Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
Front Neurosci ; 18: 1390215, 2024.
Article in English | MEDLINE | ID: mdl-38817910

ABSTRACT

Cognitive decline in Parkinson's disease (PD) is a critical premotor sign that may occur in approximately 40% of PD patients up to 10 years prior to clinical recognition and diagnosis. Delineating the mechanisms and specific behavioral signs of cognitive decline associated with PD prior to motor impairment is a critical unmet need. Rodent PD models that have an impairment in a cognitive phenotype for a time period sufficiently long enough prior to motor decline can be useful to establish viable candidate mechanisms. Arguably, the methods used to evaluate cognitive decline in rodent models should emulate methods used in the assessment of humans to optimize translation. Premotor cognitive decline in human PD can potentially be examined in the genetically altered PINK1-/- rat model, which exhibits a protracted onset of motor decline in most studies. To increase translation to cognitive assessment in human PD, we used a modified non-water multiple T-maze, which assesses attention, cognitive flexibility, and working memory similarly to the Trail Making Test (TMT) in humans. Similar to the deficiencies revealed in TMT test outcomes in human PD, 4-month-old PINK1-/- rats made more errors and took longer to complete the maze, despite a hyperkinetic phenotype, compared to wild-type rats. Thus, we have identified a potential methodological tool with cross-species translation to evaluate executive functioning in an established PD rat model.

2.
bioRxiv ; 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38293068

ABSTRACT

Cognitive decline in Parkinson's disease (PD) emerges up to 10 years before clinical recognition. Neurobiological mechanisms underlying premotor cognitive impairment in PD can potentially be examined in the PINK1 -/- rat, which exhibits a protracted motor onset. To enhance translation to human PD cognitive assessments, we tested a modified multiple T-maze, which measures cognitive flexibility similarly to the Trail-Making Test in humans. Like human PD outcomes, PINK1 -/- rats made more errors and took longer to complete the maze than wild types. Thus, we have identified a potential tool for assessing cross-species translation of cognitive functioning in an established PD animal model.

3.
J Biol Chem ; 298(8): 102167, 2022 08.
Article in English | MEDLINE | ID: mdl-35738399

ABSTRACT

The free-living amoeba Naegleria fowleri is a causative agent of primary amoebic meningoencephalitis and is highly resistant to current therapies, resulting in mortality rates >97%. As many therapeutics target G protein-centered signal transduction pathways, further understanding the functional significance of G protein signaling within N. fowleri should aid future drug discovery against this pathogen. Here, we report that the N. fowleri genome encodes numerous transcribed G protein signaling components, including G protein-coupled receptors, heterotrimeric G protein subunits, regulator of G protein signaling (RGS) proteins, and candidate Gα effector proteins. We found N. fowleri Gα subunits have diverse nucleotide cycling kinetics; Nf Gα5 and Gα7 exhibit more rapid nucleotide exchange than GTP hydrolysis (i.e., "self-activating" behavior). A crystal structure of Nf Gα7 highlights the stability of its nucleotide-free state, consistent with its rapid nucleotide exchange. Variations in the phosphate binding loop also contribute to nucleotide cycling differences among Gα subunits. Similar to plant G protein signaling pathways, N. fowleri Gα subunits selectively engage members of a large seven-transmembrane RGS protein family, resulting in acceleration of GTP hydrolysis. We show Nf Gα2 and Gα3 directly interact with a candidate Gα effector protein, RGS-RhoGEF, similar to mammalian Gα12/13 signaling pathways. We demonstrate Nf Gα2 and Gα3 each engage RGS-RhoGEF through a canonical Gα/RGS domain interface, suggesting a shared evolutionary origin with G protein signaling in the enteric pathogen Entamoeba histolytica. These findings further illuminate the evolution of G protein signaling and identify potential targets of pharmacological manipulation in N. fowleri.


Subject(s)
Amoeba , Naegleria fowleri , RGS Proteins , Animals , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Proteins/metabolism , Guanosine Triphosphate/metabolism , Mammals/metabolism , Naegleria fowleri/metabolism , RGS Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/physiology
4.
Chem Senses ; 462021 01 01.
Article in English | MEDLINE | ID: mdl-34718440

ABSTRACT

The T1R and T2R families of G protein-coupled receptors (GPCRs) initiate tastant perception by signaling via guanine nucleotide exchange and hydrolysis performed by associated heterotrimeric G proteins (Gαßγ). Heterotrimeric G protein signal termination is sped up by Gα-directed GTPase-accelerating proteins (GAPs) known as the Regulators of G protein Signaling (RGS proteins). Of this family, RGS21 is highly expressed in lingual epithelial cells and we have shown it acting in vitro to decrease the potency of bitterants on cultured cells. However, constitutive RGS21 loss in mice reduces organismal response to GPCR-mediated tastants-opposite to expectations arising from observed in vitro activity of RGS21 as a GAP and inhibitor of T2R signaling. Here, we show reduced quinine aversion and reduced sucrose preference by mice lacking RGS21 does not result from post-ingestive effects, as taste-salient brief-access tests confirm the reduced bitterant aversion and reduced sweetener preference seen using two-bottle choice testing. Eliminating Rgs21 expression after chemosensory system development, via tamoxifen-induced Cre recombination in eight week-old mice, led to a reduction in quinine aversive behavior that advanced over time, suggesting that RGS21 functions as a negative regulator to sustain stable bitter tastant reception. Consistent with this notion, we observed downregulation of multiple T2R proteins in the lingual tissue of Rgs21-deficient mice. Reduced tastant-mediated responses exhibited by mice lacking Rgs21 expression either since birth or in adulthood has highlighted the potential requirement for a GPCR GAP to maintain the full character of tastant signaling, likely at the level of mitigating receptor downregulation.


Subject(s)
RGS Proteins , Animals , GTP-Binding Proteins , Mice , RGS Proteins/genetics , RGS Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Taste
5.
Anesth Analg ; 132(2): 406-419, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33332902

ABSTRACT

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.


Subject(s)
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
6.
J Psychopharmacol ; 34(12): 1393-1407, 2020 12.
Article in English | MEDLINE | ID: mdl-32842837

ABSTRACT

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.


Subject(s)
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
7.
J Pharmacol Exp Ther ; 371(2): 487-499, 2019 11.
Article in English | MEDLINE | ID: mdl-31492823

ABSTRACT

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.


Subject(s)
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
8.
PLoS One ; 14(8): e0216167, 2019.
Article in English | MEDLINE | ID: mdl-31408461

ABSTRACT

Regulators of G Protein Signaling (RGS proteins) inhibit G protein-coupled receptor (GPCR) signaling by accelerating the GTP hydrolysis rate of activated Gα subunits. Some RGS proteins exert additional signal modulatory functions, and RGS12 is one such protein, with five additional, functional domains: a PDZ domain, a phosphotyrosine-binding domain, two Ras-binding domains, and a Gα·GDP-binding GoLoco motif. RGS12 expression is temporospatially regulated in developing mouse embryos, with notable expression in somites and developing skeletal muscle. We therefore examined whether RGS12 is involved in the skeletal muscle myogenic program. In the adult mouse, RGS12 is expressed in the tibialis anterior (TA) muscle, and its expression is increased early after cardiotoxin-induced injury, suggesting a role in muscle regeneration. Consistent with a potential role in coordinating myogenic signals, RGS12 is also expressed in primary myoblasts; as these cells undergo differentiation and fusion into myotubes, RGS12 protein abundance is reduced. Myoblasts isolated from mice lacking Rgs12 expression have an impaired ability to differentiate into myotubes ex vivo, suggesting that RGS12 may play a role as a modulator/switch for differentiation. We also assessed the muscle regenerative capacity of mice conditionally deficient in skeletal muscle Rgs12 expression (via Pax7-driven Cre recombinase expression), following cardiotoxin-induced damage to the TA muscle. Eight days post-damage, mice lacking RGS12 in skeletal muscle had attenuated repair of muscle fibers. However, when mice lacking skeletal muscle expression of Rgs12 were cross-bred with mdx mice (a model of human Duchenne muscular dystrophy), no increase in muscle degeneration was observed over time. These data support the hypothesis that RGS12 plays a role in coordinating signals during the myogenic program in select circumstances, but loss of the protein may be compensated for within model syndromes of prolonged bouts of muscle damage and repair.


Subject(s)
Cell Differentiation , Cell Proliferation , Muscle, Skeletal/cytology , Muscular Dystrophy, Animal/pathology , Myoblasts/cytology , RGS Proteins/physiology , Animals , Cardiotoxins/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Signal Transduction
9.
Int Arch Allergy Immunol ; 180(1): 72-78, 2019.
Article in English | MEDLINE | ID: mdl-31137020

ABSTRACT

BACKGROUND: Chronic rhinosinusitis (CRS) is a multifaceted disease with a significant genetic component. The importance of taste receptor signaling has recently been highlighted in CRS; single nucleotide polymorphisms (SNPs) of bitter tastant-responsive G-protein-coupled receptors have been linked with CRS and with altered innate immune responses to multiple bacterially derived signals. OBJECTIVE: To determine in CRS the frequency of six SNPs in genes with known bitter tastant signaling function. METHODS: Genomic DNA was isolated from 74 CRS volunteers in West Virginia, and allele frequency was determined and compared with demographically matched data from the 1,000 Genomes database. RESULTS: For two SNPs in a gene recently associated with bitterant signaling regulation, RGS21, there were no associations with CRS (although the frequency of the minor allele of RGS21, rs7528947, was seen to increase with increasing Lund-Mackay CT staging score). Two TAS2R bitter taste receptor gene variants (TAS2R19 rs10772420 and TAS2R38 rs713598), identified in prior CRS genetics studies, were found to have similar associations in this study. CONCLUSION: Unique to our study is the establishment of an association between CRS in this patient population and GNB3 SNP rs5443, a variation in an established G protein component downstream of bitterant receptor signal transduction.


Subject(s)
Heterotrimeric GTP-Binding Proteins/genetics , Polymorphism, Single Nucleotide , Receptors, G-Protein-Coupled/genetics , Rhinitis/diagnosis , Rhinitis/genetics , Sinusitis/diagnosis , Sinusitis/genetics , Adult , Aged , Alleles , Chronic Disease , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Rhinitis/immunology , Risk Factors , Sinusitis/immunology
10.
J Opioid Manag ; 15(2): 103-109, 2019.
Article in English | MEDLINE | ID: mdl-31057342

ABSTRACT

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.


Subject(s)
Opioid-Related Disorders/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Analgesics, Opioid , Genetic Association Studies , Humans , Pilot Projects , West Virginia
11.
Neuropsychopharmacology ; 44(10): 1728-1741, 2019 09.
Article in English | MEDLINE | ID: mdl-31141817

ABSTRACT

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.


Subject(s)
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
12.
Mol Immunol ; 106: 12-21, 2019 02.
Article in English | MEDLINE | ID: mdl-30576947

ABSTRACT

Chemerin receptor (CMKLR1) is a G protein-coupled receptor (GPCR) implicated in macrophage-mediated inflammation and in several forms of human arthritis. Analogous to other GPCR, CMKLR1 is likely regulated by G protein-coupled receptor kinase (GRK) phosphorylation of intracellular domains in an activation-dependent manner, which leads to recruitment and termination of intracellular signaling via desensitization and internalization of the receptor. The ubiquitously expressed GRK family members include GRK2, GRK3, GRK5, and GRK6, but it is unknown which GRK regulates CMKLR1 cellular and signaling functions. Our data show that activation of CMKLR1 by chemerin in primary macrophages leads to signaling and functional outcomes that are regulated by GRK6 and ß-arrestin 2. We show that arrestin recruitment to CMKLR1 following chemerin stimulation is enhanced with co-expression of GRK6. Further, internalization of endogenous CMKLR1, following the addition of chemerin, is decreased in inflammatory macrophages from GRK6- and ß-arrestin 2-deficient mice. These GRK6- and ß-arrestin 2-deficient macrophages display increased migration toward chemerin and altered AKT and Extracellular-signal Related Kinase (ERK) signaling. Our findings show that chemerin-activated CMKLR1 regulation in inflammatory macrophages is largely GRK6 and ß-arrestin mediated, which may impact innate immunity and have therapeutic implications in rheumatic disease.


Subject(s)
Chemokines/immunology , G-Protein-Coupled Receptor Kinases/immunology , Immunity, Innate , Intercellular Signaling Peptides and Proteins/immunology , Macrophages/immunology , Receptors, G-Protein-Coupled/immunology , beta-Arrestin 2/immunology , Animals , Cell Line , Chemokines/genetics , G-Protein-Coupled Receptor Kinases/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/genetics , Macrophages/pathology , Mice , Mice, Knockout , Receptors, Chemokine , Receptors, G-Protein-Coupled/genetics , Rheumatic Diseases/genetics , Rheumatic Diseases/immunology , Rheumatic Diseases/pathology , beta-Arrestin 2/genetics
13.
Drug Alcohol Depend ; 191: 14-24, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30071445

ABSTRACT

BACKGROUND: A subset of cannabis users develop some degree of Cannabis Use Disorder (CUD). Although behavioral therapy has some success in treating CUD, many users relapse, often citing altered sleep, mood, and irritability. Preclinical animal tests of cannabinoid withdrawal focus primarily on somatic-related behaviors precipitated by a cannabinoid receptor antagonist. The goal of the present study was to develop novel cannabinoid withdrawal assays that are either antagonist-precipitated or spontaneously induced by abstinence. METHODS: C57BL/6 J mice were repeatedly administered the phytocannabinoid Δ9-tetrahydrocannabinol (THC; 1, 10 or 50 mg/kg, s.c.), the synthetic cannabinoid receptor agonist JWH-018 (1 mg/kg, s.c.), or vehicle (1:1:18 parts ethanol:Kolliphor EL:saline, s.c.) for 6 days. Withdrawal was precipitated with the cannabinoid receptor inverse agonist rimonabant (3 mg/kg, i.p.) or elicited via abstinence (i.e., spontaneous withdrawal), and putative stress-related behavior was scored. Classic somatic signs of cannabinoid withdrawal were also quantified. RESULTS: Precipitated THC withdrawal significantly increased plasma corticosterone. Precipitated withdrawal from either THC or JWH-018 suppressed marble burying, increased struggling in the tail suspension test, and elicited somatic withdrawal behaviors. The monoacylglycerol lipase inhibitor JZL184 attenuated somatic precipitated withdrawal but had no effect on marble burying or struggling. Spontaneous THC or JWH-018 withdrawal-induced paw tremors, head twitches, and struggled in the tail suspension test after 24-48 h abstinence. JZL184 or THC attenuated these spontaneous withdrawal-induced behaviors. CONCLUSION: Outcomes from tail suspension and marble burying tests reveal that THC withdrawal is multifaceted, eliciting and suppressing behaviors in these tests, in addition to inducing well-documented somatic signs of withdrawal.


Subject(s)
Behavior, Animal/drug effects , Cannabinoid Receptor Agonists/adverse effects , Marijuana Abuse/etiology , Substance Withdrawal Syndrome/etiology , Animals , Benzodioxoles/adverse effects , Dronabinol/adverse effects , Indoles/adverse effects , Male , Mice , Mice, Inbred C57BL , Naphthalenes/adverse effects , Piperidines/adverse effects , Pyrazoles/adverse effects , Rimonabant
14.
Chem Senses ; 43(5): 367-378, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29701767

ABSTRACT

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.


Subject(s)
Food Preferences , RGS Proteins/metabolism , Taste , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , RGS Proteins/deficiency , RGS Proteins/genetics
15.
J Psychopharmacol ; 32(2): 191-203, 2018 02.
Article in English | MEDLINE | ID: mdl-29364035

ABSTRACT

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.


Subject(s)
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
16.
PLoS One ; 12(1): e0170269, 2017.
Article in English | MEDLINE | ID: mdl-28107494

ABSTRACT

The GTPase-accelerating protein, regulator of G-protein signalling 2 (RGS2) reduces signalling from G-protein-coupled receptors (GPCRs) that signal via Gαq. In humans, RGS2 expression is up-regulated by inhaled corticosteroids (ICSs) and long-acting ß2-adrenoceptor agonists (LABAs) such that synergy is produced in combination. This may contribute to the superior clinical efficacy of ICS/LABA therapy in asthma relative to ICS alone. In a murine model of house dust mite (HDM)-induced airways inflammation, three weeks of intranasal HDM (25 µg, 3×/week) reduced lung function and induced granulocytic airways inflammation. Compared to wild type animals, Rgs2-/- mice showed airways hyperresponsiveness (increased airways resistance and reduced compliance). While HDM increased pulmonary inflammation observed on hematoxylin and eosin-stained sections, there was no difference between wild type and Rgs2-/- animals. HDM-induced mucus hypersecretion was also unaffected by RGS2 deficiency. However, inflammatory cell counts in the bronchoalveolar lavage fluid of Rgs2-/- animals were significantly increased (57%) compared to wild type animals and this correlated with increased granulocyte (neutrophil and eosinophil) numbers. Likewise, cytokine and chemokine (IL4, IL17, IL5, LIF, IL6, CSF3, CXCLl, CXCL10 and CXCL11) release was increased by HDM exposure. Compared to wild type, Rgs2-/- animals showed a trend towards increased expression for many cytokines/chemokines, with CCL3, CCL11, CXCL9 and CXCL10 being significantly enhanced. As RGS2 expression was unaffected by HDM exposure, these data indicate that RGS2 exerts tonic bronchoprotection in HDM-induced airways inflammation. Modest anti-inflammatory and anti-remodelling roles for RGS2 are also suggested. If translatable to humans, therapies that maximize RGS2 expression may prove advantageous.


Subject(s)
Bronchitis/physiopathology , Disease Models, Animal , Pneumonia/physiopathology , Pyroglyphidae/immunology , RGS Proteins/physiology , Animals , Bronchitis/immunology , Bronchoalveolar Lavage Fluid , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/immunology , RGS Proteins/genetics
18.
Blood ; 126(24): 2611-20, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26407691

ABSTRACT

Most platelet agonists activate platelets by binding to G-protein-coupled receptors. We have shown previously that a critical node in the G-protein signaling network in platelets is formed by a scaffold protein, spinophilin (SPL), the tyrosine phosphatase, Src homology region 2 domain-containing phosphatase-1 (SHP-1), and the regulator of G-protein signaling family member, RGS18. Here, we asked whether SPL and other RGS18 binding proteins such as 14-3-3γ regulate platelet reactivity by sequestering RGS18 and, if so, how this is accomplished. The results show that, in resting platelets, free RGS18 levels are relatively low, increasing when platelets are activated by thrombin. Free RGS18 levels also rise when platelets are rendered resistant to activation by exposure to prostaglandin I2 (PGI2) or forskolin, both of which increase platelet cyclic adenosine monophosphate (cAMP) levels. However, the mechanism for raising free RGS18 is different in these 2 settings. Whereas thrombin activates SHP-1 and causes dephosphorylation of SPL tyrosine residues, PGI2 and forskolin cause phosphorylation of SPL Ser94 without reducing tyrosine phosphorylation. Substituting alanine for Ser94 blocks cAMP-induced dissociation of the SPL/RGS/SHP-1 complex. Replacing Ser94 with aspartate prevents formation of the complex and produces a loss-of-function phenotype when expressed in mouse platelets. Together with the defect in platelet function we previously observed in SPL(-/-) mice, these data show that (1) regulated sequestration and release of RGS18 by intracellular binding proteins provides a mechanism for coordinating activating and inhibitory signaling networks in platelets, and (2) differential phosphorylation of SPL tyrosine and serine residues provides a key to understanding both.


Subject(s)
Platelet Activation/physiology , RGS Proteins/physiology , Animals , Blood Platelets/drug effects , CHO Cells , Colforsin/pharmacology , Cricetinae , Cricetulus , Cyclic AMP/physiology , Epoprostenol/pharmacology , Fetal Tissue Transplantation , Liver/embryology , Liver Transplantation , Mice , Microfilament Proteins/deficiency , Microfilament Proteins/physiology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/physiology , Phosphorylation , Phosphoserine/metabolism , Phosphotyrosine/metabolism , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Radiation Chimera , Receptors, Thrombin/agonists , Second Messenger Systems/physiology , Signal Transduction/physiology , Thrombin/pharmacology
19.
Mol Cell ; 58(5): 794-803, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25982117

ABSTRACT

G protein-coupled receptors (GPCRs) comprise the largest family of cell surface receptors, regulate a wide range of physiological processes, and are the major targets of pharmaceutical drugs. Canonical signaling from GPCRs is relayed to intracellular effector proteins by trimeric G proteins, composed of α, ß, and γ subunits (Gαßγ). Here, we report that G protein ß subunits (Gß) bind to DDB1 and that Gß2 targets GRK2 for ubiquitylation by the DDB1-CUL4A-ROC1 ubiquitin ligase. Activation of GPCR results in PKA-mediated phosphorylation of DDB1 at Ser645 and its dissociation from Gß2, leading to increase of GRK2 protein. Deletion of Cul4a results in cardiac hypertrophy in male mice that can be partially rescued by the deletion of one Grk2 allele. These results reveal a non-canonical function of the Gß protein as a ubiquitin ligase component and a mechanism of feedback regulation of GPCR signaling.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Protein beta Subunits/physiology , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , DNA-Binding Proteins/metabolism , Female , HEK293 Cells , Humans , Male , Mice, Knockout , Protein Stability , Proteolysis , Rats , Rats, Wistar , Signal Transduction
20.
Biochemistry ; 54(2): 404-12, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25529118

ABSTRACT

Rho family GTPases modulate actin cytoskeleton dynamics by signaling through multiple effectors, including the p21-activated kinases (PAKs). The intestinal parasite Entamoeba histolytica expresses ∼20 Rho family GTPases and seven isoforms of PAK, two of which have been implicated in pathogenesis-related processes such as amoebic motility and invasion and host cell phagocytosis. Here, we describe two previously unstudied PAK isoforms, EhPAK4 and EhPAK5, as highly specific effectors of EhRacC. A structural model based on 2.35 Å X-ray crystallographic data of a complex between EhRacC(Q65L)·GTP and the EhPAK4 p21 binding domain (PBD) reveals a fairly well-conserved Rho/effector interface despite deviation of the PBD α-helix. A structural comparison with EhRho1 in complex with EhFormin1 suggests likely determinants of Rho family GTPase signaling specificity in E. histolytica. These findings suggest a high degree of Rho family GTPase diversity and specificity in the single-cell parasite E. histolytica. Because PAKs regulate pathogenesis-related processes in E. histolytica, they may be valid pharmacologic targets for anti-amoebiasis drugs.


Subject(s)
Entamoeba histolytica/enzymology , Entamoebiasis/parasitology , p21-Activated Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Entamoeba histolytica/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Signal Transduction , p21-Activated Kinases/chemistry , rho GTP-Binding Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...