Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 01.
Article in English | MEDLINE | ID: mdl-38746266

ABSTRACT

Adolescence is a period of increased risk taking, including increased alcohol and drug use. Multiple clinical studies report a positive relationship between adolescent alcohol consumption and risk of developing an alcohol use disorder (AUD) in adulthood. However, few preclinical studies have attempted to tease apart the biological contributions of adolescent alcohol exposure, independent of other social, environmental, and stress factors, and studies that have been conducted show mixed results. Here we use several adolescent voluntary consumption of alcohol models, conducted across three institutes and with two rodent species, to investigate the ramifications of adolescent alcohol consumption on adulthood alcohol consumption in controlled, pre-clinical environments. We consistently demonstrate a lack of increase in adulthood alcohol consumption. This work highlights that risks seen in both human datasets and other murine drinking models may be due to unique social and environmental factors - some of which may be unique to humans. HIGHLIGHTS: Adolescent drinking-in-the-dark (DID) binge drinking does not increase adulthood consumption in a DID model or a two bottle choice model in male and female SST-Cre:Ai9 miceAdolescent pair-housed intermittent access consumption of alcohol does not increase adulthood consumption in an identical adulthood model in male and female C57BL/6J miceAdolescent intermittent access to alcohol does not increase adulthood consumption in male and female Wistar ratsThese complementary datasets across murine models and institutions highlight the need to consider human social factors as well as biological factors.

3.
Mol Psychiatry ; 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38509197

ABSTRACT

Post-traumatic stress disorder (PTSD) and alcohol use disorder (AUD) are often comorbid. Few treatments exist to reduce comorbid PTSD/AUD. Elucidating the mechanisms underlying their comorbidity could reveal new avenues for therapy. Here, we employed a model of comorbid PTSD/AUD, in which rats were subjected to a stressful shock in a familiar context followed by alcohol drinking. We then examined fear overgeneralization and irritability in these rats. Familiar context stress elevated drinking, increased fear overgeneralization, increased alcohol-related aggressive signs, and elevated peripheral stress hormones. We then examined transcripts of stress- and fear-relevant genes in the central amygdala (CeA), a locus that regulates stress-mediated alcohol drinking. Compared with unstressed rats, stressed rats exhibited increases in CeA transcripts for Crh and Fkbp5 and decreases in transcripts for Bdnf and Il18. Levels of Nr3c1 mRNA, which encodes the glucocorticoid receptor, increased in stressed males but decreased in stressed females. Transcripts of Il18 binding protein (Il18bp), Glp-1r, and genes associated with calcitonin gene-related peptide signaling (Calca, Ramp1, Crlr-1, and Iapp) were unaltered. Crh, but not Crhr1, mRNA was increased by stress; thus, we tested whether inhibiting CeA neurons that express corticotropin-releasing factor (CRF) suppress PTSD/AUD-like behaviors. We used Crh-Cre rats that had received a Cre-dependent vector encoding hM4D(Gi), an inhibitory Designer Receptors Exclusively Activated by Designer Drugs. Chemogenetic inhibition of CeA CRF neurons reduced alcohol intake but not fear overgeneralization or irritability-like behaviors. Our findings suggest that CeA CRF modulates PTSD/AUD comorbidity, and inhibiting CRF neural activity is primarily associated with reducing alcohol drinking but not trauma-related behaviors that are associated with PTSD/AUD.

4.
Neurobiol Stress ; 25: 100547, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37547774

ABSTRACT

Impairments in the function of the hypothalamic-pituitary-adrenal (HPA) axis and enhanced glucocorticoid receptor (GR) activity in the central amygdala (CeA) are critical mechanisms in the pathogenesis of alcohol use disorder (AUD). The GR antagonist mifepristone attenuates craving in AUD patients, alcohol consumption in AUD models, and decreases CeA γ-aminobutyric acid (GABA) transmission in alcohol-dependent rats. Previous studies suggest elevated GR activity in the CeA of male alcohol-preferring Marchigian-Sardinian (msP) rats, but its contribution to heightened CeA GABA transmission driving their characteristic post-dependent phenotype is largely unknown. We determined Nr3c1 (the gene encoding GR) gene transcription in the CeA in male and female msP and Wistar rats using in situ hybridization and studied acute effects of mifepristone (10 µM) and its interaction with ethanol (44 mM) on pharmacologically isolated spontaneous inhibitory postsynaptic currents (sIPSCs) and electrically evoked inhibitory postsynaptic potentials (eIPSPs) in the CeA using ex vivo slice electrophysiology. Female rats of both genotypes expressed more CeA GRs than males, suggesting a sexually dimorphic GR regulation of CeA activity. Mifepristone reduced sIPSC frequencies (GABA release) and eIPSP amplitudes in msP rats of both sexes, but not in their Wistar counterparts; however, it did not prevent acute ethanol-induced increase in CeA GABA transmission in male rats. In msP rats, GR regulates CeA GABAergic signaling under basal conditions, indicative of intrinsically active GR. Thus, enhanced GR function in the CeA represents a key mechanism contributing to maladaptive behaviors associated with AUD.

5.
Cells ; 12(15)2023 07 27.
Article in English | MEDLINE | ID: mdl-37566022

ABSTRACT

Alcohol use disorder (AUD) and anxiety disorders are frequently comorbid and share dysregulated neuroimmune-related pathways. Here, we used our established rat model of comorbid post-traumatic stress disorder (PTSD)/AUD to characterize the interleukin 18 (IL-18) system in the central amygdala (CeA). Male and female rats underwent novel (NOV) and familiar (FAM) shock stress, or no stress (unstressed controls; CTL) followed by voluntary alcohol drinking and PTSD-related behaviors, then all received renewed alcohol access prior to the experiments. In situ hybridization revealed that the number of CeA positive cells for Il18 mRNA increased, while for Il18bp decreased in both male and female FAM stressed rats versus CTL. No changes were observed in Il18r1 expression across groups. Ex vivo electrophysiology showed that IL-18 reduced GABAA-mediated miniature inhibitory postsynaptic currents (mIPSCs) frequencies in CTL, suggesting reduced CeA GABA release, regardless of sex. Notably, this presynaptic effect of IL-18 was lost in both NOV and FAM males, while it persisted in NOV and FAM females. IL-18 decreased mIPSC amplitude in CTL female rats, suggesting postsynaptic effects. Overall, our results suggest that stress in rats with alcohol access impacts CeA IL-18-system expression and, in sex-related fashion, IL-18's modulatory function at GABA synapses.


Subject(s)
Alcoholism , Central Amygdaloid Nucleus , Stress Disorders, Post-Traumatic , Rats , Male , Female , Animals , Alcoholism/complications , Central Amygdaloid Nucleus/metabolism , Interleukin-18/metabolism , Ethanol/pharmacology , Alcohol Drinking , gamma-Aminobutyric Acid/metabolism
6.
Br J Pharmacol ; 180(24): 3130-3145, 2023 12.
Article in English | MEDLINE | ID: mdl-37488777

ABSTRACT

BACKGROUND AND PURPOSE: The endocannabinoid (eCB) system plays an important homeostatic role in the regulation of stress circuits and has emerged as a therapeutic target to treat stress disorders and alcohol use disorder (AUD). Extensive research has elucidated a role for the eCB anandamide (AEA), but less is known about 2-arachidonoylglycerol (2-AG) mediated signalling. EXPERIMENTAL APPROACH: We pharmacologically enhanced eCB signalling by inhibiting the 2-AG metabolizing enzyme, monoacylglycerol lipase (MAGL), in male and female Marchigian Sardinian alcohol-preferring (msP) rats, a model of innate alcohol preference and stress hypersensitivity, and in control Wistar rats. We tested the acute effect of the selective MAGL inhibitor MJN110 in alleviating symptoms of alcohol drinking, anxiety, irritability and fear. KEY RESULTS: A single systemic administration of MJN110 increased 2-AG levels in the central amygdala, prelimbic and infralimbic cortex but did not acutely alter alcohol drinking. MAGL inhibition reduced aggressive behaviours in female msPs, and increased defensive behaviours in male msPs, during the irritability test. Moreover, in the novelty-induced hypophagia test, MJN110 selectively enhanced palatable food consumption in females, mitigating stress-induced food suppression. Lastly, msP rats showed increased conditioned fear behaviour compared with Wistar rats, and MJN110 reduced context-associated conditioned fear responses, but not cue-probed fear expression, in male msPs. CONCLUSIONS AND IMPLICATIONS: Acute inhibition of MAGL attenuated some stress-related responses in msP rats but not voluntary alcohol drinking. Our results provide new insights into the sex dimorphism documented in stress-induced responses. Sex-specific eCB-based approaches should be considered in the clinical development of therapeutics.


Subject(s)
Monoacylglycerol Lipases , Monoglycerides , Rats , Male , Female , Animals , Rats, Wistar , Ethanol/pharmacology , Endocannabinoids/metabolism
7.
Neuropsychopharmacology ; 48(8): 1144-1154, 2023 07.
Article in English | MEDLINE | ID: mdl-36396784

ABSTRACT

Post-traumatic stress disorder (PTSD) leads to enhanced alcohol drinking and development of alcohol use disorder (AUD). Identifying shared neural mechanisms might help discover new therapies for PTSD/AUD. Here, we employed a rat model of comorbid PTSD/AUD to evaluate compounds that inhibit FK506-binding protein 51 (FKBP5), a co-chaperone modulator of glucocorticoid receptors implicated in stress-related disorders. Male and female rats received a familiar avoidance-based shock stress followed by voluntary alcohol drinking. We then assessed trauma-related behaviors through sleep bout cycles, hyperarousal, fear overgeneralization, and irritability. To evaluate the role of stress and alcohol history on the sensitivity to FKBP5 inhibitors, in two separate studies, we administered two FKBP5 inhibitors, benztropine (Study 1) or SAFit2 (Study 2). FKBP5 inhibitors were administered on the last alcohol drinking session and prior to each trauma-related behavioral assessment. We also measured plasma corticosterone to assess the actions of FKBP5 inhibitors after familiar shock stress and alcohol drinking. Benztropine reduced alcohol preference in stressed males and females, while aggressive bouts were reduced in benztropine-treated stressed females. During hyperarousal, benztropine reduced several startle response outcomes across stressed males and females. Corticosterone was reduced in benztropine-treated stressed males. The selective FKBP5 inhibitor, SAFit2, reduced alcohol drinking in stressed males but not females, with no differences in irritability. Importantly, SAFit2 decreased fear overgeneralization in stressed males and females. SAFit2 also reduced corticosterone across stressed males and females. Neither FKBP5 inhibitor changed sleep bout structure. These findings indicate that FKBP5 inhibitors modulate stress-related alcohol drinking and partially modulate trauma-related behaviors. This work supports the hypothesis that targeting FKBP5 may alleviate PTSD/AUD comorbidity.


Subject(s)
Alcoholism , Stress Disorders, Post-Traumatic , Male , Rats , Animals , Alcoholism/drug therapy , Alcoholism/epidemiology , Stress Disorders, Post-Traumatic/metabolism , Corticosterone , Tacrolimus Binding Proteins/metabolism , Benztropine/therapeutic use , Alcohol Drinking , Comorbidity
8.
Am J Drug Alcohol Abuse ; 49(3): 321-332, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36206520

ABSTRACT

Background: Although alcohol and nicotine are often used together, the biological consequences of these substances are not well understood. Identifying shared targets will inform cessation pharmacotherapies and provide a deeper understanding of how co-use of alcohol and nicotine impacts health, including biomarkers of stress and inflammation.Objective: We examined the effects of nicotine exposure and withdrawal on alcohol self-administration (SA), stress and inflammatory biomarkers, and a G-protein coupled receptor subunit (Gß) in brain areas associated with drug use.Methods: Male rats were trained to SA alcohol and then received a nicotine pump (n = 7-8 per group). We assessed alcohol intake for 12 days during nicotine exposure and then following pump removal to elicit withdrawal. After the behavioral studies, we assessed plasma leptin, corticosterone, and interleukin-1ß (IL-1ß), and Gß protein expression in the amygdala, nucleus accumbens (NAc), and prefrontal cortex (PFC).Results: Nicotine exposure or withdrawal did not alter alcohol intake (p > .05). Alcohol and nicotine withdrawal elevated corticosterone levels (p = .015) and decreased Gß levels in the PFC (p = .004). In the absence of nicotine, alcohol SA suppressed IL-1ß levels (p = .039). Chronic exposure to nicotine or withdrawal during alcohol SA did not alter leptin levels or Gß expression in the amygdala or NAc (p's > .05).Conclusions: The combination of alcohol SA and nicotine withdrawal produced a persistent increase in stress biomarkers and a suppression in Gß expression in the PFC, providing an important first step toward understanding the common biological mechanisms of alcohol/nicotine misuse.


Subject(s)
Nicotine , Substance Withdrawal Syndrome , Rats , Male , Animals , Nicotine/adverse effects , Leptin/metabolism , Leptin/pharmacology , Leptin/therapeutic use , Corticosterone/metabolism , Corticosterone/pharmacology , Corticosterone/therapeutic use , Rats, Wistar , Substance Withdrawal Syndrome/drug therapy , Prefrontal Cortex , Ethanol/adverse effects
9.
Alcohol Res ; 42(1): 03, 2022.
Article in English | MEDLINE | ID: mdl-35223337

ABSTRACT

PURPOSE: A growing body of evidence has implicated the endocannabinoid (eCB) system in the acute, chronic, and withdrawal effects of alcohol/ethanol on synaptic function. These eCB-mediated synaptic effects may contribute to the development of alcohol use disorder (AUD). Alcohol exposure causes neurobiological alterations similar to those elicited by chronic cannabinoid (CB) exposure. Like alcohol, cannabinoids alter many central processes, such as cognition, locomotion, synaptic transmission, and neurotransmitter release. There is a strong need to elucidate the effects of ethanol on the eCB system in different brain regions to understand the role of eCB signaling in AUD. SEARCH METHODS: For the scope of this review, preclinical studies were identified through queries of the PubMed database. SEARCH RESULTS: This search yielded 459 articles. Clinical studies and papers irrelevant to the topic of this review were excluded. DISCUSSION AND CONCLUSIONS: The endocannabinoid system includes, but is not limited to, cannabinoid receptors 1 (CB1), among the most abundantly expressed neuronal receptors in the brain; cannabinoid receptors 2 (CB2); and endogenously formed CB1 ligands, including arachidonoylethanolamide (AEA; anandamide), and 2-arachidonoylglycerol (2-AG). The development of specific CB1 agonists, such as WIN 55,212-2 (WIN), and antagonists, such as SR 141716A (rimonabant), provide powerful pharmacological tools for eCB research. Alcohol exposure has brain region-specific effects on the eCB system, including altering the synthesis of endocannabinoids (e.g., AEA, 2-AG), the synthesis of their precursors, and the density and coupling efficacy of CB1. These alcohol-induced alterations of the eCB system have subsequent effects on synaptic function including neuronal excitability and postsynaptic conductance. This review will provide a comprehensive evaluation of the current literature on the synaptic interactions of alcohol exposure and eCB signaling systems, with an emphasis on molecular and physiological synaptic effects of alcohol on the eCB system. A limited volume of studies has focused on the underlying interactions of alcohol and the eCB system at the synaptic level in the brain. Thus, the data on synaptic interactions are sparse, and future research addressing these interactions is much needed.


Subject(s)
Alcoholism , Cannabinoids , Cannabinoids/pharmacology , Endocannabinoids/pharmacology , Ethanol/pharmacology , Humans , Receptors, Cannabinoid , Synaptic Transmission
10.
Int J Mol Sci ; 22(8)2021 Apr 17.
Article in English | MEDLINE | ID: mdl-33920737

ABSTRACT

Alcoholism is a chronically relapsing disorder characterized by high alcohol intake and a negative emotional state during abstinence, which contributes to excessive drinking and susceptibility to relapse. Stress, dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and alterations in glucocorticoid receptor (GR) function have been linked to transition from recreational consumption to alcohol use disorder (AUD). Here, we investigated the effect of pharmacological antagonisms of GR on alcohol self-administration (SA) using male and female Wistar and Marchigian Sardinian alcohol-preferring (msP) rats, a rodent line genetically selected for excessive alcohol drinking and highly sensitive to stress. Animals were trained to self-administer 10% (v/v) alcohol. Once a stable alcohol SA baseline was reached, we tested the effect of the GR antagonists mifepristone (0.0, 10, 30 and 60 mg/kg; i.p.) and CORT113176 (0.0, 10, 30 and 60 mg/kg) on alcohol SA. To evaluate whether the effects of the two compounds were specific for alcohol, the two drugs were tested on a similar saccharin SA regimen. Finally, basal blood corticosterone (CORT) levels before and after alcohol SA were determined. Systemic injection with mifepristone dose-dependently reduced alcohol SA in male and female Wistars but not in msPs. Administration of CORT113176 decreased alcohol SA in male and female Wistars as well as in female msPs but not in male msP rats. At the highest dose, mifepristone also reduced saccharin SA in male Wistars and female msPs, suggesting the occurrence of some nonspecific effects at 60 mg/kg of the drug. Similarly, the highest dose of CORT113176 (60 mg/kg) decreased saccharin intake in male Wistars. Analysis of CORT levels revealed that females of both rat lines had higher blood levels of CORT compared to males. Alcohol consumption reduced CORT in females but not in males. Overall, these findings indicate that selective blockade of GR selectively reduces alcohol SA, and genetically selected msP rats are less sensitive to this pharmacological manipulation compared to heterogeneous Wistars. Moreover, results suggest sex differences in response to GR antagonism and the ability of alcohol to regulate GR transmission.


Subject(s)
Alcoholism/drug therapy , Hormone Antagonists/therapeutic use , Receptors, Glucocorticoid/antagonists & inhibitors , Alcoholism/genetics , Animals , Female , Hormone Antagonists/pharmacology , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Male , Mifepristone/pharmacology , Mifepristone/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Rats , Rats, Wistar
11.
Int J Mol Sci ; 22(6)2021 Mar 18.
Article in English | MEDLINE | ID: mdl-33803557

ABSTRACT

Marchigian Sardinian alcohol-preferring (msP) rats serve as a unique model of heightened alcohol preference and anxiety disorders. Their innate enhanced stress and poor stress-coping strategies are driven by a genetic polymorphism of the corticotropin-releasing factor receptor 1 (CRF1) in brain areas involved in glucocorticoid signaling. The activation of glucocorticoid receptors (GRs) regulates the stress response, making GRs a candidate target to treat stress and anxiety. Here, we examined whether mifepristone, a GR antagonist known to reduce alcohol drinking in dependent rats, decreases innate symptoms of anxiety in msPs. Male and female msPs were compared to non-selected Wistar counterparts across three separate behavioral tests. We assessed anxiety-like behavior via the novelty-induced hypophagia (NIH) assay. Since sleep disturbances and hyperarousal are common features of stress-related disorders, we measured sleeping patterns using the comprehensive lab monitoring system (CLAMS) and stress sensitivity using acoustic startle measures. Rats received an acute administration of vehicle or mifepristone (60 mg/kg) 90 min prior to testing on NIH, acoustic startle response, and CLAMS. Our results revealed that both male and female msPs display greater anxiety-like behaviors as well as enhanced acoustic startle responses compared to Wistar counterparts. Male msPs also displayed reduced sleeping bout duration versus Wistars, and female msPs displayed greater acoustic startle responses versus male msPs. Importantly, the enhanced anxiety-like behavior and startle responses were not reduced by mifepristone. Together, these findings suggest that increased expression of stress-related behaviors in msPs are not solely mediated by acute activation of GRs.


Subject(s)
Anxiety/pathology , Behavior, Animal , Mifepristone/pharmacology , Receptors, Glucocorticoid/antagonists & inhibitors , Animals , Anxiety/complications , Anxiety/physiopathology , Arousal/drug effects , Behavior, Animal/drug effects , Female , Male , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Sleep Wake Disorders/complications , Sleep Wake Disorders/physiopathology
12.
Neuropsychopharmacology ; 45(8): 1330-1338, 2020 07.
Article in English | MEDLINE | ID: mdl-32375160

ABSTRACT

The endocannabinoid system is a key regulator of the response to psychological stress. Inhibitors of monoacylglycerol lipase (MGL), the enzyme that deactivates the endocannabinoid 2-arachidonoyl-sn-glycerol (2-AG), exert anxiolytic-like effects in rodent models via 2-AG-dependent activation of CB1 cannabinoid receptors. In the present study, we examined whether the MGL inhibitor JZL184 might modulate persistent predator-induced fear in rats, a model that captures features of human post-traumatic stress disorder. Exposure to 2,5-dihydro-2,4,5-trimethylthiazoline (TMT), a volatile chemical that is innately aversive to some rodent species, produced in male rats a long-lasting anxiety-like state that was measured 7 days later in the elevated plus maze test. Systemic administration of JZL184 [4, 8 and 16 mg/kg, intraperitoneal (IP)] 4 h before testing caused dose-dependent inhibition of MGL activity and elevation of 2-AG content in brain tissue. Concomitantly, the inhibitor suppressed TMT-induced fear behaviors with a median effective dose (ED50) of 4 mg/kg. A similar behavioral response was observed with another MGL inhibitor, KML29 (4 and 16 mg/kg, IP). Surprisingly, the effect of JZL184 was prevented by co-administration of the CB2 inverse agonist AM630 (5 mg/kg, IP), but not the CB1 inverse agonist rimonabant (1 mg/kg, IP). Supporting mediation of the response by CB2 receptors, the CB2 agonist JWH133 (0.3, 1 and 3 mg/kg, IP) also produced anxiolytic-like effects in TMT-stressed rats, which were suppressed by AM630. Notably, (i) JWH133 was behaviorally ineffective in animals that had no prior experience with TMT; and (ii) CB2 mRNA levels in rat prefrontal cortex were elevated 7 days after exposure to the aversive odorant. The results suggest that JZL184 attenuates the behavioral consequences of predator stress through a mechanism that requires 2-AG-mediated activation of CB2 receptors, whose transcription may be induced by the stress itself.


Subject(s)
Anti-Anxiety Agents , Cannabinoids , Animals , Anti-Anxiety Agents/pharmacology , Arachidonic Acids/pharmacology , Cannabinoids/pharmacology , Endocannabinoids , Fear , Male , Monoacylglycerol Lipases , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1 , Receptor, Cannabinoid, CB2 , Receptors, Cannabinoid
13.
J Pharmacol Exp Ther ; 374(1): 151-160, 2020 07.
Article in English | MEDLINE | ID: mdl-32345621

ABSTRACT

We investigated the pharmacokinetic properties of Δ9-tetrahydrocannabinol (Δ9-THC), the main psychoactive constituent of cannabis, in adolescent and adult male mice. The drug was administered at logarithmically ascending doses (0.5, 1.6, and 5 mg/kg, i.p.) to pubertal adolescent (37-day-old) and adult (70-day-old) mice. Δ9-THC and its first-pass metabolites-11-hydroxy-Δ9-THC and 11-nor-9-carboxy-Δ9-THC (11-COOH-THC)-were quantified in plasma, brain, and white adipose tissue (WAT) using a validated isotope-dilution liquid chromatography/tandem mass spectrometry assay. Δ9-THC (5 mg/kg) reached 50% higher circulating concentration in adolescent mice than in adult mice. A similar age-dependent difference was observed in WAT. Conversely, 40%-60% lower brain concentrations and brain-to-plasma ratios for Δ9-THC and 50%-70% higher brain concentrations for Δ9-THC metabolites were measured in adolescent animals relative to adult animals. Liver microsomes from adolescent mice converted Δ9-THC into 11-COOH-THC twice as fast as adult microsomes. Moreover, the brains of adolescent mice contained higher mRNA levels of the multidrug transporter breast cancer resistance protein, which may extrude Δ9-THC from the brain, and higher mRNA levels of claudin-5, a protein that contributes to blood-brain barrier integrity. Finally, administration of Δ9-THC (5 mg/kg) reduced spontaneous locomotor activity in adult, but not adolescent, animals. The results reveal the existence of multiple differences in the distribution and metabolism of Δ9-THC between adolescent and adult male mice, which might influence the pharmacological response to the drug. SIGNIFICANCE STATEMENT: Animal studies suggest that adolescent exposure to Δ9-tetrahydrocannabinol (Δ9-THC), the intoxicating constituent of cannabis, causes persistent changes in brain function. These studies generally overlook the impact that age-dependent changes in the distribution and metabolism of the drug might exert on its pharmacological effects. This report provides a comparative analysis of the pharmacokinetic properties of Δ9-THC in adolescent and adult male mice and outlines multiple functionally significant dissimilarities in the distribution and metabolism of Δ9-THC between these two age groups.


Subject(s)
Dronabinol/pharmacokinetics , ATP-Binding Cassette Transporters/genetics , Aging/metabolism , Animals , Claudin-5/genetics , Dronabinol/blood , Gene Expression Regulation , Male , Mice , RNA, Messenger/genetics , Tissue Distribution
14.
J Pharm Pharmacol ; 71(12): 1762-1773, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31579946

ABSTRACT

OBJECTIVES: URB937, a peripheral fatty acid amide hydrolase (FAAH) inhibitor, exerts profound analgesic effects in animal models. We examined, in rats, (1) the pharmacokinetic profile of oral URB937; (2) the compound's ability to elevate levels of the representative FAAH substrate, oleoylethanolamide (OEA); and (3) the compound's tolerability after oral administration. METHODS: We developed a liquid chromatography/tandem mass spectrometry (LC/MS-MS) method to measure URB937 and used a pre-existing LC/MS-MS assay to quantify OEA. FAAH activity was measured using a radioactive substrate. The tolerability of single or repeated (once daily for 2 weeks) oral administration of supramaximal doses of URB937 (100, 300, 1000 mg/kg) was assessed by monitoring food intake, water intake and body weight, followed by post-mortem evaluation of organ structure. KEY FINDINGS: URB937 was orally available in male rats (F = 36%), but remained undetectable in brain when administered at doses that maximally inhibit FAAH activity and elevate OEA in plasma and liver. Acute and subchronic treatment with high doses of URB937 was well-tolerated and resulted in FAAH inhibition in brain. CONCLUSIONS: Pain remains a major unmet medical need. The favourable pharmacokinetic and pharmacodynamic properties of URB937, along with its tolerability, encourage further development studies on this compound.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Cannabinoids/administration & dosage , Enzyme Inhibitors/administration & dosage , Administration, Oral , Animals , Brain/drug effects , Brain/metabolism , Cannabinoids/pharmacokinetics , Cannabinoids/toxicity , Chromatography, Liquid , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Female , Male , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Tissue Distribution
15.
Int J Mol Sci ; 20(16)2019 Aug 17.
Article in English | MEDLINE | ID: mdl-31426457

ABSTRACT

Previous studies have shown that the sphingolipid-derived mediator sphingosine-1-phosphate (S1P) reduces food intake by activating G protein-coupled S1P receptor-1 (S1PR1) in the hypothalamus. Here, we examined whether feeding regulates hypothalamic mobilization of S1P and other sphingolipid-derived messengers. We prepared lipid extracts from the hypothalamus of C57Bl6/J male mice subjected to one of four conditions: free feeding, 12 h fasting, and 1 h or 6 h refeeding. Liquid chromatography/tandem mass spectrometry was used to quantify various sphingolipid species, including sphinganine (SA), sphingosine (SO), and their bioactive derivatives SA-1-phosphate (SA1P) and S1P. In parallel experiments, transcription of S1PR1 (encoded in mice by the S1pr1 gene) and of key genes of sphingolipid metabolism (Sptlc2, Lass1, Sphk1, Sphk2) was measured by RT-PCR. Feeding increased levels of S1P (in pmol-mg-1 of wet tissue) and SA1P. This response was accompanied by parallel changes in SA and dihydroceramide (d18:0/18:0), and was partially (SA1P) or completely (S1P) reversed by fasting. No such effects were observed with other sphingolipid species targeted by our analysis. Feeding also increased transcription of Sptlc2, Lass1, Sphk2, and S1pr1. Feeding stimulates mobilization of endogenous S1PR1 agonists S1P and SA1P in mouse hypothalamus, via a mechanism that involves transcriptional up-regulation of de novo sphingolipid biosynthesis. The results support a role for sphingolipid-mediated signaling in the central control of energy balance.


Subject(s)
Eating , Hypothalamus/metabolism , Lysophospholipids/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Gene Expression Regulation , Hypothalamus/physiology , Male , Mice , Mice, Inbred C57BL , Sphingolipids/metabolism , Sphingosine/metabolism
16.
Cannabis Cannabinoid Res ; 4(2): 110-123, 2019.
Article in English | MEDLINE | ID: mdl-31236476

ABSTRACT

Introduction: Few animal studies have evaluated the pharmacological effects of Δ9-tetrahydrocannabinol (THC) in relation to its pharmacokinetic properties. Understanding this relationship is essential, however, if comparisons are to be drawn across conditions-such as sex, age, and route of administration-which are associated with variations in the absorption, metabolism, and distribution of THC. As a first step toward addressing this gap, in this report, we describe a rapid, sensitive, and accurate method for the quantification of THC and its main oxidative metabolites, and apply it to representative rodent tissues. Materials and Methods: The sample workup procedure consisted of two steps: bulk protein precipitation with cold acetonitrile (ACN) followed by phospholipid removal by elution through Captiva-Enhanced Matrix Removal cartridges. The liquid chromatography/tandem mass spectrometry (LC/MS-MS) protocol utilized a commercially available C18 reversed-phase column and a simple methanol/water gradient system. The new method was validated following Food and Drug Administration (FDA) guidelines, and was applied to the quantification of THC and its main oxidative metabolites-11-hydroxy-Δ9-tetrahydrocannabinol (11-OH-THC) and 11-nor-9-carboxy-Δ9-tetrahydrocannabinol (11-COOH-THC)-in plasma and brain of mice treated with a single intraperitoneal dose of THC (10 mg/kg). Results: ACN precipitation and column elution effectively depleted matrix constituents-most notably choline-containing phospholipids-which are known to interfere with THC analysis, with average recovery values of >85% for plasma and >80% for brain. The LC conditions yielded baseline separation of all analytes in a total run time of 7 min (including re-equilibration). The 10-point calibration curves showed excellent linearity (R 2>0.99) over a wide range of concentrations (1-1000 pmol/100 µL). Lowest limit of quantification was 2 pmol/100 µL for all analytes, and lowest limits of detection were 0.5 pmol/100 µL for THC and 11-OH-THC, and 1 pmol/100 µL for 11-COOH-THC. Intraday and interday accuracy and precision values were within the FDA-recommended range (±15% of nominal concentration). An application of the method to adult male mice is presented. Conclusions: We present a fast and sensitive method for the analysis of THC, which should facilitate studies aimed at linking the pharmacokinetics and pharmacodynamics of this compound in animal models.

17.
Aging (Albany NY) ; 11(1): 73-88, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30620722

ABSTRACT

Circulating ceramide levels are abnormally elevated in age-dependent pathologies such as cardiovascular diseases, obesity and Alzheimer's disease. Nevertheless, the potential impact of age on plasma ceramide levels has not yet been systematically examined. In the present study, we quantified a focused panel of plasma ceramides and dihydroceramides in a cohort of 164 subjects (84 women) 19 to 80 years of age. After adjusting for potential confounders, multivariable linear regression analysis revealed a positive association between age and ceramide (d18:1/24:0) (ß (SE) = 5.67 (2.38); p = .0198) and ceramide (d18:1/24:1) (ß (SE) = 2.88 (.61); p < .0001) in women, and between age and ceramide (d18:1/24:1) in men (ß (SE) = 1.86 (.77); p = .0179). In women of all ages, but not men, plasma ceramide (d18:1/24:1) was negatively correlated with plasma estradiol (r = -0.294; p = .007). Finally, in vitro experiments in human cancer cells expressing estrogen receptors showed that incubation with estradiol (10 nM, 24 h) significantly decreased ceramide accumulation. Together, the results suggest that aging is associated with an increase in circulating ceramide levels, which in post-menopausal women is at least partially associated with lower estradiol levels.


Subject(s)
Aging/blood , Ceramides/blood , Adult , Aged , Aging/physiology , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Postmenopause , Young Adult
18.
Cell Metab ; 29(1): 91-102.e5, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30318340

ABSTRACT

The conversion of lipolysis-derived fatty acids into ketone bodies (ketogenesis) is a crucial metabolic adaptation to prolonged periods of food scarcity. The process occurs primarily in liver mitochondria and is initiated by fatty-acid-mediated stimulation of the ligand-operated transcription factor, peroxisome proliferator-activated receptor-α (PPAR-α). Here, we present evidence that mast cells contribute to the control of fasting-induced ketogenesis via a paracrine mechanism that involves secretion of histamine into the hepatic portal circulation, stimulation of liver H1 receptors, and local biosynthesis of the high-affinity PPAR-α agonist, oleoylethanolamide (OEA). Genetic or pharmacological interventions that disable any one of these events, including mast cell elimination, deletion of histamine- or OEA-synthesizing enzymes, and H1 blockade, blunt ketogenesis without affecting lipolysis. The results reveal an unexpected role for mast cells in the regulation of systemic fatty-acid homeostasis, and suggest that OEA may act in concert with lipolysis-derived fatty acids to activate liver PPAR-α and promote ketogenesis.


Subject(s)
Endocannabinoids/metabolism , Histamine/physiology , Ketone Bodies/biosynthesis , Liver/metabolism , Mast Cells/metabolism , Oleic Acids/metabolism , PPAR alpha/metabolism , Animals , Hep G2 Cells , Humans , Liver/cytology , Male , Mast Cells/cytology , Mice , Mice, Inbred C57BL , Receptors, Histamine H1/metabolism
19.
Psychopharmacology (Berl) ; 235(11): 3211-3221, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30251159

ABSTRACT

RATIONALE: The endocannabinoid neurotransmitter, anandamide, has been implicated in the central modulation of stress responses. Previous animal experiments have shown that inhibitors of the anandamide-degrading enzyme, fatty acid amide hydrolase (FAAH), enhance the ability to cope with acute and chronic stress. OBJECTIVES: Here, we investigated the effects of the globally active FAAH inhibitor URB597 in a rat model of predator stress-induced long-term anxiety. RESULTS: Rats exposed to 2,5-dihydro-2,4,5-trimethylthiazoline (TMT), a chemical constituent of fox feces, developed a persistent anxiety-like state, which was assessed 7 days after exposure using the elevated plus maze (EPM) test. Systemic administration of URB597 [0.03-0.1-0.3 mg/kg, intraperitoneal (ip)] 2 h before testing suppressed TMT-induced behaviors with a median effective dose (IC50) of 0.075 mg/kg. This effect was strongly correlated with inhibition of brain FAAH activity (r2 = 1.0) and was accompanied by increased brain levels of three FAAH substrates: the endocannabinoid anandamide and the endogenous peroxisome proliferator-activated receptor-α (PPAR-α) agonists, oleoylethanolamide (OEA), and palmitoylethanolamide (PEA). The anxiolytic-like effects of URB597 were blocked by co-administration of the CB1 receptor antagonist rimonabant (1 mg/kg, ip), but not of the PPAR-α antagonist GW6471 (1 mg/kg, ip). Finally, when administered 18 h after TMT exposure (i.e., 6 days before the EPM test), URB597 (0.3 mg/kg, ip) prevented the consolidation of anxiety-like behavior in a CB1-dependent manner. CONCLUSIONS: The results support the hypothesis that anandamide-mediated signaling at CB1 receptors serves an important regulatory function in the stress response, and confirm that FAAH inhibition may offer a potential therapeutic strategy for post-traumatic stress disorder.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anxiety/drug therapy , Anxiety/psychology , Benzamides/therapeutic use , Carbamates/therapeutic use , Disease Models, Animal , Amidohydrolases/metabolism , Animals , Anxiety/metabolism , Benzamides/pharmacology , Brain/drug effects , Brain/metabolism , Cannabinoid Receptor Agonists/pharmacology , Carbamates/pharmacology , Dose-Response Relationship, Drug , Foxes , Male , PPAR alpha/agonists , PPAR alpha/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Time Factors
20.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(12): 1502-1511, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28855145

ABSTRACT

Sphingolipids have been implicated in age-related neurodegeneration. Previous studies have reported elevated ceramide levels in the brain of old rodents, but a systematic investigation of the impact of age on brain sphingolipid metabolism is still lacking. Here we quantified 17 key sphingolipid species in the hippocampus of young (3months), middle-aged (12months) and old (21months) male and female mice. Lipids were extracted and quantified by liquid chromatography/mass spectrometry; transcription of enzymes involved in sphingolipid biosynthesis was evaluated by qPCR. Age-dependent changes of multiple sphingolipid species - including ceramide (d18:1/18:0), sphingomyelin (d34:1), hexosylceramide (d18:1/16:0), ceramide (d18:1/24:0) - were found in mice of both sexes. Moreover, sex-dependent changes were seen with hexosylceramide (d18:1/18:0), ceramide (d18:1/22:0), sphingomyelin (d36:1) and sphingomyelin (d42:1). Importantly, an age-dependent accumulation of sphingolipids containing nervonic acid (24:1) was observed in 21month-old male (p=0.04) and female mice (p<0.0001). Consistent with this increase, transcription of the nervonic acid-synthesizing enzyme, stearoyl-CoA desaturase (Scd1 and Scd2), was upregulated in 21month-old female mice (Scd1 p=0.006; Scd2 p=0.009); a similar trend was observed in males (Scd1 p=0.07). In conclusion, the results suggest that aging is associated with profound sex-dependent and -independent changes in hippocampal sphingolipid profile. The results also highlight the need to examine the contribution of sphingolipids, and particularly of those containing nervonic acid, in normal and pathological brain aging.


Subject(s)
Aging/metabolism , Fatty Acids, Monounsaturated/metabolism , Hippocampus/metabolism , Sphingolipids/metabolism , Animals , Female , Male , Mice , Stearoyl-CoA Desaturase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...