Disorders/etiology , RGS Proteins/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction , Structure-Activity Relationship
3.
J Psychopharmacol ; 34(12): 1393-1407, 2020 12.
Article En | MEDLINE | ID: mdl-32842837

BACKGROUND: Regulator of G protein Signaling (RGS) proteins inhibit G protein-coupled receptor (GPCR) signaling, including the signals that arise from neurotransmitter release. We have shown that RGS12 loss diminishes locomotor responses of C57BL/6J mice to dopamine transporter (DAT)-targeting psychostimulants. This diminution resulted from a brain region-specific upregulation of DAT expression and function in RGS12-null mice. This effect on DAT prompted us to investigate whether the serotonin transporter (SERT) exhibits similar alterations upon RGS12 loss in C57BL/6J mice. AIMS: Does RGS12 loss affect (a) hyperlocomotion to the preferentially SERT-targeting psychostimulant 3,4-methylenedioxymethamphetamine (MDMA), (b) SERT expression and function in relevant brain regions, and/or (c) serotonergically modulated behaviors? METHODS: Open-field and spontaneous home-cage locomotor activities were quantified. 5-HT, 5-HIAA, and SERT levels in brain-region homogenates, as well as SERT expression and function in brain-region tissue preparations, were measured using appropriate biochemical assays. Serotonergically modulated behaviors were assessed using forced swim and tail suspension paradigms, elevated plus and elevated zero maze tests, and social interaction assays. RESULTS: RGS12-null mice displayed no hyperlocomotion to 10 mg/kg MDMA. There were brain region-specific alterations in SERT expression and function associated with RGS12 loss. Drug-naïve RGS12-null mice displayed increases in both anxiety-like and anti-depressive-like behaviors. CONCLUSION: RGS12 is a critical modulator of serotonergic neurotransmission and serotonergically modulated behavior in mice; lack of hyperlocomotion to low dose MDMA in RGS12-null mice is related to an alteration of steady-state SERT expression and 5-HT uptake.


Behavior, Animal/physiology , Locomotion/physiology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , RGS Proteins/physiology , Serotonin Agents/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Behavior, Animal/drug effects , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , RGS Proteins/genetics , Serotonin Agents/administration & dosage , Social Behavior
4.
J Pharmacol Exp Ther ; 371(2): 487-499, 2019 11.
Article En | MEDLINE | ID: mdl-31492823

Mu opioid receptor (MOR)-targeting analgesics are efficacious pain treatments, but notorious for their abuse potential. In preclinical animal models, coadministration of traditional kappa opioid receptor (KOR)-targeting agonists with MOR-targeting analgesics can decrease reward and potentiate analgesia. However, traditional KOR-targeting agonists are well known for inducing antitherapeutic side effects (psychotomimesis, depression, anxiety, dysphoria). Recent data suggest that some functionally selective, or biased, KOR-targeting agonists might retain the therapeutic effects of KOR activation without inducing undesirable side effects. Nalfurafine, used safely in Japan since 2009 for uremic pruritus, is one such functionally selective KOR-targeting agonist. Here, we quantify the bias of nalfurafine and several other KOR agonists relative to an unbiased reference standard (U50,488) and show that nalfurafine and EOM-salvinorin-B demonstrate marked G protein-signaling bias. While nalfurafine (0.015 mg/kg) and EOM-salvinorin-B (1 mg/kg) produced spinal antinociception equivalent to 5 mg/kg U50,488, only nalfurafine significantly enhanced the supraspinal analgesic effect of 5 mg/kg morphine. In addition, 0.015 mg/kg nalfurafine did not produce significant conditioned place aversion, yet retained the ability to reduce morphine-induced conditioned place preference in C57BL/6J mice. Nalfurafine and EOM-salvinorin-B each produced robust inhibition of both spontaneous and morphine-stimulated locomotor behavior, suggesting a persistence of sedative effects when coadministered with morphine. Taken together, these findings suggest that nalfurafine produces analgesic augmentation, while also reducing opioid-induced reward with less risk of dysphoria. Thus, adjuvant administration of G protein-biased KOR agonists like nalfurafine may be beneficial in enhancing the therapeutic potential of MOR-targeting analgesics, such as morphine.


Analgesia/methods , Drug Delivery Systems/methods , Morphinans/administration & dosage , Morphine/administration & dosage , Pain Measurement/drug effects , Receptors, Opioid, mu/metabolism , Spiro Compounds/administration & dosage , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Drug Synergism , Female , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Pain Measurement/methods , Random Allocation , Receptors, Opioid, kappa/administration & dosage , Receptors, Opioid, mu/agonists
5.
J Opioid Manag ; 15(2): 103-109, 2019.
Article En | MEDLINE | ID: mdl-31057342

OBJECTIVE: Pilot study to assess utility in opioid use disorder (OUD) of a panel of single nucleotide polymorphisms in genes previously related to substance use disorder (SUD) and/or phenotypes that predispose individuals to OUD/SUD. DESIGN: Genetic association study. SETTING: West Virginia University's Chestnut Ridge Center Comprehensive Opioid Abuse Treatment (COAT) clinic for individuals diagnosed with OUD. PATIENTS: Sixty patients 18 years of age or older with OUD undergoing medication (buprenorphine/naloxone)-assisted treatment (MAT); all sixty patients recruited contributed samples for genetic analysis. OUTCOME MEASURES: Minor allele frequencies for single nucleotide polymorphisms. RESULTS: Four of the fourteen single nucleotide polymorphisms examined were present at frequencies that are statistically significantly different than in a demographically-matched general population. CONCLUSIONS: For the purposes of testing WV individuals via genetic means for predisposition to OUD, at least four single nucleotide polymorphisms in three genes are likely to have utility in predicting susceptibility. Additional studies with larger populations will need to be conducted to confirm these results before use in a clinical setting.


Opioid-Related Disorders/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Analgesics, Opioid , Genetic Association Studies , Humans , Pilot Projects , West Virginia
6.
Neuropsychopharmacology ; 44(10): 1728-1741, 2019 09.
Article En | MEDLINE | ID: mdl-31141817

Kappa opioid receptor (KOR) agonists show promise in ameliorating disorders, such as addiction and chronic pain, but are limited by dysphoric and aversive side effects. Clinically beneficial effects of KOR agonists (e.g., analgesia) are predominantly mediated by heterotrimeric G protein signaling, whereas ß-arrestin signaling is considered central to their detrimental side effects (e.g., dysphoria/aversion). Here we show that Regulator of G protein Signaling-12 (RGS12), via independent signaling mechanisms, simultaneously attenuates G protein signaling and augments ß-arrestin signaling downstream of KOR, exhibiting considerable selectivity in its actions for KOR over other opioid receptors. We previously reported that RGS12-null mice exhibit increased dopamine transporter-mediated dopamine (DA) uptake in the ventral (vSTR), but not dorsal striatum (dSTR), as well as reduced psychostimulant-induced hyperlocomotion; in the current study, we found that these phenotypes are reversed following KOR antagonism. Fast-scan cyclic voltammetry studies of dopamine (DA) release and reuptake suggest that striatal disruptions to KOR-dependent DAergic neurotransmission in RGS12-null mice are restricted to the nucleus accumbens. In both ventral striatal tissue and transfected cells, RGS12 and KOR are seen to interact within a protein complex. Ventral striatal-specific increases in KOR levels and KOR-induced G protein activation are seen in RGS12-null mice, as well as enhanced sensitivity to KOR agonist-induced hypolocomotion and analgesia-G protein signaling-dependent behaviors; a ventral striatal-specific increase in KOR levels was also observed in ß-arrestin-2-deficient mice, highlighting the importance of ß-arrestin signaling to establishing steady-state KOR levels in this particular brain region. Conversely, RGS12-null mice exhibited attenuated KOR-induced conditioned place aversion (considered a ß-arrestin signaling-dependent behavior), consistent with the augmented KOR-mediated ß-arrestin signaling seen upon RGS12 over-expression. Collectively, our findings highlight a role for RGS12 as a novel, differential regulator of both G protein-dependent and -independent signaling downstream of KOR activation.


Dopamine/metabolism , Nucleus Accumbens/metabolism , RGS Proteins/genetics , Receptors, Opioid, kappa/metabolism , Ventral Striatum/metabolism , beta-Arrestins/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, Leucine-2-Alanine/pharmacology , Female , Locomotion/drug effects , Male , Mice , Mice, Knockout , Nucleus Accumbens/drug effects , Receptors, Opioid, kappa/agonists , Signal Transduction , Synaptic Transmission/drug effects , Ventral Striatum/drug effects
7.
Chem Senses ; 43(5): 367-378, 2018 05 23.
Article En | MEDLINE | ID: mdl-29701767

The mammalian tastes of sweet, umami, and bitter are initiated by activation of G protein-coupled receptors (GPCRs) of the T1R and T2R families on taste receptor cells. GPCRs signal via nucleotide exchange and hydrolysis, the latter hastened by GTPase-accelerating proteins (GAPs) that include the Regulators of G protein Signaling (RGS) protein family. We previously reported that RGS21, uniquely expressed in Type II taste receptor cells, decreases the potency of bitter-stimulated T2R signaling in cultured cells, consistent with its in vitro GAP activity. However, the role of RGS21 in organismal responses to GPCR-mediated tastants was not established. Here, we characterized mice lacking the Rgs21 fifth exon. Eliminating Rgs21 expression had no effect on body mass accumulation (a measure of alimentation), fungiform papillae number and morphology, circumvallate papillae morphology, and taste bud number. Two-bottle preference tests, however, revealed that Rgs21-null mice have blunted aversion to quinine and denatonium, and blunted preference for monosodium glutamate, the sweeteners sucrose and SC45647, and (surprisingly) NaCl. Observed reductions in GPCR-mediated tastant responses upon Rgs21 loss are opposite to original expectations, given that loss of RGS21-a GPCR signaling negative regulator-should lead to increased responsiveness to tastant-mediated GPCR signaling (all else being equal). Yet, reduced organismal tastant responses are consistent with observations of reduced chorda tympani nerve recordings in Rgs21-null mice. Reduced tastant-mediated responses and behaviors exhibited by adult mice lacking Rgs21 expression since birth have thus revealed an underappreciated requirement for a GPCR GAP to establish the full character of tastant signaling.


Food Preferences , RGS Proteins/metabolism , Taste , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , RGS Proteins/deficiency , RGS Proteins/genetics
8.
J Psychopharmacol ; 32(2): 191-203, 2018 02.
Article En | MEDLINE | ID: mdl-29364035

Regulators of G protein signaling are proteins that accelerate the termination of effector stimulation after G protein-coupled receptor activation. Many regulators of G protein signaling proteins are highly expressed in the brain and therefore considered potential drug discovery targets for central nervous system pathologies; for example, here we show that RGS12 is highly expressed in microdissected mouse ventral striatum. Given a role for the ventral striatum in psychostimulant-induced locomotor activity, we tested whether Rgs12 genetic ablation affected behavioral responses to amphetamine and cocaine. RGS12 loss significantly decreased hyperlocomotion to lower doses of both amphetamine and cocaine; however, other outcomes of administration (sensitization and conditioned place preference) were unaffected, suggesting that RGS12 does not function in support of the rewarding properties of these psychostimulants. To test whether observed response changes upon RGS12 loss were caused by changes to dopamine transporter expression and/or function, we prepared crude membranes from the brains of wild-type and RGS12-null mice and measured dopamine transporter-selective [3H]WIN 35428 binding, revealing an increase in dopamine transporter levels in the ventral-but not dorsal-striatum of RGS12-null mice. To address dopamine transporter function, we prepared striatal synaptosomes and measured [3H]dopamine uptake. Consistent with increased [3H]WIN 35428 binding, dopamine transporter-specific [3H]dopamine uptake in RGS12-null ventral striatal synaptosomes was found to be increased. Decreased amphetamine-induced locomotor activity and increased [3H]WIN 35428 binding were recapitulated with an independent RGS12-null mouse strain. Thus, we propose that RGS12 regulates dopamine transporter expression and function in the ventral striatum, affecting amphetamine- and cocaine-induced increases in dopamine levels that specifically elicit acute hyperlocomotor responses.


Central Nervous System Stimulants/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Locomotion/drug effects , RGS Proteins/genetics , Amphetamine/administration & dosage , Amphetamine/pharmacology , Animals , Central Nervous System Stimulants/administration & dosage , Cocaine/administration & dosage , Cocaine/analogs & derivatives , Cocaine/pharmacology , Dopamine/metabolism , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Mice, Knockout , Reward , Signal Transduction/drug effects , Ventral Striatum/drug effects , Ventral Striatum/metabolism
...