1
|
Chen Y, Xiao T, Kimbrough A. Escalation of intravenous fentanyl self-administration and assessment of withdrawal behavior in male and female mice. Psychopharmacology (Berl) 2025; 242:1419-1435. [PMID: 39730840 DOI: 10.1007/s00213-024-06739-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
RATIONALE The rise in overdose deaths from synthetic opioids, especially fentanyl, necessitates the development of preclinical models to study fentanyl use disorder (FUD). While there has been progress with rodent models, additional translationally relevant models are needed to examine excessive fentanyl intake and withdrawal signs. OBJECTIVE The current study aimed to develop a translationally relevant preclinical mouse model of FUD by employing chronic intravenous fentanyl self-administration (IVSA). METHODS The study performed intravenous self-administration (IVSA) of fentanyl in male and female C57BL/6J mice for 14 days. Mechanical pain sensitivity during withdrawal was assessed using the von Frey test. Anxiety-like behavior was evaluated via the open field test one week into abstinence, and drug seeking behavior after extended abstinence was assessed at four weeks abstinence. RESULTS Both male and female mice demonstrated a significant escalation in fentanyl intake over the 14 days of self-administration, with significant front-loading observed in the final days of self-administration. Mice showed increased mechanical pain sensitivity at 36 and 48hours withdrawal from fentanyl. At 1-week abstinence from fentanyl, mice exhibited increased anxiety-like behavior compared to naive mice. Four weeks into abstinence from fentanyl, mice maintained lever-pressing behavior on the previous reward-associated active lever, with significantly higher active lever pressing compared to inactive lever pressing. CONCLUSIONS The study establishes a translationally relevant mouse model of IVSA of fentanyl, effectively encapsulating critical aspects of FUD, including escalation of drug intake, front-loading behavior, withdrawal signs, and drug-seeking behavior into extended abstinence. This model offers a robust basis for further exploration into behavioral and neurobiological mechanisms involved in fentanyl dependence and potential therapeutic strategies.
Collapse
Affiliation(s)
- Yueyi Chen
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47904, USA
| | - Tiange Xiao
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47904, USA
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47904, USA.
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, 47904, USA.
| |
Collapse
|
2
|
Boorman DC, Rehal SK, Fazili M, Martin LJ. Sex and Strain Differences in Analgesic and Hyperlocomotor Effects of Morphine and μ-Opioid Receptor Expression in Mice. J Neurosci Res 2025; 103:e70039. [PMID: 40247818 PMCID: PMC12006896 DOI: 10.1002/jnr.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/05/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025]
Abstract
Sex and gender differences in the analgesic efficacy and side effects of opioids have been widely reported, but their underlying neurobiological mechanisms remain poorly understood. Preclinical animal models are essential tools for investigating these differences and providing insights into the neurobiology of opioid effects. Although studies in rats have revealed sex-specific effects of opioids, the sex-dependent behavioral profiles of opioids in mice, particularly across strains, remain largely unexplored. In this study, we characterized sex and strain differences in the antinociceptive and hyperlocomotor effects of morphine in the two most widely used mouse strains-CD1 and C57BL/6-and quantified regional expression of the μ-opioid receptor (MOR) in key brain and spinal cord regions. Both strains exhibited clear, dose-dependent antinociceptive and hyperlocomotor responses to morphine. While no significant sex or strain differences were observed in antinociceptive effects, C57BL/6 mice displayed significantly greater hyperlocomotor activity than CD1 mice. Western blot analyses revealed strain-specific MOR expression, with CD1 mice showing higher spinal cord and periaqueductal gray MOR levels, particularly in females, while C57BL/6 mice exhibited elevated MOR expression in the caudoputamen. Morphine treatment increased spinal MOR expression in CD1 mice but not C57BL/6, suggesting strain-dependent regulation of MOR. These findings highlight strain-specific behavioral and molecular responses to morphine, emphasizing the importance of strain and sex considerations in preclinical opioid research.
Collapse
Affiliation(s)
- Damien C. Boorman
- Department of PsychologyThe University of Toronto MississaugaMississaugaOntarioCanada
| | - Simran K. Rehal
- Department of Cell and Systems BiologyUniversity of TorontoTorontoOntarioCanada
| | - Maryam Fazili
- Department of PsychologyThe University of Toronto MississaugaMississaugaOntarioCanada
| | - Loren J. Martin
- Department of PsychologyThe University of Toronto MississaugaMississaugaOntarioCanada
- Department of Cell and Systems BiologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
3
|
Perše M. Animal Models of Human Pathology: Revision, Relevance and Refinements. Biomedicines 2024; 12:2418. [PMID: 39594985 PMCID: PMC11592039 DOI: 10.3390/biomedicines12112418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Animal Models of Human Pathology [...].
Collapse
Affiliation(s)
- Martina Perše
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
4
|
Anbalagan S. "Blind men and an elephant": The need for animals in research, drug safety studies, and understanding civilizational diseases. Animal Model Exp Med 2023; 6:627-633. [PMID: 38062666 PMCID: PMC10757217 DOI: 10.1002/ame2.12364] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/26/2023] [Indexed: 12/31/2023] Open
Abstract
Animal-based research and drug safety studies are essential to understanding the mysteries of nature and the long-term survival of humans. Due to the rapid increase in the global human population, conflict- and economically driven human migration, tourism-related activities, densely populated metropolitan areas, and local policies, humans will be affected by a multitude of novel disease-causing microorganisms and civilizational diseases. Despite disparities among countries, recent and planned changes in regulations concerning animal research and drug safety studies could have detrimental effects on both the animal research community and nations lacking sufficient social support systems. Based on existing scientific literature, I argue that we need animal research encompassing aspects such as animal development, behavior, drug safety studies, and for the understanding of future civilizational diseases. Depending on the nature of the research questions and local challenges, a suitable animal model organism should be made mandatory.
Collapse
Affiliation(s)
- Savani Anbalagan
- Faculty of Biology, Institute of Molecular Biology and BiotechnologyAdam Mickiewicz University in PoznańPoznańPoland
| |
Collapse
|
5
|
Durgin CJ, Huhn AS, Bergeria CL, Finan PH, Campbell CM, Antoine DG, Dunn KE. Within subject, double blind, examination of opioid sensitivity in participant-reported, observed, physiologic, and analgesic outcomes. DRUG AND ALCOHOL DEPENDENCE REPORTS 2023; 8:100188. [PMID: 37731966 PMCID: PMC10507188 DOI: 10.1016/j.dadr.2023.100188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/22/2023]
Abstract
Background Inter-individual differences in opioid sensitivity may underlie different opioid risk profiles but have often been researched in persons who have current or past opioid use disorder or physical dependence. This study examined how opioid sensitivity manifests across various assessments of opioid effects in a primarily opioid-naïve population. Procedures Data were harmonized from two within-subject, double-blind trials wherein healthy participants (N = 123) received placebo and 4 mg oral hydromorphone. Demographics, self-report ratings, observer ratings, physiological, and cold pressor measures were collected. Participants were categorized as being responsive or nonresponsive to the opioid dose tested and compared using mixed-models, Pearson product correlations, and paired t-tests. Findings Participants were 49.6% female, mean 33.0 (SD=9.3) years old, and 44.7% Black/African American and 41.5% White, with 89.4% reporting no prior exposure to opioids. Within-subject sensitivity to opioids varied depending on the measure. One in five participants did not respond subjectively to the 4 mg hydromorphone dose based on their "Drug Effects" rating. Persons who were responsive showed more evidence of drug-dependent effects than did persons who were not responsive on ratings of Bad Effects (p= .03), feeling High (p= .01), Nausea (p= .03), pupil diameter (p< 0.01), and on the circular lights task (p< 0.001). Conclusions This study provides initial evidence that the experience of opioids may be domain specific. Data suggest potentially clinically meaningful differences exist regarding opioid response patterns, evident following one dose among opioid inexperienced individuals.
Collapse
Affiliation(s)
- Caitlyn J. Durgin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr, Baltimore MD 21224, USA
| | - Andrew S. Huhn
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr, Baltimore MD 21224, USA
| | - Cecilia L. Bergeria
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr, Baltimore MD 21224, USA
| | - Patrick H. Finan
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Claudia M. Campbell
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr, Baltimore MD 21224, USA
| | - Denis G. Antoine
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr, Baltimore MD 21224, USA
| | - Kelly E. Dunn
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr, Baltimore MD 21224, USA
| |
Collapse
|
6
|
Elsilä LV, Harkki J, Enberg E, Martti A, Linden AM, Korpi ER. Effects of acute lysergic acid diethylamide on intermittent ethanol and sucrose drinking and intracranial self-stimulation in C57BL/6 mice. J Psychopharmacol 2022; 36:860-874. [PMID: 35695174 PMCID: PMC9247434 DOI: 10.1177/02698811221104641] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Psychedelics, like lysergic acid diethylamide (LSD), are again being studied as potential therapies for many neuropsychiatric disorders, including addictions. At the same time, the acute effects of psychedelics on rewarding behaviours have been scarcely studied. AIMS The current study aimed to clarify if LSD decreases binge-like ethanol drinking in mice, and whether the observed acute effects on ethanol consumption are generalizable to a natural reinforcer, sucrose, and if the effects resulted from aversive or reward-attenuating effects caused by LSD. METHODS The effects of acute LSD were examined using 2-bottle choice intermittent ethanol (20%) and sucrose drinking (10%), discrete-trial current-intensity threshold method of intracranial self-stimulation and short-term feeding behaviour assay in C57BL/6 male mice. RESULTS The results showed that acute 0.1 mg/kg, but not 0.05 mg/kg, dose (i.p.) of LSD reduced 2-h intermittent ethanol drinking transiently without any prolonged effects. No effects were seen in intermittent 2-h sucrose drinking. The tested LSD doses had neither effect on the intracranial self-stimulation current-intensity thresholds, nor did LSD affect the threshold-lowering, or rewarding, effects of simultaneous amphetamine treatment. Furthermore, LSD had small, acute diminishing effects on 2-h food and water intake. CONCLUSIONS Based on these results, LSD decreases binge-like ethanol drinking in mice, but only acutely. This effect is not likely to stem from reward-attenuating effects but could be in part due to reduced consummatory behaviour.
Collapse
Affiliation(s)
- Lauri V Elsilä
- Lauri V Elsilä, Department of
Pharmacology, Faculty of Medicine, University of Helsinki, P.O. Box 63
(Haartmaninkatu 8), Biomedicum Helsinki, Helsinki FI-00014, Finland.
| | | | | | | | | | | |
Collapse
|
7
|
Barbosa-Méndez S, Matus-Ortega M, Hernández-Miramontes R, Salazar-Juárez A. The morphine/heroin vaccine decreased the heroin-induced antinociceptive and reinforcing effects in three inbred strains mouse. Int Immunopharmacol 2021; 98:107887. [PMID: 34186279 DOI: 10.1016/j.intimp.2021.107887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 11/25/2022]
Abstract
Clinical trials have indicated that a vaccine must be immunogenic in genetically diverse human populations and that immunogenicity and protective efficacy in animal models are two key indices required for the approval of a new vaccine. Additionally, the immune response (immunogenicity) and immunoprotection are dependent on the mouse strain. Therefore, the objective of the present study was to determine the immune response (immunogenicity) and the protective efficacy (behavioral response) in three inbred mouse strains immunized with the M6TT vaccine. Female BALB/c, C57Bl/6, and DBA/2 inbred mice were immunized with the M6-TT vaccine. A solid-phase antibody-capture ELISA was used to monitor antibody titer responses after each booster dose in vaccinated animals. The study used tail-flick testing to evaluate the antinociceptive effects induced by heroin. Additionally, heroin-induced locomotor activity and place preference were evaluated. The M6-TT vaccine was able to generate a specific antibody titer in the three inbred mouse strains evaluated. The antibodies reduced the antinociceptive effect of different doses of heroin. In addition, they decreased the heroin-induced locomotor activity and place preference. These findings suggest that the M6-TT vaccine generates a powerful immunogenic response capable of reducing the antinociceptive and reinforcing effects of heroin in different inbred mouse strains, which supports its possible future use in clinical trials in genetically diverse human populations.
Collapse
Affiliation(s)
- Susana Barbosa-Méndez
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, México DF 14370, Mexico
| | - Maura Matus-Ortega
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, México DF 14370, Mexico
| | - Ricardo Hernández-Miramontes
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, México DF 14370, Mexico
| | - Alberto Salazar-Juárez
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, México DF 14370, Mexico.
| |
Collapse
|
8
|
Slc1a3-2A-CreERT2 mice reveal unique features of Bergmann glia and augment a growing collection of Cre drivers and effectors in the 129S4 genetic background. Sci Rep 2021; 11:5412. [PMID: 33686166 PMCID: PMC7940647 DOI: 10.1038/s41598-021-84887-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 02/22/2021] [Indexed: 12/26/2022] Open
Abstract
Genetic variation is a primary determinant of phenotypic diversity. In laboratory mice, genetic variation can be a serious experimental confounder, and thus minimized through inbreeding. However, generalizations of results obtained with inbred strains must be made with caution, especially when working with complex phenotypes and disease models. Here we compared behavioral characteristics of C57Bl/6—the strain most widely used in biomedical research—with those of 129S4. In contrast to 129S4, C57Bl/6 demonstrated high within-strain and intra-litter behavioral hyperactivity. Although high consistency would be advantageous, the majority of disease models and transgenic tools are in C57Bl/6. We recently established six Cre driver lines and two Cre effector lines in 129S4. To augment this collection, we genetically engineered a Cre line to study astrocytes in 129S4. It was validated with two Cre effector lines: calcium indicator gCaMP5g-tdTomato and RiboTag—a tool widely used to study cell type-specific translatomes. These reporters are in different genomic loci, and in both the Cre was functional and astrocyte-specific. We found that calcium signals lasted longer and had a higher amplitude in cortical compared to hippocampal astrocytes, genes linked to a single neurodegenerative disease have highly divergent expression patterns, and that ribosome proteins are non-uniformly expressed across brain regions and cell types.
Collapse
|
9
|
Baird TR, Davies RA, Glennon RA, Peace MR, Negus SS. A Strategy to Prioritize Emerging Drugs of Abuse for Analysis: Abuse Liability Testing Using Intracranial Self-Stimulation (ICSS) in Rats and Validation with α-Pyrrolidinohexanophenone (α-PHP). EMERGING TRENDS IN DRUGS, ADDICTIONS, AND HEALTH 2021; 1:100004. [PMID: 34296204 PMCID: PMC8294200 DOI: 10.1016/j.etdah.2021.100004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Novel psychoactive substances (NPS) threaten public health and safety while also straining the limited resources of forensic laboratories. To efficiently allocate the finite resources available, we propose a new strategy for prioritizing NPS with abuse liability testing using a preclinical behavioral procedure in rats known as intracranial self-stimulation (ICSS). To validate this assay, the recently-scheduled synthetic cathinone α-PHP was compared to cocaine, a mechanistically similar drug of abuse, as a positive control and saline as a negative control. Male Sprague-Dawley rats (n=6) were implanted with electrodes targeting the medial forebrain bundle and trained to respond by lever-press for electrical brain stimulation. The rats were tested with doses of 0.32, 1.0, and 3.2 mg/kg α-PHP as well as 10 mg/kg of cocaine and saline administered by intraperitoneal injection. Neither saline nor 0.32 mg/kg α-PHP altered ICSS response rates compared to baseline levels of responding; however, doses of 1.0 and 3.2 mg/kg α-PHP and 10 mg/kg cocaine facilitated ICSS responding. This ICSS profile suggests that α-PHP has high abuse potential, with a rapid onset of effects and a long duration of action, and supports the decision to schedule this compound. This study demonstrates the ability of ICSS to distinguish between compounds of low and high potential for abuse. A strategy is proposed here to screen NPS using ICSS and classify emerging drugs into four priority categories for further analysis.
Collapse
Affiliation(s)
- Tyson R. Baird
- Integrative Life Sciences Doctoral Program, Virginia Commonwealth University
- Department of Forensic Science, Virginia Commonwealth University
| | - Rachel A. Davies
- Department of Medicinal Chemistry, Virginia Commonwealth University
| | | | | | - S. Stevens Negus
- Department of Pharmacology & Toxicology, Virginia Commonwealth University
| |
Collapse
|
10
|
Negus SS, Moerke MJ. Determinants of opioid abuse potential: Insights using intracranial self-stimulation. Peptides 2019; 112:23-31. [PMID: 30391425 PMCID: PMC6342636 DOI: 10.1016/j.peptides.2018.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022]
Abstract
Intracranial self-stimulation (ICSS) is one procedure that can be used for preclinical abuse potential assessment. In ICSS procedures, subjects with microelectrodes implanted into a brain-reward region are trained to press an operant response lever for pulses of electrical brain stimulation, and drugs are evaluated for their effectiveness to increase or "facilitate" ICSS responding (an abuse-related effect) or to depress ICSS responding (an abuse-limiting effect). ICSS has been used for decades to evaluate determinants of opioid abuse potential, and this article reviews pharmacological and biological determinants of opioid abuse potential as revealed by ICSS studies in rodents. One of the most important observations from ICSS studies is that abused mu opioid receptor (MOR) agonists like morphine often fail to produce abuse-related ICSS facilitation in opioid-naïve subjects, but several days of repeated opioid exposure is sufficient for opioid-induced facilitation to emerge. Future studies with ICSS could help (a) to clarify mechanisms that increase MOR agonist abuse potential during early opioid exposure or during chronic exposure leading to dependence, (b) to evaluate novel opioids either developed as candidate analgesics with reduced abuse potential or identified as designer opioids being synthesized and distributed for illicit use, and (c) to test candidate pharmacotherapies for treatment of opioid abuse in non-dependent and dependent subjects.
Collapse
Affiliation(s)
- S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond VA 23298, United States.
| | - Megan J Moerke
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond VA 23298, United States
| |
Collapse
|
11
|
|
12
|
Differential Expression of Munc13-2 Produces Unique Synaptic Phenotypes in the Basolateral Amygdala of C57BL/6J and DBA/2J Mice. J Neurosci 2017; 36:10964-10977. [PMID: 27798178 DOI: 10.1523/jneurosci.1785-16.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/28/2016] [Indexed: 11/21/2022] Open
Abstract
C57BL/6J (B6) and DBA/2J (D2) mice are well known to differentially express a number of behavioral phenotypes, including anxiety-like behavior, fear conditioning, and drug self-administration. However, the cellular mechanisms contributing to these differences remain unclear. Given the basolateral amygdala (BLA) contributes to these behaviors, we characterized strain-dependent differences in presynaptic and postsynaptic function in BLA neurons by integrating electrophysiological, biochemical, and genetic approaches to identify specific molecular mechanisms. We found that D2 glutamatergic synapses expressed enhanced release probability and lower sensitivity to both the inhibitory effects of low extracellular calcium and facilitation by phorbol esters. Furthermore, repetitive stimulation of BLA afferents at low (2 Hz) or high (40 Hz) frequencies revealed that B6 terminals, relative to D2 terminals, were more sensitive to synaptic fatigue principally because of reduced vesicle recycling rates. Additionally, B6 synapses exhibited more robust augmentation of spontaneous release after repetitive stimulation relative to the D2 strain. In silico analysis of the inheritance of synaptic physiology from an array of BXD recombinant inbred strains (Jansen et al., 2011) identified a segment on chromosome 4 containing the gene encoding Munc13-2, which has calcium-/phorbol ester-binding domains and controls presynaptic function. We subsequently found that B6 mice express substantially higher levels of Munc13-2 compared with the D2 strain whereas expression of several release-related proteins, including Munc13-1, was equivalent. We then knocked down the expression of Munc13-2 in B6 mice using a short hairpin RNA and found this recapitulated the presynaptic phenotype of D2 BLA synapses. SIGNIFICANCE STATEMENT DBA/2J and C57BL/6J mice have been used to understand the genetic mechanisms controlling behaviors related to a number of psychiatric illnesses. However, the fundamental neurobiological mechanisms producing these behavioral characteristics remain unresolved. Here we identify a critical family of presynaptic proteins differentially expressed by these strains that control strain-dependent synaptic physiology. This family of proteins regulates excitation/secretion coupling, vesicle recycling, and short-term plasticity throughout the CNS. Thus, differential inheritance of proteins like Munc13-2 has broad implications for genetic control over a wide variety of pathological behaviors. Importantly, these proteins also contain a large number of modulatory sites, making them attractive potential targets for the development of novel neuropharmaceutical treatments.
Collapse
|
13
|
Vivian CJ, Brinker AE, Graw S, Koestler DC, Legendre C, Gooden GC, Salhia B, Welch DR. Mitochondrial Genomic Backgrounds Affect Nuclear DNA Methylation and Gene Expression. Cancer Res 2017; 77:6202-6214. [PMID: 28663334 DOI: 10.1158/0008-5472.can-17-1473] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 12/19/2022]
Abstract
Mitochondrial DNA (mtDNA) mutations and polymorphisms contribute to many complex diseases, including cancer. Using a unique mouse model that contains nDNA from one mouse strain and homoplasmic mitochondrial haplotypes from different mouse strain(s)-designated Mitochondrial Nuclear Exchange (MNX)-we showed that mtDNA could alter mammary tumor metastasis. Because retrograde and anterograde communication exists between the nuclear and mitochondrial genomes, we hypothesized that there are differential mtDNA-driven changes in nuclear (n)DNA expression and DNA methylation. Genome-wide nDNA methylation and gene expression were measured in harvested brain tissue from paired wild-type and MNX mice. Selective differential DNA methylation and gene expression were observed between strains having identical nDNA, but different mtDNA. These observations provide insights into how mtDNA could be altering epigenetic regulation and thereby contribute to the pathogenesis of metastasis. Cancer Res; 77(22); 6202-14. ©2017 AACR.
Collapse
Affiliation(s)
- Carolyn J Vivian
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas.,Heartland Center for Mitochondrial Medicine, Phoenix, Arizona
| | - Amanda E Brinker
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas.,Heartland Center for Mitochondrial Medicine, Phoenix, Arizona.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Stefan Graw
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas
| | - Devin C Koestler
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas.,The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas
| | | | | | - Bodour Salhia
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Danny R Welch
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas. .,Heartland Center for Mitochondrial Medicine, Phoenix, Arizona.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas.,The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
14
|
Neelakantan H, Ward SJ, Walker EA. Effects of paclitaxel on mechanical sensitivity and morphine reward in male and female C57Bl6 mice. Exp Clin Psychopharmacol 2016; 24:485-495. [PMID: 27929349 PMCID: PMC5157702 DOI: 10.1037/pha0000097] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study evaluated the hypothesis that a paclitaxel treatment regimen sufficient to produce mechanical allodynia would alter sensitivities of male and female mice to the conditioned rewarding and reinforcing effects of morphine. Saline or paclitaxel were administered on Days 1, 3, 5, and 7 in male and female C57Bl/6 mice to induce morphine-reversible mechanical allodynia as measured by the Von Frey filament test. Paclitaxel treatment did not change sensitivity to morphine conditioned place preference (CPP) relative to saline treatment in either male or female mice. Morphine produced peak self-administration under a fixed ratio-1 (FR1) schedule of reinforcement for 0.03 mg/kg morphine per infusion in female mice and 0.1 mg/kg morphine per infusion in male mice. During the progressive ratio experiments, saline treatment in male mice decreased the number of morphine infusions for 12 days whereas the paclitaxel-treated male mice maintained responding for morphine similar to baseline levels during the same time period. However, paclitaxel did not have an overall effect on the reinforcing efficacy of morphine assessed over a limited dose range during the course of the repeated self-administration. These results suggest that the reward-related behavioral effects of morphine are overall not robustly altered by the presence of paclitaxel treatment under the current dosing regimen, with the exception of maintaining a small yet significant higher baseline than saline treatment during the development of allodynia in male mice. (PsycINFO Database Record
Collapse
Affiliation(s)
| | | | - Ellen Ann Walker
- Department of Pharmaceutical Sciences & Center for Substance Abuse Research, Temple University
| |
Collapse
|
15
|
Robinson JE, Vardy E, DiBerto JF, Chefer VI, White KL, Fish EW, Chen M, Gigante E, Krouse MC, Sun H, Thorsell A, Roth BL, Heilig M, Malanga CJ. Receptor Reserve Moderates Mesolimbic Responses to Opioids in a Humanized Mouse Model of the OPRM1 A118G Polymorphism. Neuropsychopharmacology 2015; 40:2614-22. [PMID: 25881115 PMCID: PMC4569952 DOI: 10.1038/npp.2015.109] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 11/09/2022]
Abstract
The OPRM1 A118G polymorphism is the most widely studied μ-opioid receptor (MOR) variant. Although its involvement in acute alcohol effects is well characterized, less is known about the extent to which it alters responses to opioids. Prior work has shown that both electrophysiological and analgesic responses to morphine but not to fentanyl are moderated by OPRM1 A118G variation, but the mechanism behind this dissociation is not known. Here we found that humanized mice carrying the 118GG allele (h/mOPRM1-118GG) were less sensitive than h/mOPRM1-118AA littermates to the rewarding effects of morphine and hydrocodone but not those of other opioids measured with intracranial self-stimulation. Reduced morphine reward in 118GG mice was associated with decreased dopamine release in the nucleus accumbens and reduced effects on GABA release in the ventral tegmental area that were not due to changes in drug potency or efficacy in vitro or receptor-binding affinity. Fewer MOR-binding sites were observed in h/mOPRM1-118GG mice, and pharmacological reduction of MOR availability unmasked genotypic differences in fentanyl sensitivity. These findings suggest that the OPRM1 A118G polymorphism decreases sensitivity to low-potency agonists by decreasing receptor reserve without significantly altering receptor function.
Collapse
Affiliation(s)
- J Elliott Robinson
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eyal Vardy
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jeffrey F DiBerto
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Vladimir I Chefer
- Intramural Research Program, National Institute on Drug Abuse (NIDA), Baltimore, MD, USA
| | - Kate L White
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eric W Fish
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Meng Chen
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eduardo Gigante
- Intramural Research Program, National Institute on Drug Abuse (NIDA), Baltimore, MD, USA
| | - Michael C Krouse
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hui Sun
- Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA), Bethesda, MD, USA
| | - Annika Thorsell
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Bryan L Roth
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,NIMH Psychoactive Drug Screening Program (PDSP), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Markus Heilig
- Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA), Bethesda, MD, USA,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - C J Malanga
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Department of Neurology, University of North Carolina School of Medicine, Physicians' Office Building, 170 Manning Drive, CB 7025, Chapel Hill, NC 27599-7025, USA, Tel: +1 919 966 1683, Fax: +1 919 843 4576, E-mail:
| |
Collapse
|
16
|
The Convergence of Systems and Reductionist Approaches in Complex Trait Analysis. Cell 2015; 162:23-32. [PMID: 26140590 DOI: 10.1016/j.cell.2015.06.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Indexed: 01/16/2023]
Abstract
Research into the genetic and environmental factors behind complex trait variation has traditionally been segregated into distinct scientific camps. The reductionist approach aims to decrypt phenotypic variability bit by bit, founded on the underlying hypothesis that genome-to-phenome relations are largely constructed from the additive effects of their molecular players. In contrast, the systems approach aims to examine large-scale interactions of many components simultaneously, on the premise that interactions in gene networks can be both linear and non-linear. Both approaches are complementary, and they are becoming increasingly intertwined due to developments in gene editing tools, omics technologies, and population resources. Together, these strategies are beginning to drive the next era in complex trait research, paving the way to improve agriculture and toward more personalized medicine.
Collapse
|
17
|
Ethanol self-administration in mice under a second-order schedule. Alcohol 2015; 49:561-70. [PMID: 26254963 DOI: 10.1016/j.alcohol.2015.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 06/09/2015] [Accepted: 06/20/2015] [Indexed: 11/23/2022]
Abstract
Long Fixed-Interval (FI) schedules, particularly second-order schedules, can engender substantial responding before drug or ethanol delivery that is uninfluenced by the direct effects of the drug or ethanol. Thus, these schedules can be used to study the effects of medications upon drug- or ethanol-seeking, uninfluenced by the direct effects of the self-administered drug or ethanol. Long FI second-order schedules are frequently used in primates and occasionally in rats. Under second-order schedules, completion of one response requirement, e.g., a Fixed Ratio 10 (FR10:S), produces a brief stimulus presentation, e.g., a 1-s 80-dB 4-kHZ tone, and this FR10:S serves as the response unit under another schedule, e.g., an FI 1800-s. Thus, the first FR10 completed after 1800 s would result in delivery both of the tone and of reinforcement, e.g., 10 × 0.01 mL 16% (w/v) ethanol. To examine if such schedules could be effectively used in mice, which have advantages in neurobiological and genetic studies, we trained eight C57BL/6J mice to respond under the schedule just described. This schedule maintained substantial responding. The temporal pattern of behavior was typical of an FI schedule with responding accelerating across the interval. We also examined the effects of acute and chronic administration of fluvoxamine on this responding, and these were modest. Finally, we examined responding when alcohol and/or tone deliveries were withheld, and found that extinction occurred most rapidly when both were withheld. This work demonstrates that long FI schedules of ethanol delivery may be useful in studying ethanol seeking in mice.
Collapse
|
18
|
Patrono E, Di Segni M, Patella L, Andolina D, Valzania A, Latagliata EC, Felsani A, Pompili A, Gasbarri A, Puglisi-Allegra S, Ventura R. When chocolate seeking becomes compulsion: gene-environment interplay. PLoS One 2015; 10:e0120191. [PMID: 25781028 PMCID: PMC4363151 DOI: 10.1371/journal.pone.0120191] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 02/04/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Eating disorders appear to be caused by a complex interaction between environmental and genetic factors, and compulsive eating in response to adverse circumstances characterizes many eating disorders. MATERIALS AND METHODS We compared compulsion-like eating in the form of conditioned suppression of palatable food-seeking in adverse situations in stressed C57BL/6J and DBA/2J mice, two well-characterized inbred strains, to determine the influence of gene-environment interplay on this behavioral phenotype. Moreover, we tested the hypothesis that low accumbal D2 receptor (R) availability is a genetic risk factor of food compulsion-like behavior and that environmental conditions that induce compulsive eating alter D2R expression in the striatum. To this end, we measured D1R and D2R expression in the striatum and D1R, D2R and α1R levels in the medial prefrontal cortex, respectively, by western blot. RESULTS Exposure to environmental conditions induces compulsion-like eating behavior, depending on genetic background. This behavioral pattern is linked to decreased availability of accumbal D2R. Moreover, exposure to certain environmental conditions upregulates D2R and downregulates α1R in the striatum and medial prefrontal cortex, respectively, of compulsive animals. These findings confirm the function of gene-environment interplay in the manifestation of compulsive eating and support the hypothesis that low accumbal D2R availability is a "constitutive" genetic risk factor for compulsion-like eating behavior. Finally, D2R upregulation and α1R downregulation in the striatum and medial prefrontal cortex, respectively, are potential neuroadaptive responses that parallel the shift from motivated to compulsive eating.
Collapse
Affiliation(s)
- Enrico Patrono
- Department of Applied Clinical Science and Biotechnology, University of L'Aquila, Coppito, Italy
| | - Matteo Di Segni
- Santa Lucia Foundation, Rome, Italy
- Department of Psychology and Center “Daniel Bovet,” Sapienza University, Rome, Italy
| | - Loris Patella
- Department of Applied Clinical Science and Biotechnology, University of L'Aquila, Coppito, Italy
| | - Diego Andolina
- Santa Lucia Foundation, Rome, Italy
- Department of Applied Clinical Science and Biotechnology, University of L'Aquila, Coppito, Italy
| | - Alessandro Valzania
- Santa Lucia Foundation, Rome, Italy
- Department of Psychology and Center “Daniel Bovet,” Sapienza University, Rome, Italy
| | - Emanuele Claudio Latagliata
- Santa Lucia Foundation, Rome, Italy
- Department of Psychology and Center “Daniel Bovet,” Sapienza University, Rome, Italy
| | - Armando Felsani
- CNR, Institute of Cellular Biology and Neurobiology, Rome, Italy
| | - Assunta Pompili
- Department of Applied Clinical Science and Biotechnology, University of L'Aquila, Coppito, Italy
| | - Antonella Gasbarri
- Department of Applied Clinical Science and Biotechnology, University of L'Aquila, Coppito, Italy
| | - Stefano Puglisi-Allegra
- Santa Lucia Foundation, Rome, Italy
- Department of Psychology and Center “Daniel Bovet,” Sapienza University, Rome, Italy
| | - Rossella Ventura
- Santa Lucia Foundation, Rome, Italy
- Department of Psychology and Center “Daniel Bovet,” Sapienza University, Rome, Italy
- * E-mail:
| |
Collapse
|
19
|
Katsidoni V, Alexiou P, Fotiadou M, Pelecanou M, Sagnou M, Panagis G. Curcumin, demethoxycurcumin and bisdemethoxycurcumin differentially inhibit morphine's rewarding effect in rats. Psychopharmacology (Berl) 2014; 231:4467-78. [PMID: 24838368 DOI: 10.1007/s00213-014-3603-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/12/2014] [Indexed: 12/19/2022]
Abstract
RATIONALE Recent animal studies reported that curcumin, the active constituent of Curcuma longa, has several central actions and may attenuate morphine tolerance. OBJECTIVES In the present study, we utilized the intracranial self-stimulation (ICSS) paradigm to examine the effects of the commercially available curcuminoid mixture and each one of its components, individually, on brain stimulation reward and on the reward-facilitating effect of morphine. METHODS Male Sprague-Dawley rats were implanted with an electrode into the medial forebrain bundle and trained to respond for electrical stimulation using a rate-frequency paradigm. In the first study, rats were injected with graded doses either of the curcuminoid mixture, or curcumin I, or II, or III. In the second study, we examined whether a low dose of the curcuminoid mixture or each individual curcumin analogue composing it could counteract the reward-facilitating effect of morphine. RESULTS At low doses, both the curcuminoid mixture and curcumin I did not affect brain stimulation reward, whereas, higher doses increased ICSS thresholds. Curcumin II and curcumin III did not affect brain stimulation reward at any doses. Subthreshold doses of the curcuminoid mixture and curcumin I inhibited the reward-facilitating effect of morphine. CONCLUSION Both the curcuminoid mixture and curcumin I lack hedonic properties and moderate the reward-facilitating effect of morphine. Our data suggest that curcumin interferes with brain reward mechanisms responsible for the expression of the acute reinforcing properties of opioids and provide evidence that curcumin may be a promising adjuvant for attenuating morphine's rewarding effects in patients who are under long-term opioid therapy.
Collapse
Affiliation(s)
- Vicky Katsidoni
- Laboratory of Behavioral Neuroscience, Department of Psychology, School of Social Sciences, University of Crete, 74100, Rethymno, Crete, Greece
| | | | | | | | | | | |
Collapse
|
20
|
Fish EW, DiBerto JF, Krouse MC, Robinson JE, Malanga CJ. Different contributions of dopamine D1 and D2 receptor activity to alcohol potentiation of brain stimulation reward in C57BL/6J and DBA/2J mice. J Pharmacol Exp Ther 2014; 350:322-9. [PMID: 24917543 PMCID: PMC4109490 DOI: 10.1124/jpet.114.216135] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/06/2014] [Indexed: 02/02/2023] Open
Abstract
C57BL/6J (C57) and DBA/2J (DBA) mice respond differently to drugs that affect dopamine systems, including alcohol. The current study compared effects of D1 and D2 receptor agonists and antagonists, and the interaction between D1/D2 antagonists and alcohol, on intracranial self-stimulation in male C57 and DBA mice to determine the role of dopamine receptors in the effects of alcohol on brain stimulation reward (BSR). In the initial strain comparison, dose effects on BSR thresholds and maximum operant response rates were determined for the D1 receptor agonist SKF-82958 (±-6-chloro-7,8-dihydroxy-3-allyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine; 0.1-0.56 mg/kg) and antagonist SCH 23390 (+-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepinehydrochloride; 0.003-0.056 mg/kg), and the D2 receptor agonist quinpirole (0.1-3.0 mg/kg) and antagonist raclopride (0.01-0.56 mg/kg). For the alcohol interaction, SCH 23390 (0.003 mg/kg) or raclopride (0.03 mg/kg) was given before alcohol (0.6-2.4 g/kg p.o.). D1 antagonism dose-dependently elevated and SKF-82958 dose-dependently lowered BSR threshold in both strains; DBA mice were more sensitive to SKF-82958 effects. D2 antagonism dose-dependently elevated BSR threshold only in C57 mice. Low doses of quinpirole elevated BSR threshold equally in both strains, whereas higher doses of quinpirole lowered BSR threshold only in C57 mice. SCH 23390, but not raclopride, prevented lowering of BSR threshold by alcohol in DBA mice. Conversely, raclopride, but not SCH 23390, prevented alcohol potentiation of BSR in C57 mice. These results extend C57 and DBA strain differences to D1/D2 sensitivity of BSR, and suggest differential involvement of D1 and D2 receptors in the acute rewarding effects of alcohol in these two mouse strains.
Collapse
Affiliation(s)
- Eric W Fish
- Bowles Center for Alcohol Studies (E.W.F., J.E.R., C.J.M.), Department of Neurology, University of North Carolina School of Medicine (J.F.D., M.C.K., J.E.R., C.J.M.), and Neurobiology Curriculum, University of North Carolina School of Medicine (J.E.R., C.J.M.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jeffrey F DiBerto
- Bowles Center for Alcohol Studies (E.W.F., J.E.R., C.J.M.), Department of Neurology, University of North Carolina School of Medicine (J.F.D., M.C.K., J.E.R., C.J.M.), and Neurobiology Curriculum, University of North Carolina School of Medicine (J.E.R., C.J.M.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael C Krouse
- Bowles Center for Alcohol Studies (E.W.F., J.E.R., C.J.M.), Department of Neurology, University of North Carolina School of Medicine (J.F.D., M.C.K., J.E.R., C.J.M.), and Neurobiology Curriculum, University of North Carolina School of Medicine (J.E.R., C.J.M.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - J Elliott Robinson
- Bowles Center for Alcohol Studies (E.W.F., J.E.R., C.J.M.), Department of Neurology, University of North Carolina School of Medicine (J.F.D., M.C.K., J.E.R., C.J.M.), and Neurobiology Curriculum, University of North Carolina School of Medicine (J.E.R., C.J.M.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - C J Malanga
- Bowles Center for Alcohol Studies (E.W.F., J.E.R., C.J.M.), Department of Neurology, University of North Carolina School of Medicine (J.F.D., M.C.K., J.E.R., C.J.M.), and Neurobiology Curriculum, University of North Carolina School of Medicine (J.E.R., C.J.M.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
21
|
Doyle GA, Schwebel CL, Ruiz SE, Chou AD, Lai AT, Wang MJ, Smith GG, Buono RJ, Berrettini WH, Ferraro TN. Analysis of candidate genes for morphine preference quantitative trait locus Mop2. Neuroscience 2014; 277:403-16. [PMID: 25058503 DOI: 10.1016/j.neuroscience.2014.07.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 07/14/2014] [Accepted: 07/14/2014] [Indexed: 11/26/2022]
Abstract
Compared to DBA/2J (D2), C57BL/6J (B6) inbred mice exhibit strong morphine preference when tested using a two-bottle choice drinking paradigm. A morphine preference quantitative trait locus (QTL), Mop2, was originally mapped to proximal chromosome (Chr) 10 using a B6xD2 F2 intercross population, confirmed with reciprocal congenic strains and fine mapped with recombinant congenic strains. These efforts identified a ∼ 10-Million base pair (Mbp) interval, underlying Mop2, containing 35 genes. To further reduce the interval, mice from the D2.B6-Mop2-P1 congenic strain were backcrossed to parental D2 mice and two new recombinant strains of interest were generated: D2.B6-Mop2-P1.pD.dB and D2.B6-Mop2-P1.pD.dD. Results obtained from testing these strains in the two-bottle choice drinking paradigm suggest that the gene(s) responsible for the Mop2 QTL is one or more of 22 remaining within the newly defined interval (∼ 7.6 Mbp) which includes Oprm1 and several other genes related to opioid pharmacology. Real-time qRT-PCR analysis of Oprm1 and opioid-related genes Rgs17, Ppp1r14c, Vip, and Iyd revealed both between-strain and within-strain expression differences in comparisons of saline- and morphine-treated B6 and D2 mice. Analysis of Rgs17 protein levels also revealed both between-strain and within-strain differences in comparisons of saline- and morphine-treated B6 and D2 mice. Results suggest that the Mop2 QTL represents the combined influence of multiple genetic variants on morphine preference in these two strains. Relative contributions of each variant remain to be determined.
Collapse
Affiliation(s)
- G A Doyle
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - C L Schwebel
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - S E Ruiz
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - A D Chou
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - A T Lai
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - M-J Wang
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - G G Smith
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Research Services, Department of Veterans Affairs Medical Center, Coatesville, PA, USA
| | - R J Buono
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - W H Berrettini
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - T N Ferraro
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| |
Collapse
|
22
|
Abstract
Intracranial self-stimulation (ICSS) is a behavioral procedure in which operant responding is maintained by pulses of electrical brain stimulation. In research to study abuse-related drug effects, ICSS relies on electrode placements that target the medial forebrain bundle at the level of the lateral hypothalamus, and experimental sessions manipulate frequency or amplitude of stimulation to engender a wide range of baseline response rates or response probabilities. Under these conditions, drug-induced increases in low rates/probabilities of responding maintained by low frequencies/amplitudes of stimulation are interpreted as an abuse-related effect. Conversely, drug-induced decreases in high rates/probabilities of responding maintained by high frequencies/amplitudes of stimulation can be interpreted as an abuse-limiting effect. Overall abuse potential can be inferred from the relative expression of abuse-related and abuse-limiting effects. The sensitivity and selectivity of ICSS to detect abuse potential of many classes of abused drugs is similar to the sensitivity and selectivity of drug self-administration procedures. Moreover, similar to progressive-ratio drug self-administration procedures, ICSS data can be used to rank the relative abuse potential of different drugs. Strengths of ICSS in comparison with drug self-administration include 1) potential for simultaneous evaluation of both abuse-related and abuse-limiting effects, 2) flexibility for use with various routes of drug administration or drug vehicles, 3) utility for studies in drug-naive subjects as well as in subjects with controlled levels of prior drug exposure, and 4) utility for studies of drug time course. Taken together, these considerations suggest that ICSS can make significant contributions to the practice of abuse potential testing.
Collapse
Affiliation(s)
- S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Laurence L Miller
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
23
|
Modeling combined schizophrenia-related behavioral and metabolic phenotypes in rodents. Behav Brain Res 2014; 276:130-42. [PMID: 24747658 DOI: 10.1016/j.bbr.2014.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/08/2014] [Accepted: 04/09/2014] [Indexed: 12/11/2022]
Abstract
Schizophrenia is a chronic, debilitating disorder with a complex behavioral and cognitive phenotype underlined by a similarly complex etiology involving an interaction between susceptibility genes and environmental factors during early development. Limited progress has been made in developing novel pharmacotherapy, partly due to a lack of valid animal models. The recent recognition of the potentially causal role of central and peripheral energy metabolism in the pathophysiology of schizophrenia raises the need of research on animal models that combine both behavioral and metabolic phenotypic domains, similar to what have been identified in humans. In this review we focus on selected genetic (DBA/2J mice, leptin receptor mutants, and PSD-93 knockout mice), early neurodevelopmental (maternal protein deprivation) and pharmacological (acute phencyclidine) animal models that capture the combined behavioral and metabolic abnormalities shown by schizophrenic patients. In reviewing behavioral phenotypes relevant to schizophrenia we apply the principles established by the Research Domain Criteria (RDoC) for better translation. We demonstrate that etiologically diverse manipulations such as specific breeding, deletion of genes that are primarily involved in metabolic regulation and in synaptic plasticity, as well as early metabolic deprivation and adult pharmacological challenge of the glutamate system can lead to schizophrenia-related behavioral and metabolic phenotypes, which suggest that these pathways might be interlinked. We propose that using animal models that combine different domains of schizophrenia can be used as a translationally valid approach to capture the system-level complex interplay between peripheral and central processes in the development of psychopathology.
Collapse
|
24
|
Bauer CT, Banks ML, Blough BE, Negus SS. Use of intracranial self-stimulation to evaluate abuse-related and abuse-limiting effects of monoamine releasers in rats. Br J Pharmacol 2013; 168:850-62. [PMID: 22978626 DOI: 10.1111/j.1476-5381.2012.02214.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 08/17/2012] [Accepted: 08/30/2012] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Monoamine releasers constitute a class of drugs that promote the release of dopamine (DA), serotonin (5-HT) and/or norepinephrine. Although some drugs in this class are well-known drugs of abuse (amphetamine, methamphetamine), others are thought to have reduced (3,4-methylenedioxy-N-methylamphetamine [MDMA]) or no (fenfluramine) abuse potential. The purpose of this study was to further elucidate the role of dopamine versus serotonin selectivity on expression of abuse-related effects produced by monoamine releasers in an assay of intracranial self-stimulation (ICSS) in rats. EXPERIMENTAL APPROACH This study evaluated effects produced in a frequency-rate ICSS procedure by 11 monoamine releasers that vary in selectivity to release DA versus 5-HT. KEY RESULTS Efficacy of monoamine releasers to facilitate ICSS correlated with DA-selectivity, such that DA-selective releasers exclusively facilitated ICSS, a 5-HT-selective releaser exclusively depressed ICSS, and mixed-action releasers both facilitated low ICSS rates and depressed high ICSS rates. Fixed-proportion mixtures of a DA-selective releaser and a 5-HT-selective releaser recapitulated effects of mixed-action releasers. Efficacy of monoamine releasers to facilitate ICSS also correlated with previously published data on efficacy to maintain self-administration in rhesus monkeys responding under a progressive-ratio schedule of reinforcement. CONCLUSIONS AND IMPLICATIONS These data support the importance of selectivity for DA versus 5-HT in determining abuse potential of monoamine releasers and demonstrate a novel correlation between rat ICSS and nonhuman primate self-administration measures of abuse-related effects. Taken together, these results support the use of ICSS in rats as an experimental tool to study the expression and pharmacological determinants of abuse-related effects of monoamine releasers.
Collapse
Affiliation(s)
- C T Bauer
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | |
Collapse
|
25
|
Tapocik JD, Luu TV, Mayo CL, Wang BD, Doyle E, Lee AD, Lee NH, Elmer GI. Neuroplasticity, axonal guidance and micro-RNA genes are associated with morphine self-administration behavior. Addict Biol 2013; 18:480-95. [PMID: 22804800 DOI: 10.1111/j.1369-1600.2012.00470.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neuroadaptations in the ventral striatum (VS) and ventral midbrain (VMB) following chronic opioid administration are thought to contribute to the pathogenesis and persistence of opiate addiction. In order to identify candidate genes involved in these neuroadaptations, we utilized a behavior-genetics strategy designed to associate contingent intravenous drug self-administration with specific patterns of gene expression in inbred mice differentially predisposed to the rewarding effects of morphine. In a Yoked-control paradigm, C57BL/6J mice showed clear morphine-reinforced behavior, whereas DBA/2J mice did not. Moreover, the Yoked-control paradigm revealed the powerful consequences of self-administration versus passive administration at the level of gene expression. Morphine self-administration in the C57BL/6J mice uniquely up- or down-regulated 237 genes in the VS and 131 genes in the VMB. Interestingly, only a handful of the C57BL/6J self-administration genes (<3%) exhibited a similar expression pattern in the DBA/2J mice. Hence, specific sets of genes could be confidently assigned to regional effects of morphine in a contingent- and genotype-dependent manner. Bioinformatics analysis revealed that neuroplasticity, axonal guidance and micro-RNAs (miRNAs) were among the key themes associated with drug self-administration. Noteworthy were the primary miRNA genes H19 and micro-RNA containing gene (Mirg), processed, respectively, to mature miRNAs miR-675 and miR-154, because they are prime candidates to mediate network-like changes in responses to chronic drug administration. These miRNAs have postulated roles in dopaminergic neuron differentiation and mu-opioid receptor regulation. The strategic approach designed to focus on reinforcement-associated genes provides new insight into the role of neuroplasticity pathways and miRNAs in drug addiction.
Collapse
Affiliation(s)
| | - Truong V. Luu
- Department of Pharmacology and Physiology; The George Washington University Medical Center; Washington; DC; USA
| | - Cheryl L. Mayo
- Department of Psychiatry; Maryland Psychiatric Research Center; University of Maryland School of Medicine; Baltimore; MD; USA
| | - Bi-Dar Wang
- Department of Pharmacology and Physiology; The George Washington University Medical Center; Washington; DC; USA
| | - Erin Doyle
- Department of Pharmacology and Physiology; The George Washington University Medical Center; Washington; DC; USA
| | - Alec D. Lee
- Department of Pharmacology and Physiology; The George Washington University Medical Center; Washington; DC; USA
| | - Norman H. Lee
- Department of Pharmacology and Physiology; The George Washington University Medical Center; Washington; DC; USA
| | - Greg I. Elmer
- Department of Psychiatry; Maryland Psychiatric Research Center; University of Maryland School of Medicine; Baltimore; MD; USA
| |
Collapse
|
26
|
Balter RE, Dykstra LA. Thermal sensitivity as a measure of spontaneous morphine withdrawal in mice. J Pharmacol Toxicol Methods 2013; 67:162-8. [PMID: 23416790 DOI: 10.1016/j.vascn.2013.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 02/04/2013] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Opioid withdrawal syndrome is a critical component of opioid abuse and consists of a wide array of symptoms including increases in pain sensitivity (hyperalgesia). A reliable preclinical model of hyperalgesia during opioid withdrawal is needed to evaluate possible interventions to alleviate withdrawal. The following study describes a method for assessing increases in thermal sensitivity on the hotplate in a mouse model of spontaneous morphine withdrawal. METHODS C57BL/6J mice received 5.5days of 30, 56, or 100mg/kg morphine or saline (s.c., twice daily). In Experiment I, thermal sensitivity data were collected at baseline and at 8, 24, 32, 48h and 1week following the final injection. Thermal sensitivity was assessed by examining latency to respond on a hotplate across a range of temperatures (50, 52, 54, and 56°C). In Experiment II, 0.01mg/kg buprenorphine was administered 30min prior to each testing session during the withdrawal period. In Experiment III, jumping during a 30min period was assessed at baseline and at 0, 8, 24, 32, and 48h following the final morphine injection. RESULTS During the withdrawal period, thermal sensitivity increased significantly in all morphine-treated mice as compared to saline-treated mice. Thermal sensitivity was greater in mice treated with 56mg/kg morphine compared to 30mg/kg and peaked earlier than in mice treated with 100mg/kg (32h v 1wk). The increase in thermal sensitivity following 56mg/kg morphine was attenuated by a dose of buprenorphine that did not produce antinociception alone (i.e., 0.01mg/kg). In general, the results of the jumping experiment paralleled those obtained in Experiment I. DISCUSSION Response latency on the hotplate is a reliable and sensitive measure of spontaneous morphine withdrawal in mice, making it an ideal behavior for assessing the potential of medications and environmental interventions to alleviate opioid withdrawal.
Collapse
Affiliation(s)
- Rebecca E Balter
- Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| | | |
Collapse
|
27
|
Abstract
This paper is the thirty-fourth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2011 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
28
|
Balter RE, Dykstra LA. The effect of environmental factors on morphine withdrawal in C57BL/6J mice: running wheel access and group housing. Psychopharmacology (Berl) 2012; 224:91-100. [PMID: 22903388 DOI: 10.1007/s00213-012-2826-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 07/24/2012] [Indexed: 12/17/2022]
Abstract
RATIONALE There is evidence to suggest that the rewarding effects of drugs of abuse can be altered by environmental manipulations such as housing conditions and access to running wheels. There is less information about how these environmental manipulations alter withdrawal behaviors following the termination of chronic drug administration. OBJECTIVES The objective of this study is to examine the effects of access to running wheels and group housing on spontaneous morphine withdrawal. METHODS C57BL/6J mice were assigned to one of the three housing conditions: wheel access (singly housed), no wheels (singly housed), or group-housed (no wheels). Mice received 30 or 56 mg/kg morphine or saline (s.c.) twice daily for 5.5 days. At baseline and at 8, 24, 32, and 48 h following the final injection, latency to respond on a hot plate was determined across a range of temperatures (50, 52, 54, and 56 °C). RESULTS Latency to respond decreased as a function of temperature. Response latencies during the withdrawal period were decreased in mice without wheel access treated with both 30 and 56 mg/kg of morphine. This increase in thermal sensitivity was significantly attenuated in singly housed mice with wheel access and in group-housed mice; however, the effects were less pronounced in the group-housed mice and depended upon the time during withdrawal. CONCLUSIONS Both wheel access and group housing attenuate the increase in thermal sensitivity seen in morphine-treated mice during morphine withdrawal.
Collapse
Affiliation(s)
- Rebecca E Balter
- Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | | |
Collapse
|
29
|
de Wit H, Phillips TJ. Do initial responses to drugs predict future use or abuse? Neurosci Biobehav Rev 2012; 36:1565-76. [PMID: 22542906 PMCID: PMC3372699 DOI: 10.1016/j.neubiorev.2012.04.005] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 03/09/2012] [Accepted: 04/12/2012] [Indexed: 10/28/2022]
Abstract
Individuals vary in their initial reactions to drugs of abuse in ways that may contribute to the likelihood of subsequent drug use. In humans, most drugs of abuse produce positive subjective states such as euphoria and feelings of well-being, which may facilitate repeated use. In nonhumans, many drugs initially increase locomotor activity and produce discriminative stimulus effects, both of which have been considered to be models of human stimulant and subjective states. Both humans and nonhumans vary in their sensitivity to early acute drug effects in ways that may predict future use or self-administration, and some of these variations appear to be genetic in origin. However, it is not known exactly how the initial responses to drugs in either humans or nonhumans relate to subsequent use or abuse. In humans, positive effects of drugs facilitate continued use of a drug while negative effects discourage use, and in nonhumans, greater genetic risk for drug intake is predicted by reduced sensitivity to drug aversive effects; but whether these initial responses affect escalation of drug use, and the development of dependence is currently unknown. Although early use of a drug is a necessary step in the progression to abuse and dependence, other variables may be of greater importance in the transition from use to abuse. Alternatively, the same variables that predict initial acute drug effects and early use may significantly contribute to continued use, escalation and dependence. Here we review the existing evidence for relations between initial direct drug effects, early use, and continued use. Ultimately, these relations can only be determined from systematic longitudinal studies with comprehensive assessments from early drug responses to progression of problem drug use. In parallel, additional investigation of initial responses in animal models as predictors of drug use will shed light on the underlying mechanisms.
Collapse
Affiliation(s)
- Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, 5841 S Maryland Ave, MC3077, Chicago, IL 60637, United States.
| | | |
Collapse
|
30
|
Mephedrone (4-methylmethcathinone) and intracranial self-stimulation in C57BL/6J mice: comparison to cocaine. Behav Brain Res 2012; 234:76-81. [PMID: 22728726 DOI: 10.1016/j.bbr.2012.06.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 06/07/2012] [Accepted: 06/13/2012] [Indexed: 11/21/2022]
Abstract
The recreational use of cathinone-derived synthetic stimulants, also known as "bath salts", has increased during the last five years. A commonly abused drug in this class is mephedrone (4-methylmethcathinone or "meow-meow"), which alters mood and produces euphoria in humans. Intracranial self-stimulation (ICSS) measures the behavioral effects of neuroactive compounds on brain reward circuitry. We used ICSS to investigate the ability of mephedrone and cocaine to alter responding for electrical stimulation of the medial forebrain bundle in C57BL/6J mice. Adult male C57BL/6J mice (n=6) implanted with unipolar stimulating electrodes at the level of the lateral hypothalamus responded for varying frequencies of brain stimulation reward (BSR). The frequency that supported half maximal responding (EF50), the BSR threshold (θ(0)), and the maximum response rate were determined before and after intraperitoneal administration of saline, mephedrone (1.0, 3.0, or 10.0 mg/kg), or cocaine (1.0, 3.0, or 10.0 mg/kg). Mephedrone dose-dependently decreased EF50 (max. effect=72.3% of baseline), θ(0) (max. effect=59.6% of baseline), and the maximum response rate (max. effect=67.0% of baseline) beginning 15 min after administration. Beginning immediately after administration, cocaine dose-dependently lowered EF50 (max. effect=66.4% of baseline) and θ(0) (max. effect=60.1% of baseline) but did not affect maximum response rate. These results suggest that mephedrone, like cocaine, potentiates BSR, which may indicate its potential for abuse. Given the public health concern of stimulant abuse, future studies will be necessary to determine the cellular and behavioral effects of acute and chronic mephedrone use.
Collapse
|
31
|
Fish EW, Robinson JE, Krouse MC, Hodge CW, Reed C, Phillips TJ, Malanga CJ. Intracranial self-stimulation in FAST and SLOW mice: effects of alcohol and cocaine. Psychopharmacology (Berl) 2012; 220:719-30. [PMID: 21983918 PMCID: PMC3289728 DOI: 10.1007/s00213-011-2523-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 09/21/2011] [Indexed: 11/29/2022]
Abstract
RATIONALE Sensitivity to the stimulant and rewarding effects of alcohol may be genetically correlated traits that predispose individuals to develop an alcohol use disorder. OBJECTIVE This study aimed to examine the effects of alcohol and cocaine on intracranial self-stimulation (ICSS) in FAST and SLOW mice, which were selectively bred for extremes in alcohol stimulation. METHODS Male FAST and SLOW mice were conditioned to respond for reinforcement by direct electrical stimulation of the medial forebrain bundle (i.e., brain stimulation reward). ICSS responses were determined immediately before and after oral gavage with water or alcohol (0.3-2.4 g/kg) or intraperitoneal injection with saline or cocaine (1.0-30.0 mg/kg). In separate FAST and SLOW mice, the locomotor effects of these treatments were measured in activity chambers. RESULTS Alcohol dose-dependently lowered the threshold for self-stimulation (θ (0)) and the frequency that maintained 50% of maximal responding (EF50) in FAST mice but did not significantly affect these parameters in SLOW mice. The largest effects of alcohol were after the 1.7- and 2.4-g/kg doses and were about 40% compared to water injection. Alcohol did not affect MAX response rates, but dose-dependently stimulated locomotor activity in FAST mice. Cocaine lowered thresholds equally in FAST and SLOW mice, although cocaine-stimulated locomotor activity was higher in the FAST than in the SLOW mice. CONCLUSIONS Selective breeding for alcohol locomotor stimulation also renders the mice more sensitive to the effects of alcohol, but not cocaine, on ICSS.
Collapse
Affiliation(s)
- Eric W Fish
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Potentiation of brain stimulation reward by morphine: effects of neurokinin-1 receptor antagonism. Psychopharmacology (Berl) 2012; 220:215-24. [PMID: 21909635 PMCID: PMC3484369 DOI: 10.1007/s00213-011-2469-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 08/23/2011] [Indexed: 10/17/2022]
Abstract
RATIONALE The abuse potential of opioids may be due to their reinforcing and rewarding effects, which may be attenuated by neurokinin-1 receptor (NK1R) antagonists. OBJECTIVE This study was conducted to measure the effects of opioid and NK1R blockade on the potentiation of brain stimulation reward (BSR) by morphine using the intracranial self-stimulation method. METHODS Adult male C57BL/6J mice (n = 15) were implanted with unipolar stimulating electrodes in the lateral hypothalamus and trained to respond for varying frequencies of rewarding electrical stimulation. The BSR threshold (θ(0)) and maximum response rate (MAX) were determined before and after intraperitoneal administration of saline, morphine (1.0-17.0 mg/kg), or the NK1R antagonists L-733,060 (1.0-17.0 mg/kg) and L-703,606 (1.0-17.0 mg/kg). In morphine antagonism experiments, naltrexone (0.1-1.0 mg/kg) or 10.0 mg/kg L-733,060 or L-703,606 was administered 15 min before morphine (1.0-10.0 mg/kg) or saline. RESULTS Morphine dose-dependently decreased θ(0) (maximum effect = 62% of baseline) and altered MAX when compared to saline. L-703,606 and L-733,060 altered θ(0); 10.0 mg/kg L-733,060 and L-703,606, which did not affect θ(0) or MAX, attenuated the effects of 3.0 and 10.0 mg/kg morphine, and 1.0 and 0.3 mg/kg naltrexone blocked the effects of 10.0 mg/kg morphine. Naltrexone given before saline did not affect θ(0) or MAX. CONCLUSIONS The decrease in θ(0) by morphine reflects its rewarding effects, which were attenuated by NK1R and opioid receptor blockade. These results demonstrate the importance of substance P signaling during limbic reward system activation by opioids.
Collapse
|
33
|
Klaus F, Amrein I. Running in laboratory and wild rodents: Differences in context sensitivity and plasticity of hippocampal neurogenesis. Behav Brain Res 2012; 227:363-70. [DOI: 10.1016/j.bbr.2011.04.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 03/23/2011] [Accepted: 04/19/2011] [Indexed: 01/01/2023]
|
34
|
Abstract
Operant methods are powerful behavioral tools for the study of motivated behavior. These 'self-administration' methods have been used extensively in drug addiction research due to their high construct validity. Operant studies provide researchers a tool for preclinical investigation of several aspects of the addiction process. For example, mechanisms of acute reinforcement (both drug and non-drug) can be tested using pharmacological or genetic tools to determine the ability of a molecular target to influence self-administration behavior. Additionally, drug or food seeking behaviors can be studied in the absence of the primary reinforcer, and the ability of pharmacological compounds to disrupt this process is a preclinical model for discovery of molecular targets and compounds that may be useful for the treatment of addiction. One problem with performing intravenous drug self-administration studies in the mouse is the technical difficulty of maintaining catheter patency. Attrition rates in these experiments are high and can reach 40% or higher. Another general problem with drug self-administration is discerning which pharmacologically-induced effects of the reinforcer produce specific behaviors. For example, measurement of the reinforcing and neurological effects of psychostimulants can be confounded by their psychomotor effects. Operant methods using food reinforcement can avoid these pitfalls, although their utility in studying drug addiction is limited by the fact that some manipulations that alter drug self-administration have a minimal impact on food self-administration. For example, mesolimbic dopamine lesion or knockout of the D1 dopamine receptor reduce cocaine self-administration without having a significant impact on food self-administration. Sensory stimuli have been described for their ability to support operant responding as primary reinforcers (i.e. not conditioned reinforcers). Auditory and visual stimuli are self-administered by several species, although surprisingly little is known about the neural mechanisms underlying this reinforcement. The operant sensation seeking (OSS) model is a robust model for obtaining sensory self-administration in the mouse, allowing the study of neural mechanisms important in sensory reinforcement. An additional advantage of OSS is the ability to screen mutant mice for differences in operant behavior that may be relevant to addiction. We have reported that dopamine D1 receptor knockout mice, previously shown to be deficient in psychostimulant self-administration, also fail to acquire OSS. This is a unique finding in that these mice are capable of learning an operant task when food is used as a reinforcer. While operant studies using food reinforcement can be useful in the study of general motivated behavior and the mechanisms underlying food reinforcement, as mentioned above, these studies are limited in their application to studying molecular mechanisms of drug addiction. Thus, there may be similar neural substrates mediating sensory and psychostimulant reinforcement that are distinct from food reinforcement, which would make OSS a particularly attractive model for the study of drug addiction processes. The degree of overlap between other molecular targets of OSS and drug reinforcers is unclear, but is a topic that we are currently pursuing. While some aspects of addiction such as resistance to extinction may be observed with OSS, we have found that escalation is not observed in this model. Interestingly, escalation of intake and some other aspects of addiction are observed with self-administration of sucrose. Thus, when non-drug operant procedures are desired to study addiction-related processes, food or sensory reinforcers can be chosen to best fit the particular question being asked. In conclusion, both food self-administration and OSS in the mouse have the advantage of not requiring an intravenous catheter, which allows a higher throughput means to study the effects of pharmacological or genetic manipulation of neural targets involved in motivation. While operant testing using food as a reinforcer is particularly useful in the study of the regulation of food intake, OSS is particularly apt for studying reinforcement mechanisms of sensory stimuli and may have broad applicability to novelty seeking and addiction.
Collapse
Affiliation(s)
- Christopher M Olsen
- Department of Molecular Physiology and Biophysics, Center for Molecular Neuroscience, Kennedy Center for Human Development, Vanderbilt University Medical Center, USA.
| | | |
Collapse
|