1
|
Stea SG, Grisel JE. β-Endorphin influences sedative and ataxic effects of alcohol. Alcohol 2024; 115:69-77. [PMID: 37741556 DOI: 10.1016/j.alcohol.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
Beta-endorphin (β-E) is an opioid peptide linked to the behavioral effects of ethanol. For example, β-E provides negative feedback to inhibit the hypothalamic-pituitary-adrenal (HPA) stress axis, and neuroadaptation of this system to ethanol may facilitate sex differences in disordered drinking. Locomotor sensitivity to ethanol may also influence the risk for addiction; however, the role of β-E in psychomotor effects of ethanol is not fully understood. We examined the role of β-E and sex on locomotor effects of ethanol using adult male and female wild-type C57BL/6J and β-E deficient B6.129S2-Pomctm1Low/J mice in a parallel rod floor apparatus following 0.75 or 2.0 g/kg ethanol. Beginning 15 min after intraperitoneal injection, we recorded foot slips, distance traveled, slips per meter, first instance of immobility, and total time spent off-balance (lying on the floor) over 15 min, and collected blood for analysis of ethanol concentration 60 min after injection. Overall, β-E deficient mice were more sedated and ataxic following ethanol; at the lower dose they slipped more frequently and had a higher rate of slips per meter traveled. At the higher dose, β-E deficient mice were predominantly sedated, slipping less frequently, and traveling less, as well as spending more time off-balance and becoming immobile sooner. Genotype interacted with sex in that male β-E deficient mice slipped more frequently than their female counterparts, suggesting that β-E may elicit sex-dependent effects of ethanol-induced ataxia. Blood ethanol concentration did not differ between any group, suggesting that behavioral differences result from altered sensitivity to ethanol. Our data support the contention that β-E modulates the locomotor effects of ethanol and may influence ataxia in a sex-dependent manner. These findings help elucidate the role of β-E in diverging behavioral responses to ethanol and may aid the development of targeted treatments for alcohol use disorders.
Collapse
Affiliation(s)
- Samuel G Stea
- Department of Psychology & Neuroscience Program, Bucknell University, Lewisburg, PA 17837, United States
| | - Judith E Grisel
- Department of Psychology & Neuroscience Program, Bucknell University, Lewisburg, PA 17837, United States.
| |
Collapse
|
2
|
Little KM, Kosten TA. Focus on fentanyl in females: Sex and gender differences in the physiological and behavioral effects of fentanyl. Front Neuroendocrinol 2023; 71:101096. [PMID: 37597668 DOI: 10.1016/j.yfrne.2023.101096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
The prevalence of opioid use disorder and overdose continues to harm the U.S. population and is further exacerbated by the use of the synthetic opioid, fentanyl, and its analogs. Gender differences in the effects of fentanyl are not well understood. The present article reviews evidence for gender and sex differences in the physiological and behavioral effects of fentanyl in humans and animals. Biological sex seems to be a foundational driver in addiction vulnerability and affects mechanisms related to opioid use including fentanyl. Fentanyl has distinct pharmacodynamics and enhanced efficacy relative to other opioids that highlights the need to investigate how females may be uniquely altered by its use. Behavioral and physiological responses to fentanyl are found to differ by sex and gender in many cases, including outputs like affective symptoms, analgesia, tolerance, and withdrawal emphasizing the need for further research about the role of biological sex on fentanyl use.
Collapse
Affiliation(s)
- Kaitlyn M Little
- Department of Psychology, University of Houston, Houston, TX, United States
| | - Therese A Kosten
- Department of Psychology, University of Houston, Houston, TX, United States.
| |
Collapse
|
3
|
Hinds NM, Wojtas ID, Gallagher CA, Corbett CM, Manvich DF. Effects of sex and estrous cycle on intravenous oxycodone self-administration and the reinstatement of oxycodone-seeking behavior in rats. Front Behav Neurosci 2023; 17:1143373. [PMID: 37465001 PMCID: PMC10350507 DOI: 10.3389/fnbeh.2023.1143373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/09/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction The increasing misuse of both prescription and illicit opioids has culminated in a national healthcare crisis in the United States. Oxycodone is among the most widely prescribed and misused opioid pain relievers and has been associated with a high risk for transition to compulsive opioid use. Here, we sought to examine potential sex differences and estrous cycle-dependent effects on the reinforcing efficacy of oxycodone, as well as on stress-induced or cue-induced oxycodone-seeking behavior, using intravenous (IV) oxycodone self-administration and reinstatement procedures. Methods In experiment 1, adult male and female Long-Evans rats were trained to self-administer 0.03 mg/kg/inf oxycodone according to a fixed-ratio 1 schedule of reinforcement in daily 2-h sessions, and a dose-response function was subsequently determined (0.003-0.03 mg/kg/inf). In experiment 2, a separate group of adult male and female Long-Evans rats were trained to self-administer 0.03 mg/kg/inf oxycodone for 8 sessions, followed by 0.01 mg/kg/inf oxycodone for 10 sessions. Responding was then extinguished, followed by sequential footshock-induced and cue-induced reinstatement tests. Results In the dose-response experiment, oxycodone produced a typical inverted U-shape function with 0.01 mg/kg/inf representing the maximally effective dose in both sexes. No sex differences were detected in the reinforcing efficacy of oxycodone. In the second experiment, the reinforcing effects of 0.01-0.03 mg//kg/inf oxycodone were significantly attenuated in females during proestrus/estrus as compared to metestrus/diestrus phases of the estrous cycle. Neither males nor females displayed significant footshock-induced reinstatement of oxycodone seeking, but both sexes exhibited significant cue-induced reinstatement of oxycodone seeking at magnitudes that did not differ either by sex or by estrous cycle phase. Discussion These results confirm and extend previous work suggesting that sex does not robustly influence the primary reinforcing effects of oxycodone nor the reinstatement of oxycodone-seeking behavior. However, our findings reveal for the first time that the reinforcing efficacy of IV oxycodone varies across the estrous cycle in female rats.
Collapse
Affiliation(s)
- Nicole M. Hinds
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| | - Ireneusz D. Wojtas
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| | - Corinne A. Gallagher
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| | - Claire M. Corbett
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| | - Daniel F. Manvich
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| |
Collapse
|
4
|
Hinds NM, Wojtas ID, Gallagher CA, Corbett CM, Manvich DF. Effects of sex and estrous cycle on intravenous oxycodone self-administration and the reinstatement of oxycodone-seeking behavior in rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543393. [PMID: 37333293 PMCID: PMC10274722 DOI: 10.1101/2023.06.02.543393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The increasing misuse of both prescription and illicit opioids has culminated in a national healthcare crisis in the United States. Oxycodone is among the most widely prescribed and misused opioid pain relievers and has been associated with a high risk for transition to compulsive opioid use. Here, we sought to examine potential sex differences and estrous cycle-dependent effects on the reinforcing efficacy of oxycodone, as well as on stress-induced or cue-induced oxycodone-seeking behavior, using intravenous (IV) oxycodone self-administration and reinstatement procedures. In experiment 1, adult male and female Long-Evans rats were trained to self-administer 0.03 mg/kg/inf oxycodone according to a fixed-ratio 1 schedule of reinforcement in daily 2-hr sessions, and a dose-response function was subsequently determined (0.003-0.03 mg/kg/inf). In experiment 2, a separate group of adult male and female Long-Evans rats were trained to self-administer 0.03 mg/kg/inf oxycodone for 8 sessions, followed by 0.01 mg/kg/inf oxycodone for 10 sessions. Responding was then extinguished, followed by sequential footshock-induced and cue-induced reinstatement tests. In the dose-response experiment, oxycodone produced a typical inverted U-shape function with 0.01 mg/kg/inf representing the maximally effective dose in both sexes. No sex differences were detected in the reinforcing efficacy of oxycodone. In the second experiment, the reinforcing effects of 0.01-0.03 mg//kg/inf oxycodone were significantly attenuated in females during proestrus/estrus as compared to metestrus/diestrus phases of the estrous cycle. Neither males nor females displayed significant footshock-induced reinstatement of oxycodone seeking, but both sexes exhibited significant cue-induced reinstatement of oxycodone seeking at magnitudes that did not differ either by sex or by estrous cycle phase. These results confirm and extend previous work suggesting that sex does not robustly influence the primary reinforcing effects of oxycodone nor the reinstatement of oxycodone-seeking behavior. However, our findings reveal for the first time that the reinforcing efficacy of IV oxycodone varies across the estrous cycle in female rats.
Collapse
Affiliation(s)
- Nicole M. Hinds
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Ireneusz D. Wojtas
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Corinne A. Gallagher
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Claire M. Corbett
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Daniel F. Manvich
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| |
Collapse
|
5
|
Bossert JM, Mejias-Aponte CA, Saunders T, Altidor L, Emery M, Fredriksson I, Batista A, Claypool SM, Caldwell KE, Reiner DJ, Chow JJ, Foltz M, Kumar V, Seasholtz A, Hughes E, Filipiak W, Harvey BK, Richie CT, Vautier F, Gomez JL, Michaelides M, Kieffer BL, Watson SJ, Akil H, Shaham Y. Effect of Selective Lesions of Nucleus Accumbens µ-Opioid Receptor-Expressing Cells on Heroin Self-Administration in Male and Female Rats: A Study with Novel Oprm1-Cre Knock-in Rats. J Neurosci 2023; 43:1692-1713. [PMID: 36717230 PMCID: PMC10010456 DOI: 10.1523/jneurosci.2049-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/29/2022] [Accepted: 01/18/2023] [Indexed: 02/01/2023] Open
Abstract
The brain µ-opioid receptor (MOR) is critical for the analgesic, rewarding, and addictive effects of opioid drugs. However, in rat models of opioid-related behaviors, the circuit mechanisms of MOR-expressing cells are less known because of a lack of genetic tools to selectively manipulate them. We introduce a CRISPR-based Oprm1-Cre knock-in transgenic rat that provides cell type-specific genetic access to MOR-expressing cells. After performing anatomic and behavioral validation experiments, we used the Oprm1-Cre knock-in rats to study the involvement of NAc MOR-expressing cells in heroin self-administration in male and female rats. Using RNAscope, autoradiography, and FISH chain reaction (HCR-FISH), we found no differences in Oprm1 expression in NAc, dorsal striatum, and dorsal hippocampus, or MOR receptor density (except dorsal striatum) or function between Oprm1-Cre knock-in rats and wildtype littermates. HCR-FISH assay showed that iCre is highly coexpressed with Oprm1 (95%-98%). There were no genotype differences in pain responses, morphine analgesia and tolerance, heroin self-administration, and relapse-related behaviors. We used the Cre-dependent vector AAV1-EF1a-Flex-taCasp3-TEVP to lesion NAc MOR-expressing cells. We found that the lesions decreased acquisition of heroin self-administration in male Oprm1-Cre rats and had a stronger inhibitory effect on the effort to self-administer heroin in female Oprm1-Cre rats. The validation of an Oprm1-Cre knock-in rat enables new strategies for understanding the role of MOR-expressing cells in rat models of opioid addiction, pain-related behaviors, and other opioid-mediated functions. Our initial mechanistic study indicates that lesioning NAc MOR-expressing cells had different effects on heroin self-administration in male and female rats.SIGNIFICANCE STATEMENT The brain µ-opioid receptor (MOR) is critical for the analgesic, rewarding, and addictive effects of opioid drugs. However, in rat models of opioid-related behaviors, the circuit mechanisms of MOR-expressing cells are less known because of a lack of genetic tools to selectively manipulate them. We introduce a CRISPR-based Oprm1-Cre knock-in transgenic rat that provides cell type-specific genetic access to brain MOR-expressing cells. After performing anatomical and behavioral validation experiments, we used the Oprm1-Cre knock-in rats to show that lesioning NAc MOR-expressing cells had different effects on heroin self-administration in males and females. The new Oprm1-Cre rats can be used to study the role of brain MOR-expressing cells in animal models of opioid addiction, pain-related behaviors, and other opioid-mediated functions.
Collapse
Affiliation(s)
- Jennifer M Bossert
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Carlos A Mejias-Aponte
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | | | - Lindsay Altidor
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | | | - Ida Fredriksson
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Ashley Batista
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Sarah M Claypool
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Kiera E Caldwell
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - David J Reiner
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Jonathan J Chow
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | | | - Vivek Kumar
- University of Michigan, Ann Arbor, Michigan, 48104
| | | | | | | | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Christopher T Richie
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Francois Vautier
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Juan L Gomez
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Michael Michaelides
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Brigitte L Kieffer
- University of Strasbourg-Institut National de la Santé et de la Recherche Médicale U1114, Strasbourg, France, 67084
| | | | - Huda Akil
- University of Michigan, Ann Arbor, Michigan, 48104
| | - Yavin Shaham
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| |
Collapse
|
6
|
Craft RM. Burrowing as an index of inflammatory pain in male vs. female rats. Behav Pharmacol 2023; 34:55-67. [PMID: 36473021 DOI: 10.1097/fbp.0000000000000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The study objective was to determine whether burrowing behavior is useful as a functional index of pain in both male and female rats, and whether a 'no-training' protocol can be used to increase testing efficiency. Adult Sprague-Dawley rats were injected in one or both hindpaws with oil vehicle or complete Freund's adjuvant (CFA); starting the next day, the amount of gravel each rat burrowed out of a tube in 1 h was measured daily for ≤7 days. Without preliminary training on the burrowing procedure, CFA reliably suppressed burrowing for 2-3 days compared to controls, in both sexes. However, whereas unilateral CFA completely suppressed burrowing 1-day post-CFA in nearly all males, bilateral CFA was required to do so in females. When administered 30 min before testing, once daily for 5 days post-CFA, the nonsteroidal anti-inflammatory drug ketoprofen (0.01-3.2 mg/kg) and the opioid morphine (0.1-3.2 mg/kg) significantly increased CFA-suppressed burrowing, whereas the purported cannabinoid analgesic Δ 9 -tetrahydrocannabinol (0.01-2.0 mg/kg) did not. The benzodiazepine chlordiazepoxide (1.25-10 mg/kg), included as a 'true negative' control, also did not restore CFA-suppressed burrowing in either sex. However, in CFA-treated males only, chlordiazepoxide decreased burrowing, suggesting that anxiety may contribute to burrowing in males but not females that are in pain. Overall these results suggest that burrowing is a valid, functional index of inflammatory pain in both sexes, and training on the burrowing procedure is not necessary. However, females are more avid burrowers than males, which should be considered when both sexes are used in inflammatory pain testing.
Collapse
Affiliation(s)
- Rebecca M Craft
- Department of Psychology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
7
|
Bergum N, Berezin CT, King CM, Vigh J. µ-Opioid Receptors Expressed by Intrinsically Photosensitive Retinal Ganglion Cells Contribute to Morphine-Induced Behavioral Sensitization. Int J Mol Sci 2022; 23:15870. [PMID: 36555511 PMCID: PMC9781919 DOI: 10.3390/ijms232415870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Opioid drugs are the most effective tools for treating moderate to severe pain. Despite their analgesic efficacy, long-term opioid use can lead to drug tolerance, addiction, and sleep/wake disturbances. While the link between opioids and sleep/wake problems is well-documented, the mechanism underlying opioid-related sleep/wake problems remains largely unresolved. Importantly, intrinsically photosensitive retinal ganglion cells (ipRGCs), the cells that transmit environmental light/dark information to the brain's sleep/circadian centers to regulate sleep/wake behavior, express μ-opioid receptors (MORs). In this study, we explored the potential contribution of ipRGCs to opioid-related sleep/circadian disruptions. Using implanted telemetry transmitters, we measured changes in horizontal locomotor activity and body temperature in mice over the course of a chronic morphine paradigm. Mice lacking MORs expressed by ipRGCs (McKO) exhibited reduced morphine-induced behavioral activation/sensitization compared with control littermates with normal patterns of MOR expression. Contrastingly, mice lacking MORs globally (MKO) did not acquire morphine-induced locomotor activation/sensitization. Control mice also showed morphine-induced hypothermia in both the light and dark phases, while McKO littermates only exhibited morphine-induced hypothermia in the dark. Interestingly, only control animals appeared to acquire tolerance to morphine's hypothermic effect. Morphine, however, did not acutely decrease the body temperature of MKO mice. These findings support the idea that MORs expressed by ipRGCs could contribute to opioid-related sleep/wake problems and thermoregulatory changes.
Collapse
Affiliation(s)
- Nikolas Bergum
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Casey-Tyler Berezin
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA
| | - Connie M. King
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
8
|
Brynildsen JK, Yang K, Lemchi C, Dani JA, De Biasi M, Blendy JA. A common SNP in Chrna5 enhances morphine reward in female mice. Neuropharmacology 2022; 218:109218. [PMID: 35973602 DOI: 10.1016/j.neuropharm.2022.109218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022]
Abstract
The single nucleotide polymorphism (SNP) D398N (rs16969968) in CHRNA5, the gene encoding the α5 subunit of the nicotinic acetylcholine receptors (nAChR), has been associated with both nicotine and opiate dependence in human populations. Expression of this SNP on presynaptic VTA dopaminergic (DA) neurons is known to cause a reduction in calcium signaling, leading to alterations in transmitter signaling and altered responses to drugs of abuse. To examine the impact of the Chrna5 SNP on opiate reward and underlying dopaminergic mechanisms, mice harboring two copies of the risk-associated allele (Chrna5 A/A) at a location equivalent to human rs16969968 were generated via CRISPR/cas9 genome editing. We sought to determine whether Chrna5 A/A mice show differences in sensitivity to rewarding properties of morphine using the conditioned place preference paradigm. When mice were tested two weeks after conditioning, female Chrna5 A/A mice showed significantly enhanced preference for the morphine-paired chamber relative to WT females, suggesting that this genotype may enhance opioid reward specifically in females. In contrast, Chrna5 genotype had no effect on locomotor sensitization in male or female mice. Relative to WT females, peak amplitude of ACh-gated currents recorded from VTA DA neurons in Chrna5 A/A females was potentiated 1 day after conditioning with morphine. Increased FOS expression was also observed in Chrna5 A/A mice relative to WT mice following exposure to the morphine CPP chamber. We propose that impaired α5 nAChR subunit function alters DA neuron response following repeated morphine exposures, and that this early cellular response could contribute to enhanced opiate reward two weeks after conditioning.
Collapse
Affiliation(s)
| | | | - Crystal Lemchi
- Department of Systems Pharmacology and Translational Therapeutics, USA
| | | | - Mariella De Biasi
- Department of Neuroscience, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics, USA.
| |
Collapse
|
9
|
Cippitelli A, Martinez M, Zribi G, Cami-Kobeci G, Husbands SM, Toll L. PPL-138 (BU10038): A bifunctional NOP/mu partial agonist that reduces cocaine self-administration in rats. Neuropharmacology 2022; 211:109045. [PMID: 35378170 PMCID: PMC9074796 DOI: 10.1016/j.neuropharm.2022.109045] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/11/2022] [Accepted: 03/21/2022] [Indexed: 12/30/2022]
Abstract
The search for new and effective treatments for cocaine use disorder (CUD) is a priority. We determined whether PPL-138 (BU10038), a compound with partial agonist activity at both nociceptin opioid peptide (NOP) and mu-opioid receptors, reduces cocaine consumption, reinstatement, and whether the compound itself produces reinforcing effects in rats. Using an intermittent access (IntA) cocaine self-administration procedure, we found that PPL-138 (0.1 and 0.3 mg/kg) effectively decreased the total number of cocaine infusions and burst-like cocaine intake in both male and female rats. Responses for food in an IntA model of food self-administration were not altered for either sex, although locomotor activity was increased in female but not male rats. Blockade of NOP receptors with the selective antagonist J-113397 (5 mg/kg) did not prevent the PPL-138-induced suppression of cocaine self-administration, whereas blockade of mu-opioid receptors by naltrexone (1 mg/kg) reversed such effect. Consistently, treatment with morphine (1, 3, and 10 mg/kg) dose-dependently reduced IntA cocaine self-administration measures. PPL-138 also reduced reinstatement of cocaine seeking at all doses examined. Although an initial treatment with PPL-138 (2.5, 10, and 40 μg/kg/infusion) appeared rewarding, the compound did not maintain self-administration behavior. Animals treated with PPL-138 showed initial suppression of cocaine self-administration, which was eliminated following repeated daily dosing. However, suppression of cocaine self-administration was retained when subsequent PPL-138 treatments were administered 48 h apart. These findings demonstrate that the approach of combining partial NOP/mu-opioid activation successfully reduces cocaine use, but properties of PPL-138 seem to depend on the timing of drug administration.
Collapse
Affiliation(s)
- Andrea Cippitelli
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States.
| | - Madeline Martinez
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Gilles Zribi
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Gerta Cami-Kobeci
- School of Life Sciences, Faculty of Creative Arts, Technologies & Science, University of Bedfordshire Luton, Luton, UK
| | | | - Lawrence Toll
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
10
|
Sharp JL, Pearson T, Smith MA. Sex differences in opioid receptor mediated effects: Role of androgens. Neurosci Biobehav Rev 2022; 134:104522. [PMID: 34995646 PMCID: PMC8872632 DOI: 10.1016/j.neubiorev.2022.104522] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/19/2021] [Accepted: 01/02/2022] [Indexed: 12/26/2022]
Abstract
An abundance of data indicates there are sex differences in endogenous opioid peptides and opioid receptors, leading to functional differences in sensitivity to opioid receptor mediated behaviors between males and females. Many of these sex differences are mediated by the effects of gonadal hormones on the endogenous opioid system. Whereas much research has examined the role of ovarian hormones on opioid receptor mediated endpoints, comparatively less research has examined the role of androgens. This review describes what is currently known regarding the influence of androgens on opioid receptor mediated endpoints and how androgens may contribute to sex differences in these effects. The review also addresses the clinical implications of androgenic modulation of opioid receptor mediated behaviors and suggests future lines of research for preclinical and clinical investigators. We conclude that further investigation into androgenic modulation of opioid receptor mediated effects may lead to new options for addressing conditions such as chronic pain and substance use disorders.
Collapse
Affiliation(s)
- Jessica L Sharp
- Department of Psychology and Program in Neuroscience, Davidson College, United States
| | - Tallia Pearson
- Department of Psychology and Program in Neuroscience, Davidson College, United States
| | - Mark A Smith
- Department of Psychology and Program in Neuroscience, Davidson College, United States.
| |
Collapse
|
11
|
Effects of neonatal dopaminergic lesion on oral cocaine self-administration in rats: Higher female vulnerability to cocaine consumption. Pharmacol Biochem Behav 2021; 212:173315. [PMID: 34942237 DOI: 10.1016/j.pbb.2021.173315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022]
Abstract
The dopaminergic system is associated with cocaine-seeking behaviors, being influenced by other neurotransmitters such as GABA and deregulated by chronic cocaine self-administration. Administration of 6-hydroxydopamine (6-OHDA) to neonatal rats produces a depletion of brain dopamine, mainly, that results in behavioral alterations in adulthood. This model can be applied to better understanding of the role of the dopaminergic system in cocaine use and how its behavioral effects can modulate drug intake. Though there are well-established sex differences in the pattern of drug use, there are no published studies investigating sex-dependent effects of neonatal lesions with 6-OHDA on cocaine self-administration nor regarding GABAA receptor (GABAAR) subunits expression. Herein, neurotoxic lesion was induced in male and female neonatal rats by intracisternal injection of 6-OHDA at PND 4, and locomotion was evaluated before and after cocaine self-administration. Cocaine was diluted in a sweet solution (sucrose 1.5%) and offered for 27 consecutive 3-h daily sessions via a dispenser for oral intake, in an operant chamber under a fixed-ratio 1 (FR1) schedule. The 6-OHDA lesion reduced oral cocaine self-administration in male and female rats. Female rats, independent of dopaminergic condition, consumed more cocaine-containing solution than sucrose-only solution. Furthermore, as expected, 6-OHDA-lesioned animals presented a higher basal locomotor activity when compared to sham rats. We evaluated GABAAR subunit expression and found no statistically significant differences between rats that self-administered a sucrose-only solution and those that self-administered a cocaine-containing solution. Even when the reward system is depleted, some behavioral differences remain in females, providing more data that highlight the female vulnerability to cocaine consumption.
Collapse
|
12
|
Suslov EV, Ponomarev KY, Volcho KP, Salakhutdinov NF. Azaadamantanes, a New Promising Scaffold for Medical Chemistry. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2021; 47:1133-1154. [PMID: 34931112 PMCID: PMC8675118 DOI: 10.1134/s1068162021060236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/10/2021] [Accepted: 03/14/2021] [Indexed: 12/25/2022]
Abstract
Azaadamantanes are nitrogen-containing analogs of adamantane, which contain one or more nitrogen atoms instead of carbon atoms. This substitution leads to several specific chemical and physical properties. The azaadamantane derivatives have less lipophilicity compared to their adamantane analogs, which affects both their interaction with biological targets and bioavailability. The significant increase in the number of publications during the last decade (2009-2020) concerning the study of reactivity and biological activity of azaadamantanes and their derivatives indicates a great theoretical and practical interest in these compounds. Compounds with pronounced biological activity have been already discovered among azaadamantane derivatives. The review is devoted to the biological activity of azaadamantanes and their derivatives. It presents the main methods for the synthesis of di- and triazaadamantanes and summarizes the accumulated data on studying the biological activity of these compounds. The prospects for the use of azaadamantanes in medical chemistry and pharmacology are discussed.
Collapse
Affiliation(s)
- E. V. Suslov
- Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - K. Yu. Ponomarev
- Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - K. P. Volcho
- Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - N. F. Salakhutdinov
- Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
13
|
Sharp JL, Ethridge SB, Ballard SL, Potter KM, Schmidt KT, Smith MA. The effects of chronic estradiol treatment on opioid self-administration in intact female rats. Drug Alcohol Depend 2021; 225:108816. [PMID: 34171824 PMCID: PMC8282761 DOI: 10.1016/j.drugalcdep.2021.108816] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/07/2021] [Accepted: 04/22/2021] [Indexed: 01/24/2023]
Abstract
Heroin intake decreases significantly during proestrus in normally cycling female rats, and this effect is mediated by endogenous estradiol but not endogenous progesterone. The purpose of this study was to determine whether chronic administration of exogenous estradiol decreases intake of the semi-synthetic opioid, heroin, and the fully synthetic opioid, remifentanil, in intact female rats. Normally cycling female rats were implanted with intravenous catheters and trained to self-administer heroin on a fixed ratio (FR1) schedule of reinforcement. Rats were treated chronically with daily administration of either a low dose of estradiol (0.5 mcg, sc), a high dose of estradiol (5.0 mcg, sc), or vehicle (peanut oil, sc). After two weeks of heroin self-administration training, dose-effect curves were determined for both heroin and remifentanil. Chronic administration of estradiol non-significantly decreased heroin intake and significantly decreased remifentanil intake. Estradiol-induced decreases in remifentanil intake were dose-dependent, characterized by large effect sizes, and greatest in rats treated with the high dose of estradiol. These data indicate that chronic estradiol administration decreases opioid intake in intact female rats with medium to large effect sizes across opioids. These findings suggest that estrogen-based pharmacotherapies may represent a novel treatment approach for women with opioid use disorder.
Collapse
Affiliation(s)
- Jessica L Sharp
- Department of Psychology, Davidson College, Davidson, NC, USA
| | | | | | - Kenzie M Potter
- Department of Psychology, Davidson College, Davidson, NC, USA
| | - Karl T Schmidt
- Department of Psychology, Davidson College, Davidson, NC, USA
| | - Mark A Smith
- Department of Psychology, Davidson College, Davidson, NC, USA.
| |
Collapse
|
14
|
Sex dependent alteration of epigenetic marks after chronic morphine treatment in mice organs. Food Chem Toxicol 2021; 152:112200. [PMID: 33891991 DOI: 10.1016/j.fct.2021.112200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/20/2021] [Accepted: 04/07/2021] [Indexed: 11/23/2022]
Abstract
Epigenetic marks may be also affected by several factors, such as age, lifestyle, early life experiences and exposure to chemicals or drugs, such as opioids. Previous studies have focused on how morphine epigenetically regulates different regions of the brain that are implicated in tolerance, dependence and other psychiatric disorders more related to the physio-pathological effects of opioids. Nevertheless, a significant knowledge gap remains regarding the effect of chronic treatment on other organs and biological systems. Therefore, the aim of this work is to increase our knowledge about the impact of chronic morphine exposure on DNA methylation and histone modification levels in each of the organs of male and female model mice in vivo. Our results reveal, for the first time, that chronic morphine treatment induced changes in DNA methylation/hydroxymethylation and histone modification in-vivo at the systemic level, revealing a potential physiological effect on the regulation of gene expression. Notably, morphine-induced epigenetic modification occurs in a sex-dependent manner, revealing the existence of different underlying mechanisms of epigenetic modification in male and female mice.
Collapse
|
15
|
Co-administration of nalbuphine attenuates the morphine-induced anxiety and dopaminergic alterations in morphine-withdrawn rats. Psychopharmacology (Berl) 2021; 238:1193-1211. [PMID: 33655408 DOI: 10.1007/s00213-021-05765-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
Abstract
INTRODUCTION The classical effects of exogenous opioids, such as morphine, are predominantly mediated through μ-opioid receptors. The chronic use of morphine induces anxiety-like behavior causing functional changes in the mesolimbic dopaminergic system. The mixed μ/κ-agonist, nalbuphine, used either as an analgesic or as an adjuvant with morphine, produces different and opposite effects. However, whether nalbuphine can be used to antagonize morphine-induced anxiety and dopaminergic alterations is not fully known. OBJECTIVE This study aimed to compare acute and chronic effects of nalbuphine on morphine-induced anxiety and dopaminergic alterations in rats. METHODS Male adult Wistar albino rats were made opioid-dependent by administering increasing doses of morphine (5-25 mg/kg; i.p.; b.i.d.). Withdrawal was induced by naloxone (1 mg/kg, i.p.), 4 h after the last morphine injection. Anxiety-like behavior was measured using Activity Monitor (Coulbourn Instruments, Inc. USA). Thereafter, the animals were sacrificed and the brain dissected out and the level of cAMP and the transcriptional and translational expression of TH was measured. Nalbuphine was co-administered with morphine, acutely and chronically, at various doses (0.1, 0.3, 1.0, 3.0 mg/kg, i.p.). RESULTS Morphine-dependent rats showed a significant higher anxiety and cAMP levels and a significant decrease in the expression of TH. Co-administration of chronic doses of nalbuphine attenuates the higher anxiety, cAMP levels, and upregulates the TH expressions; however, the acute nalbuphine treatment does not attenuate the morphine-induced side effects. CONCLUSION Therefore, nalbuphine might have an important role in attenuating the anxiety and the effects of the dopaminergic pathway and may have potential in the treatment of opioid addiction.
Collapse
|
16
|
Andrade AK, Renda B, Sharivker M, Lambert K, Murray JE. Sex differences in the discriminative stimulus characteristics of a morphine occasion setter in rats. Pharmacol Biochem Behav 2021; 205:173173. [PMID: 33753118 DOI: 10.1016/j.pbb.2021.173173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/19/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
The current study investigated whether the stimulus effects of morphine can function as a positive and negative feature in a Pavlovian occasion setting drug discrimination preparation in male and female rats. Sprague-Dawley rats were assigned to a feature positive (FP) or feature negative (FN) training group and all received intermixed morphine (3.2 mg/kg, IP) or saline injections 15 min before 20-min daily training sessions. For FP rats, on morphine sessions, each of eight 15-s white noise (WN) presentations was followed by 4-s access to sucrose (0.01 ml, 26% w/v); on saline sessions, sucrose was withheld. FN rats learned the reverse contingency. FP discrimination was acquired somewhat sooner than FN discrimination, and females, but not males, became sensitized to the locomotor effects of morphine, which did not influence conditioned responding. Rats then entered dose generalization testing. There was no sex difference in dose generalization for FN groups (ED50 1.26 for males and 1.57 for females). Yet for FP rats, the dose response curve for females was shifted to the right compared to males (ED50 0.54 for males and 1.94 for females). FP females exhibited enhanced responding at a dose higher than that of their original training. These findings reveal the need to reassess our notions of drug stimuli that guide appropriate associative behaviours from the perspective of sex differences.
Collapse
Affiliation(s)
- Allyson K Andrade
- Department of Psychology, University of Guelph, Guelph, ON, Canada; Collaborative Neurosciences Graduate Program, University of Guelph, Guelph, ON, Canada
| | - Briana Renda
- Department of Psychology, University of Guelph, Guelph, ON, Canada; Collaborative Neurosciences Graduate Program, University of Guelph, Guelph, ON, Canada
| | - Michael Sharivker
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Karlie Lambert
- Department of Psychology, University of Guelph, Guelph, ON, Canada
| | - Jennifer E Murray
- Department of Psychology, University of Guelph, Guelph, ON, Canada; Collaborative Neurosciences Graduate Program, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
17
|
Zebrafish early life stages as alternative model to study 'designer drugs': Concordance with mammals in response to opioids. Toxicol Appl Pharmacol 2021; 419:115483. [PMID: 33722667 DOI: 10.1016/j.taap.2021.115483] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023]
Abstract
The number of new psychoactive substances (NPS) on the illicit drug market increases fast, posing a need to urgently understand their toxicity and behavioural effects. However, with currently available rodent models, NPS assessment is limited to a few substances per year. Therefore, zebrafish (Danio rerio) embryos and larvae have been suggested as an alternative model that would require less time and resources to perform an initial assessment and could help to prioritize substances for subsequent evaluation in rodents. To validate this model, more information on the concordance of zebrafish larvae and mammalian responses to specific classes of NPS is needed. Here, we studied toxicity and behavioural effects of opioids in zebrafish early life stages. Synthetic opioids are a class of NPS that are often used in pain medication but also frequently abused, having caused multiple intoxications and fatalities recently. Our data shows that fentanyl derivatives were the most toxic among the tested opioids, with toxicity in the zebrafish embryo toxicity test decreasing in the following order: butyrfentanyl>3-methylfentanyl>fentanyl>tramadol> O-desmethyltramadol>morphine. Similar to rodents, tramadol as well as fentanyl and its derivatives led to hypoactive behaviour in zebrafish larvae, with 3-methylfentanyl being the most potent. Physico-chemical properties-based predictions of chemicals' uptake into zebrafish embryos and larvae correlated well with the effects observed. Further, the biotransformation pattern of butyrfentanyl in zebrafish larvae was reminiscent of that in humans. Comparison of toxicity and behavioural responses to opioids in zebrafish and rodents supports zebrafish as a suitable alternative model for rapidly testing synthetic opioids.
Collapse
|
18
|
Bossert JM, Kiyatkin E, Korah H, Hoots JK, Afzal A, Perekopskiy D, Thomas S, Fredriksson I, Blough BE, Negus SS, Epstein DH, Shaham Y. In a Rat Model of Opioid Maintenance, the G Protein-Biased Mu Opioid Receptor Agonist TRV130 Decreases Relapse to Oxycodone Seeking and Taking and Prevents Oxycodone-Induced Brain Hypoxia. Biol Psychiatry 2020; 88:935-944. [PMID: 32305216 PMCID: PMC7483192 DOI: 10.1016/j.biopsych.2020.02.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Maintenance treatment with opioid agonists (buprenorphine, methadone) is effective for opioid addiction but does not eliminate opioid use in all patients. We modeled maintenance treatment in rats that self-administered the prescription opioid oxycodone. The maintenance medication was either buprenorphine or the G protein-biased mu opioid receptor agonist TRV130. We then tested prevention of oxycodone seeking and taking during abstinence using a modified context-induced reinstatement procedure, a rat relapse model. METHODS We trained rats to self-administer oxycodone (6 hours/day, 14 days) in context A; infusions were paired with discrete tone-light cues. We then implanted osmotic pumps containing buprenorphine or TRV130 (0, 3, 6, or 9 mg/kg/day) and performed 3 consecutive tests: lever pressing reinforced by oxycodone-associated discrete cues in nondrug context B (extinction responding), context-induced reinstatement of oxycodone seeking in context A, and reacquisition of oxycodone self-administration in context A. We also tested whether TRV130 maintenance would protect against acute oxycodone-induced decreases in nucleus accumbens oxygen levels. RESULTS In male rats, buprenorphine and TRV130 decreased extinction responding and reacquisition of oxycodone self-administration but had a weaker (nonsignificant) effect on context-induced reinstatement. In female rats, buprenorphine decreased responding in all 3 tests, while TRV130 decreased only extinction responding. In both sexes, TRV130 prevented acute brain hypoxia induced by moderate doses of oxycodone. CONCLUSIONS TRV130 decreased oxycodone seeking and taking during abstinence in a partly sex-specific manner and prevented acute oxycodone-induced brain hypoxia. We propose that G protein-biased mu opioid receptor agonists, currently in development as analgesics, should be considered as relapse prevention maintenance treatment for opioid addiction.
Collapse
Affiliation(s)
- Jennifer M. Bossert
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A.,Correspondence: Jennifer M. Bossert ()
| | - Eugene Kiyatkin
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Hannah Korah
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | | | - Anum Afzal
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | | | - Shruthi Thomas
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Ida Fredriksson
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Bruce E. Blough
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC, U.S.A
| | - S. Stevens Negus
- Dept. of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - David H. Epstein
- Clinical Pharmacology and Therapeutics Research Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Yavin Shaham
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| |
Collapse
|
19
|
Bryant CD, Healy AF, Ruan QT, Coehlo MA, Lustig E, Yazdani N, Luttik KP, Tran T, Swancy I, Brewin LW, Chen MM, Szumlinski KK. Sex‐dependent effects of an
Hnrnph1
mutation on fentanyl addiction‐relevant behaviors but not antinociception in mice. GENES BRAIN AND BEHAVIOR 2020; 20:e12711. [DOI: 10.1111/gbb.12711] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Camron D. Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry Boston University School of Medicine Boston Massachusetts USA
| | - Aidan F. Healy
- Department of Psychological and Brain Sciences University of California Santa Barbara California USA
| | - Qiu T. Ruan
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry Boston University School of Medicine Boston Massachusetts USA
- T32 Biomolecular Pharmacology Ph.D. Program Boston University School of Medicine Boston Massachusetts USA
- Transformative Training Program in Addiction Science Boston University Boston Massachusetts USA
| | - Michal A. Coehlo
- Department of Psychological and Brain Sciences University of California Santa Barbara California USA
| | - Elijah Lustig
- Department of Psychological and Brain Sciences University of California Santa Barbara California USA
| | - Neema Yazdani
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry Boston University School of Medicine Boston Massachusetts USA
- T32 Biomolecular Pharmacology Ph.D. Program Boston University School of Medicine Boston Massachusetts USA
- Transformative Training Program in Addiction Science Boston University Boston Massachusetts USA
| | - Kimberly P. Luttik
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry Boston University School of Medicine Boston Massachusetts USA
- Undergraduate Research Opportunity Program (UROP) Boston University Boston Massachusetts USA
| | - Tori Tran
- Department of Psychological and Brain Sciences University of California Santa Barbara California USA
| | - Isaiah Swancy
- Department of Psychological and Brain Sciences University of California Santa Barbara California USA
| | - Lindsey W. Brewin
- Department of Psychological and Brain Sciences University of California Santa Barbara California USA
| | - Melanie M. Chen
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry Boston University School of Medicine Boston Massachusetts USA
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences University of California Santa Barbara California USA
- Department of Molecular, Developmental and Cellular Biology and the Neuroscience Research Institute University of California Santa Barbara California USA
| |
Collapse
|
20
|
Gutierrez A, Creehan KM, Taffe MA. A vapor exposure method for delivering heroin alters nociception, body temperature and spontaneous activity in female and male rats. J Neurosci Methods 2020; 348:108993. [PMID: 33130050 DOI: 10.1016/j.jneumeth.2020.108993] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/02/2020] [Accepted: 10/23/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND The ongoing crisis related to non-medical use of opioids makes it of continued importance to understand the risk factors for opioid addiction, the behavioral and neurobiological consequences of opioid exposure and to seek potential avenues for therapy. Pre-clinical rodent models have been critical to advancing understanding of opioid consequences for decades, but have been mostly limited to drug delivery by injection or by oral dosing. Inhalation, a significant route for many human users, has not been as well-established. METHOD We adapted an e-cigarette based exposure system, previously shown efficacious for delivery of other drugs to rats, to deliver heroin vapor. Effectsin vivo were assessed in male and female Sprague-Dawley rats using a warm-water assay for anti-nociception and an implanted radiotelemetry system for evaluating changes in body temperature and spontaneous activity rate. RESULTS Inhalation of vapor created by heroin 100 mg/mL in the propylene glycol (PG) vehicle significantly slowed tail-withdrawal from a 52 °C water bath, bi-phasically altered activity, and increased temperature in male and female rats. Inhalation of heroin 50 mg/mL for 15 min produced significant effects, as the lower bound on efficacy, whereas inhalation of heroin 100 mg/mL for 30 min produced robust effects across all endpoints and groups. CONCLUSIONS This work shows that e-cigarette devices deliver psychoactive doses of heroin to rats, using concentrations of ∼50-100 mg/mL and inhalation durations of 15-30 min. This technique may be useful to assess the health consequences of inhaled heroin and other opioid drugs.
Collapse
Affiliation(s)
- Arnold Gutierrez
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Kevin M Creehan
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Michael A Taffe
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
21
|
Lopresti NM, Esguerra M, Mermelstein PG. Sex Differences in Animal Models of Opioid Reward. CURRENT SEXUAL HEALTH REPORTS 2020; 12:186-194. [PMID: 33574737 PMCID: PMC7872138 DOI: 10.1007/s11930-020-00266-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW This review aims to discuss sex differences observed in preclinical rodent models of opioid reward. RECENT FINDINGS Utilizing a variety of methodological approaches and drug regimens, no clear consensus has emerged regarding the effects of opiates between males and females. This is quite dissimilar to work examining psychostimulants, where female animals reliably exhibit stronger behavioral responses. SUMMARY With opioid research quickly expanding to determine the neural underpinnings of opioid addiction, further research is essential to determine the conditions wherein sex differences may occur and how they may relate to the human condition.
Collapse
Affiliation(s)
| | - Manuel Esguerra
- University of Minnesota, Twin Cities, Department of Neuroscience
| | | |
Collapse
|
22
|
Caporoso J, Moses M, Koper K, Tillman TS, Jiang L, Brandon N, Chen Q, Tang P, Xu Y. A Thermal Place Preference Test for Discovery of Neuropathic Pain Drugs. ACS Chem Neurosci 2020; 11:1006-1012. [PMID: 32191433 DOI: 10.1021/acschemneuro.0c00013] [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] [Indexed: 12/25/2022] Open
Abstract
Developing potent non-opioid pain medications is an integral part of the battle to conquer both chronic pain and the current opioid crisis. Although most screening approaches use in vitro surrogate targets, in vivo screening of analgesic candidates is a necessary preclinical step in drug discovery. Here, we report the design of a new automated behavioral testing apparatus based on the principle of a thermal place preference test (TPPT). This new design can detect, quantify, and differentiate behavioral responses to cold stimuli between sham and chronic constriction injury (CCI) rodents with up to 12 animals tested simultaneously. At an optimized temperature pair of 12.5 °C vs 30.0 °C (±0.5 °C), the TPPT design has captured the antinociceptive effects of morphine and pregabalin on CCI rats in individual 10 min tests. Moreover, it can differentiate analgesic effects by morphine or pregabalin from anxiolytic effects by diazepam. The results, along with the relatively low cost to construct the apparatus and moderately high throughput, make our TPPT design applicable for behavioral studies of chronic pain in rodents and for high-throughput in vivo screening of the next generation of pain medications.
Collapse
Affiliation(s)
- Joel Caporoso
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Mark Moses
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Kerryann Koper
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Tommy S. Tillman
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Lingling Jiang
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Nicole Brandon
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Qiang Chen
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Pei Tang
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Yan Xu
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
23
|
Datta U, Schoenrock SE, Bubier JA, Bogue MA, Jentsch JD, Logan RW, Tarantino LM, Chesler EJ. Prospects for finding the mechanisms of sex differences in addiction with human and model organism genetic analysis. GENES, BRAIN, AND BEHAVIOR 2020; 19:e12645. [PMID: 32012419 PMCID: PMC7060801 DOI: 10.1111/gbb.12645] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Despite substantial evidence for sex differences in addiction epidemiology, addiction-relevant behaviors and associated neurobiological phenomena, the mechanisms and implications of these differences remain unknown. Genetic analysis in model organism is a potentially powerful and effective means of discovering the mechanisms that underlie sex differences in addiction. Human genetic studies are beginning to show precise risk variants that influence the mechanisms of addiction but typically lack sufficient power or neurobiological mechanistic access, particularly for the discovery of the mechanisms that underlie sex differences. Our thesis in this review is that genetic variation in model organisms are a promising approach that can complement these investigations to show the biological mechanisms that underlie sex differences in addiction.
Collapse
Affiliation(s)
- Udita Datta
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| | - Sarah E. Schoenrock
- Center for Systems Neurogenetics of Addiction, Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Jason A. Bubier
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| | - Molly A. Bogue
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| | - James D. Jentsch
- Center for Systems Neurogenetics of Addiction, PsychologyState University of New York at BinghamtonBinghamtonNew York
| | - Ryan W. Logan
- Center for Systems Neurogenetics of Addiction, PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Lisa M. Tarantino
- Center for Systems Neurogenetics of Addiction, Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Elissa J. Chesler
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| |
Collapse
|
24
|
Mansouri MT, Fidler JA, Meng QC, Eckenhoff RG, García PS. Sex effects on behavioral markers of emergence from propofol and isoflurane anesthesia in rats. Behav Brain Res 2019; 367:59-67. [PMID: 30898682 DOI: 10.1016/j.bbr.2019.03.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 01/18/2023]
Abstract
Clinical studies have demonstrated sex-related differences in recovery from surgical anesthesia. This study aimed to characterize the emergence pattern following two anesthesia regimens in both sexes of rats. We considered six different markers of emergence from anesthesia: sigh, eye blinking, forelimb movement, mastication, neck extension, and recovery of the righting reflex (RORR). Spontaneous motor activity 24 h after the anesthesia induction was also examined. Our results showed that the rank order of the emergence latency after intraperitoneal propofol, PRO, exposure was forelimb movement < sigh < blink < mastication < neck extension < RORR, while after inhaled isoflurane, ISO, anesthesia the sequence was changed as sigh < blink < mastication < forelimb movement < neck extension < RORR in both male and female rats. Moreover, the latency to emergence after PRO in female rats was significantly higher than male rats, although following ISO there was no difference between the sexes (P < 0.001; P > 0.05, respectively). Open-field testing revealed no difference in PRO and ISO spontaneous locomotor activity due to drug administration (P > 0.05). These two anesthetics presented different emergence sequences. Although clinical data suggests that females arouse faster than males from anesthesia with propofol, our intraperitoneal technique in a rodent model had the opposite effect. Pharmacokinetic analysis demonstrated increased absorption of injected propofol for the female rats in our study, emphasizing the role of sexual dimorphism in drug distribution in rodents. Despite these pharmacokinetic differences, the pharmacodynamic effects of the drugs were remarkably consistent among both sexes through emergence.
Collapse
Affiliation(s)
- Mohammed Taghi Mansouri
- Neuroanesthesia Laboratory, Atlanta VA Medical Center/Emory University, Atlanta, GA, USA; Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Jonathan A Fidler
- Neuroanesthesia Laboratory, Atlanta VA Medical Center/Emory University, Atlanta, GA, USA; Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Qing Cheng Meng
- Department of Anesthesiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Roderic G Eckenhoff
- Department of Anesthesiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul S García
- Neuroanesthesia Laboratory, Atlanta VA Medical Center/Emory University, Atlanta, GA, USA; Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
25
|
Pedrón VT, Varani AP, Bettler B, Balerio GN. GABA B receptors modulate morphine antinociception: Pharmacological and genetic approaches. Pharmacol Biochem Behav 2019; 180:11-21. [PMID: 30851293 DOI: 10.1016/j.pbb.2019.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 10/27/2022]
Abstract
Previous studies in our laboratory showed an interaction between the GABAergic and opioid systems involved in the analgesic effect of baclofen (BAC). Furthermore, it is known that sex differences exist regarding various pharmacological responses of morphine (MOR) and they are related to an increased sensitivity to MOR effects in males. The aims of the present study were to evaluate the possible involvement of the GABAB receptors in the antinociceptive responses induced by MOR (1, 3 and 9 mg/kg, s.c.) administration using both pharmacological (BAC 2 mg/kg, i.p.; and 2-OH-saclofen, SAC 0.3 mg/kg, intra cisterna magna) and genetic approaches (GABAB1 knockout mice; GABAB1 KO) in mice of both sexes. In addition, we explored the alterations in c-Fos expression of different brain areas involved in the antinociceptive effect of MOR using both approaches. The pharmacological approach showed a higher dose-dependent antinociceptive effect of MOR in male mice compared to female mice. BAC and SAC pretreatment potentiated and attenuated the antinociceptive effect of MOR, respectively, in both sexes. The genetic approach revealed a dose-dependent antinociceptive effect of MOR in the wild type mice, but not in the GABAB1 KO mice and no sex differences were observed. Additionally, BAC and SAC pretreatment and the lack of GABAB1 subunit of the GABAB receptor prevented the changes observed in c-Fos expression in the cingulate cortex and nucleus accumbens of male mice. Our results suggest that the GABAB receptors are involved in the MOR antinociceptive effect of both male and female mice.
Collapse
Affiliation(s)
- Valeria T Pedrón
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Farmacológicas (ININFA), Buenos Aires, Argentina
| | - Andrés P Varani
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Farmacológicas (ININFA), Buenos Aires, Argentina
| | - Bernhard Bettler
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland
| | - Graciela N Balerio
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Farmacológicas (ININFA), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Buenos Aires, Argentina.
| |
Collapse
|
26
|
Peecher DL, Binder AK, Gabriel KI. Rodent models of mental illness in polycystic ovary syndrome: the potential role of hypothalamic-pituitary-adrenal dysregulation and lessons for behavioral researchers. Biol Reprod 2019; 100:590-600. [PMID: 30388193 DOI: 10.1093/biolre/ioy233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/04/2018] [Accepted: 10/31/2018] [Indexed: 12/20/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most commonly diagnosed endocrine disorder in women of reproductive age, with phenotypes including ovarian and metabolic dysfunctions. Women with PCOS also show increased rates of mental illness, dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, and altered responsiveness to stressors that may contribute to the higher rates of mental illness, specifically depression and anxiety. Animal models of PCOS have provided insight into the ovarian and metabolic mechanisms that underlie the syndrome, and several models have been used to study the behavioral consequences associated with PCOS in the laboratory. Several studies in rodent models of PCOS demonstrate changes in anxiety-like behavior, but researchers often neglect to report procedural details or behavioral data crucial to interpreting the differences observed in those studies. Additionally, the impact of potential HPA dysregulation in animal models of PCOS may influence behavioral findings, although only three studies to date have examined this. As such, researchers should consider and report stress-associated variables (e.g., time of day, light/dark cycle, light intensity, housing, and procedures to control experimenter and litter effects) that may influence depression- and anxiety-like behaviors in rodents. This review will summarize the behavioral and HPA-related studies in women with PCOS and rodent models of the disease, and provide considerations for future studies.
Collapse
Affiliation(s)
- Danielle L Peecher
- Department of Psychology, Central Washington University, Ellensburg, Washington, USA.,Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - April K Binder
- Center for Reproductive Biology, Washington State University, Pullman, Washington, USA.,Department of Biological Sciences, Central Washington University, Ellensburg, Washington, USA
| | - Kara I Gabriel
- Department of Psychology, Central Washington University, Ellensburg, Washington, USA
| |
Collapse
|
27
|
Ryan JD, Zhou Y, Contoreggi NH, Bshesh FK, Gray JD, Kogan JF, Ben KT, McEwen BS, Jeanne Kreek M, Milner TA. Sex Differences in the Rat Hippocampal Opioid System After Oxycodone Conditioned Place Preference. Neuroscience 2018; 393:236-257. [PMID: 30316908 PMCID: PMC6246823 DOI: 10.1016/j.neuroscience.2018.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/10/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022]
Abstract
Although opioid addiction has risen dramatically, the role of gender in addiction has been difficult to elucidate. We previously found sex-dependent differences in the hippocampal opioid system of Sprague-Dawley rats that may promote associative learning relevant to drug abuse. The present studies show that although female and male rats acquired conditioned place preference (CPP) to the mu-opioid receptor (MOR) agonist oxycodone (3 mg/kg, I.P.), hippocampal opioid circuits were differentially altered. In CA3, Leu-Enkephalin-containing mossy fibers had elevated levels in oxycodone CPP (Oxy) males comparable to those in females and sprouted in Oxy-females, suggesting different mechanisms for enhancing opioid sensitivity. Electron microscopy revealed that in Oxy-males delta opioid receptors (DORs) redistributed to mossy fiber-CA3 synapses in a manner resembling females that we previously showed is important for opioid-mediated long-term potentiation. Moreover, in Oxy-females DORs redistributed to CA3 pyramidal cell spines, suggesting the potential for enhanced plasticity processes. In Saline-injected (Sal) females, dentate hilar parvalbumin-containing basket interneuron dendrites had fewer MORs, however plasmalemmal and total MORs increased in Oxy-females. In dentate hilar GABAergic dendrites that contain neuropeptide Y, Sal-females compared to Sal-males had higher plasmalemmal DORs, and near-plasmalemmal DORs increased in Oxy-females. This redistribution of MORs and DORs within hilar interneurons in Oxy-females would potentially enhance disinhibition of granule cells via two different circuits. Together, these results indicate that oxycodone CPP induces sex-dependent redistributions of opioid receptors in hippocampal circuits in a manner facilitating opioid-associative learning processes and may help explain the increased susceptibility of females to opioid addiction acquisition and relapse.
Collapse
Affiliation(s)
- James D Ryan
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States.
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Farah K Bshesh
- Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, P.O. Box 24144 Doha, Qatar
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Joshua F Kogan
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Konrad T Ben
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States.
| |
Collapse
|
28
|
Abstract
There are several biological factors that might play a role in physiological response to opioids and/or the onset of problematic opioid use; however, sex-based differences in non-analgesic opioid-based effects are poorly understood. The goal of this review is to provide a current analysis of the pre-clinical literature on sex-based differences in response to endogenous and exogenous opioids, including the interplay between sex hormones and opioid receptor-mediated neuronal activity and associated behaviours. A systematic search was performed on the following terms within PubMed between March and April 2018: 'opioid oestrogen', 'opioid progesterone', 'opioid oestradiol', and 'opioid testosterone'. Pre-clinical research on the non-analgesic, sex-based effects of opioids is disparate, both in terms of methodology and outcomes, which prohibits a cohesive summary of the results. Themes from the pre-clinical literature suggest that opioid receptor binding, coupling, and density vary as a function of hormone exposure. Findings also suggest that interactions between endogenous opioid and stress systems may differ between males and females as a function of ovarian hormones. Given the current opioid-related public health crisis, there is a pressing need to increase systematic pre-clinical and clinical research on sex-based differences in opioid-effects and opioid use disorder.
Collapse
Affiliation(s)
- Andrew S. Huhn
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Meredith S. Berry
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kelly E. Dunn
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Legakis LP, Negus SS. Repeated Morphine Produces Sensitization to Reward and Tolerance to Antiallodynia in Male and Female Rats with Chemotherapy-Induced Neuropathy. J Pharmacol Exp Ther 2018; 365:9-19. [PMID: 29363579 DOI: 10.1124/jpet.117.246215] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/22/2018] [Indexed: 01/14/2023] Open
Abstract
Paclitaxel is a cancer chemotherapy drug with adverse effects that include chemotherapy-induced neuropathic pain (CINP) as well as depression of behavior and mood. In the clinical setting, opioids are often used concurrently with or after chemotherapy to treat pain related to the cancer or CINP, but repeated opioid exposure can also increase the risk of opioid abuse. In this study, male and female Sprague-Dawley rats were used to test the hypothesis that repeated 3.2-mg/kg doses of morphine would induce tolerance to its antinociceptive effects in a mechanical sensitivity assay and increased expression of its abuse-related rewarding effects in an assay of intracranial self-stimulation (ICSS). Three weeks after four injections of vehicle or 2.0 mg/kg of paclitaxel, the initial morphine dose-effect curves were determined in both assays. Subsequently, rats were treated with 3.2 mg/kg per day morphine for 6 days. On the final day of testing, morphine dose-effect curves were redetermined in both assays. On initial exposure, morphine produced dose-dependent antiallodynia in the assay of mechanical sensitivity, but it produced little or no rewarding effects in the assay of ICSS. After 6 days of repeated treatment, morphine antiallodynia decreased, and morphine reward increased. Females exhibited greater morphine reward on initial exposure than males, but repeated morphine eliminated this sex difference. These results suggest that repeated morphine may produce tolerance to therapeutically beneficial analgesic effects of morphine but increased sensitivity to abuse-related rewarding effects of morphine in subjects treated with paclitaxel.
Collapse
Affiliation(s)
- L P Legakis
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - S S Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
30
|
Antagonism of mGlu2/3 receptors in the nucleus accumbens prevents oxytocin from reducing cued methamphetamine seeking in male and female rats. Pharmacol Biochem Behav 2017; 161:13-21. [PMID: 28870523 DOI: 10.1016/j.pbb.2017.08.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 11/23/2022]
Abstract
Methamphetamine (meth) addiction is a prevalent health concern worldwide, yet remains without approved pharmacological treatments. Preclinical evidence suggests that oxytocin may decrease relapse, but the neuronal underpinnings driving this effect remain unknown. Here we investigate whether oxytocin's effect is dependent on presynaptic glutamatergic regulation in the nucleus accumbens core (NAcore) by blocking metabotropic glutamate receptors 2/3 (mGluR2/3). Male and female Sprague-Dawley rats self-administered meth or sucrose on an escalating fixed ratio, followed by extinction and cue-induced reinstatement sessions. Reinstatement tests consisted of systemic (Experiment 1) or site-specific application of the drugs into the NAcore (Experiments 2 and 3). Before reinstatement sessions, rats received LY341495, an mGluR2/3 antagonist, or its vehicle followed by a second infusion/injection of oxytocin or saline. As expected, both males and females reinstated lever pressing to meth associated cues, and LY341495 alone did not impact this behavior. Oxytocin injected systemically or infused into the NAcore decreased cued meth seeking. Importantly, combined LY341495 and oxytocin administration restored meth cued reinstatement. Interestingly, neither oxytocin nor LY341495 impacted sucrose-cued reinstatement, suggesting distinct mechanisms between meth and sucrose. These findings were consistent between males and females. Overall, we report that oxytocin reduced responding to meth-associated cues and blocking presynaptic mGluR2/3 reversed this effect. Further, oxytocin's effects were specific to meth cues as NAcore oxytocin was without an effect on sucrose cued reinstatement. Results are discussed in terms of oxytocin receptor localization in the NAcore and modulation of presynaptic regulation of glutamate in response to drug associated cues.
Collapse
|
31
|
Wang X, Fitts RH. Ventricular action potential adaptation to regular exercise: role of β-adrenergic and KATP channel function. J Appl Physiol (1985) 2017; 123:285-296. [DOI: 10.1152/japplphysiol.00197.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/21/2017] [Accepted: 05/15/2017] [Indexed: 01/06/2023] Open
Abstract
Regular exercise training is known to affect the action potential duration (APD) and improve heart function, but involvement of β-adrenergic receptor (β-AR) subtypes and/or the ATP-sensitive K+ (KATP) channel is unknown. To address this, female and male Sprague-Dawley rats were randomly assigned to voluntary wheel-running or control groups; they were anesthetized after 6–8 wk of training, and myocytes were isolated. Exercise training significantly increased APD of apex and base myocytes at 1 Hz and decreased APD at 10 Hz. Ca2+ transient durations reflected the changes in APD, while Ca2+ transient amplitudes were unaffected by wheel running. The nonselective β-AR agonist isoproterenol shortened the myocyte APD, an effect reduced by wheel running. The isoproterenol-induced shortening of APD was largely reversed by the selective β1-AR blocker atenolol, but not the β2-AR blocker ICI 118,551, providing evidence that wheel running reduced the sensitivity of the β1-AR. At 10 Hz, the KATP channel inhibitor glibenclamide prolonged the myocyte APD more in exercise-trained than control rats, implicating a role for this channel in the exercise-induced APD shortening at 10 Hz. A novel finding of this work was the dual importance of altered β1-AR responsiveness and KATP channel function in the training-induced regulation of APD. Of physiological importance to the beating heart, the reduced response to adrenergic agonists would enhance cardiac contractility at resting rates, where sympathetic drive is low, by prolonging APD and Ca2+ influx; during exercise, an increase in KATP channel activity would shorten APD and, thus, protect the heart against Ca2+ overload or inadequate filling. NEW & NOTEWORTHY Our data demonstrated that regular exercise prolonged the action potential and Ca2+ transient durations in myocytes isolated from apex and base regions at 1-Hz and shortened both at 10-Hz stimulation. Novel findings were that wheel running shifted the β-adrenergic receptor agonist dose-response curve rightward compared with controls by reducing β1-adrenergic receptor responsiveness and that, at the high activation rate, myocytes from trained animals showed higher KATP channel function.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Robert H. Fitts
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| |
Collapse
|
32
|
Mavrikaki M, Pravetoni M, Page S, Potter D, Chartoff E. Oxycodone self-administration in male and female rats. Psychopharmacology (Berl) 2017; 234:977-987. [PMID: 28127624 PMCID: PMC7250466 DOI: 10.1007/s00213-017-4536-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/10/2017] [Indexed: 02/04/2023]
Abstract
RATIONALE Oxycodone is one of the most widely prescribed painkillers in the USA. However, its use is complicated by high abuse potential. As sex differences have been described in drug addiction, the present study tested for sex differences in intravenous oxycodone self-administration in rats. METHODS Male and female Sprague-Dawley rats were implanted with jugular vein catheters and trained to self-administer oxycodone (0.03 mg/kg/infusion) under fixed ratio 1 (FR1), FR2, and FR5 schedules of reinforcement followed by a dose-response study to assess sensitivity to the reinforcing effects of oxycodone. In separate rats, sucrose pellet self-administration was assessed under an FR1 schedule to determine whether sex differences in oxycodone self-administration could be generalized across reinforcers. In separate rats, oxycodone distribution to plasma and brain was measured after intravenous drug delivery. RESULTS In the first 3 trials under an FR1 schedule of reinforcement, male rats self-administered more oxycodone than females. In contrast, females self-administered more sucrose pellets. Under FR2 and FR5 schedules, no significant sex differences in oxycodone intake were observed, although female rats had significantly more inactive lever presses. Male and female rats showed similar inverted U-shaped dose-effect functions, with females tending to self-administer more oxycodone than males at higher doses. No significant sex differences were observed in plasma or brain oxycodone levels, suggesting that sex differences in oxycodone self-administration behavior were not due to pharmacokinetics. CONCLUSION Our results suggest subtle sex differences in oxycodone self-administration, which may influence the abuse liability of oxycodone and have ramifications for prescription opioid addiction treatment.
Collapse
Affiliation(s)
- Maria Mavrikaki
- Department of Psychiatry, Harvard Medical School, McLean Hospital, 115 Mill Street, Belmont, MA, 02478, USA.
| | - Marco Pravetoni
- Department of Medicine and Pharmacology, Minneapolis Medical Research Foundation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sarah Page
- Department of Psychiatry, Harvard Medical School, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA
| | - David Potter
- Department of Psychiatry, Harvard Medical School, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA
| | - Elena Chartoff
- Department of Psychiatry, Harvard Medical School, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA
| |
Collapse
|
33
|
Kandasamy R, Calsbeek JJ, Morgan MM. Analysis of inflammation-induced depression of home cage wheel running in rats reveals the difference between opioid antinociception and restoration of function. Behav Brain Res 2016; 317:502-507. [PMID: 27746208 DOI: 10.1016/j.bbr.2016.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 02/06/2023]
Abstract
Opioids are effective at inhibiting responses to noxious stimuli in rodents, but have limited efficacy and many side effects in chronic pain patients. One reason for this disconnect is that nociception is typically assessed using withdrawal from noxious stimuli in animals, whereas chronic pain patients suffer from abnormal pain that disrupts normal activity. We hypothesized that assessment of home cage wheel running in rats would provide a much more clinically relevant method to assess opioid efficacy to restore normal behavior. Intraplantar injection of Complete Freund's Adjuvant (CFA) into the right hindpaw depressed wheel running and caused mechanical allodynia measured with the von Frey test in both male and female rats. Administration of an ED50 dose of morphine (3.2mg/kg) reversed mechanical allodynia, but did not reverse CFA-induced depression of wheel running. In contrast, administration of a low dose of morphine (1.0mg/kg) restored running for one hour in both sexes, but had no effect on mechanical allodynia. Administration of the atypical opioid buprenorphine had no effect on inflammation-induced depression of wheel running in male or female rats, but attenuated mechanical allodynia in male rats. Administration of buprenorphine and higher doses of morphine depressed wheel running in non-inflamed rats, suggesting that the side effects of opioids interfere with restoration of function. These data indicate that restoration of pain-depressed function requires antinociception in the absence of disruptive side effects. The disruptive side effects of opioids are consistent with the major limitation of opioid use in human pain patients.
Collapse
Affiliation(s)
- Ram Kandasamy
- Graduate Program in Neuroscience, Washington State University, Pullman, WA, United States.
| | - Jonas J Calsbeek
- Department of Psychology, Washington State University Vancouver, Vancouver, WA, United States
| | - Michael M Morgan
- Graduate Program in Neuroscience, Washington State University, Pullman, WA, United States; Department of Psychology, Washington State University Vancouver, Vancouver, WA, United States
| |
Collapse
|
34
|
Collins D, Reed B, Zhang Y, Kreek MJ. Sex differences in responsiveness to the prescription opioid oxycodone in mice. Pharmacol Biochem Behav 2016; 148:99-105. [PMID: 27316549 DOI: 10.1016/j.pbb.2016.06.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 06/12/2016] [Accepted: 06/13/2016] [Indexed: 11/29/2022]
Abstract
Over-prescription and increased nonmedical use of oxycodone has become a major concern. Despite its increased use, preclinical data concerning oxycodone's effects are still limited, especially in rodent models. To address this, we examined oxycodone's effects on place preference, locomotor activation, corticosterone levels, and thermal analgesia across a range of doses (between 0.3 and 10mg/kg) in gonadally intact, adult male and female C57BL/6J mice. Males and females showed oxycodone-induced conditioned place preference and did not show significant between-sex differences in their place preference behavior. During both CPP conditioning sessions and open field assay, locomotor activity was increased by 1, 3, and 10mg/kg oxycodone in females and by 3 and 10mg/kg oxycodone in males. Plasma corticosterone levels were higher in females (compared to males) at baseline as well as following acute oxycodone injection and open field testing. The time course of oxycodone-induced analgesia was similar in males and females, however the total antinociceptive effect (AUC0-120min) was larger in males compared to females at the highest dose tested (10mg/kg). Taken together, these data suggest that male and female mice are modestly different in their responses to oxycodone.
Collapse
Affiliation(s)
- Devon Collins
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States.
| | - Brian Reed
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Yong Zhang
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| |
Collapse
|
35
|
Macúchová E, Ševčíková M, Hrebíčková I, Nohejlová K, Šlamberová R. How various drugs affect anxiety‐related behavior in male and female rats prenatally exposed to methamphetamine. Int J Dev Neurosci 2016; 51:1-11. [DOI: 10.1016/j.ijdevneu.2016.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/04/2016] [Accepted: 04/07/2016] [Indexed: 01/23/2023] Open
Affiliation(s)
- E. Macúchová
- Charles University in PragueThird Faculty of Medicine, Department of Normal, Pathological and Clinical PhysiologyPragueCzech Republic
| | - M. Ševčíková
- Charles University in PragueThird Faculty of Medicine, Department of Normal, Pathological and Clinical PhysiologyPragueCzech Republic
| | - I. Hrebíčková
- Charles University in PragueThird Faculty of Medicine, Department of Normal, Pathological and Clinical PhysiologyPragueCzech Republic
| | - K. Nohejlová
- Charles University in PragueThird Faculty of Medicine, Department of Normal, Pathological and Clinical PhysiologyPragueCzech Republic
| | - R. Šlamberová
- Charles University in PragueThird Faculty of Medicine, Department of Normal, Pathological and Clinical PhysiologyPragueCzech Republic
| |
Collapse
|
36
|
Ponomarev K, Pavlova A, Suslov E, Ardashov O, Korchagina D, Nefedov A, Tolstikova T, Volcho K, Salakhutdinov N. Synthesis and analgesic activity of new compounds combining azaadamantane and monoterpene moieties. Med Chem Res 2015. [DOI: 10.1007/s00044-015-1464-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
37
|
Anoush M, Jani A, Sahebgharani M, Jafari MR. Effects of Estrogen Receptor Modulators on Morphine Induced Sensitization in Mice Memory. IRANIAN JOURNAL OF PSYCHIATRY 2015; 10:192-9. [PMID: 26877753 PMCID: PMC4749689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE In this study, the effects of estradiol valerate and raloxifenea selective estrogen receptor modulator; (SERM) on morphine induced sensitization were examined in mice memory, according to the step-down passive avoidance task. METHOD The mice received morphine or estradiol and raloxifene for three days alone or in combination with morphine. After a drug free period of 5 days, the subjects received saline or morphine as pre- training treatments followed by a pre-test saline administration. The memory retrieval was evaluated using step-down passive avoidance test both on the training and test day. RESULTS The results illustrated that the three- day administration of morphine induced sensitization through the enhancement of memory retrieval (morphine induced sensitization in mice memory). Both the three- day administration of estradiol valerate alone and with morphine (5 mg/kg) restored memory. On the other hand, the three- day administration of raloxifene had no effect on memory retrieval alone, but declined morphine induced sensitization in mice memory. CONCLUSION The results of the study indicated that there is an interaction between estrogen receptor modulators and morphine induced sensitization in mice memory.
Collapse
Affiliation(s)
- Mahdieh Anoush
- Department of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Jani
- Department of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Moosa Sahebgharani
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Jafari
- Department of Physiology and Pharmacology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
38
|
Zhou L, Ghee SM, See RE, Reichel CM. Oxytocin differentially affects sucrose taking and seeking in male and female rats. Behav Brain Res 2015; 283:184-90. [PMID: 25647756 PMCID: PMC4387851 DOI: 10.1016/j.bbr.2015.01.050] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/17/2014] [Accepted: 01/23/2015] [Indexed: 02/08/2023]
Abstract
Oxytocin has a modulatory role in natural and drug reward processes. While the role of oxytocin in pair bonding and reproduction has been extensively studied, sex differences in conditioned and unconditioned behavioral responses to oxytocin treatment have not been fully characterized. Here, we determined whether male and female rats would show similar dose response curves in response to acute oxytocin on measures of locomotor activity, sucrose seeking, and sucrose intake. Male and freely cycling female rats received vehicle or oxytocin (0.1, 0.3, 1, 3 mg/kg, IP) injections before behavioral tests designed to assess general motor activity, as well as sucrose self-administration and seeking. Lower doses of oxytocin decreased motor activity in a novel environment in females relative to males. Likewise, lower doses of oxytocin in females decreased responding for sucrose during maintenance of sucrose self-administration and reinstatement to sucrose-conditioned cues. However, sucrose seeking in response to a sucrose prime was only decreased by the highest oxytocin dose in both sexes. In general, oxytocin had similar effects in both sexes. However, females were more sensitive to lower doses of oxytocin than males. These findings are consistent with the notion that oxytocin regulates many of the same behaviors in males and females, but that the effects are typically more profound in females. Therapeutic use of oxytocin should include sex as a factor in determining dose regimens.
Collapse
Affiliation(s)
- Luyi Zhou
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Shannon M Ghee
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ronald E See
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Carmela M Reichel
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
39
|
Growth restriction alters adult spatial memory and sensorimotor gating in a sex-specific manner. J Dev Orig Health Dis 2014; 3:59-68. [PMID: 25101812 DOI: 10.1017/s2040174411000729] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In Western society, impaired uteroplacental blood flow is the major cause of human intrauterine growth restriction. Infants born small and who experience late childhood accelerated growth have an increased risk of developing adult diseases. Recent studies also suggest a link between birth weight and altered adult behavior, particularly relating to motor function, learning and memory, depression and schizophrenia. The aim of this study was to determine the relative influence of prenatal and postnatal growth restriction on adult behavioral outcomes in male and female rats. Uteroplacental insufficiency was induced in Wistar Kyoto rats by bilateral uterine vessel ligation on day 18 of gestation producing growth-restricted offspring (Restricted group). The Control group had sham surgery. Another group underwent sham surgery, with a reduction in litter size to five at birth equivalent to the Restricted litter size (Reduced Litter group). At 6 months of age, a series of behavioral tests were conducted in male and female offspring. Growth restriction did not impair motor function. In fact, Restricted and Reduced Litter males showed enhanced motor performance compared with Controls (P < 0.05). Spatial memory was greater in Restricted females only (P < 0.05). The Porsolts test was unremarkable, however, males exhibited more depressive-like behavior than females (P < 0.05). A reduction in sensorimotor gating function was identified in Reduced Litter males and females (P < 0.05). We have demonstrated that growth restriction and/or a poor lactational environment can affect adult rat behavior, particularly balance and coordination, memory and learning, and sensorimotor gating function, in a sex-specific manner.
Collapse
|
40
|
McNabb CT, White MM, Harris AL, Fuchs PN. The elusive rat model of conditioned placebo analgesia. Pain 2014; 155:2022-32. [PMID: 25026214 DOI: 10.1016/j.pain.2014.07.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 06/30/2014] [Accepted: 07/03/2014] [Indexed: 10/25/2022]
Abstract
Recent research on human placebo analgesia has suggested the need for rodent models to further elucidate the neural substrates of the placebo effect. This series of 3 experiments therefore was performed in an attempt to develop a model of placebo analgesia in rats. In each study, female Sprague-Dawley rats received an L5 spinal nerve ligation to induce a neuropathic pain condition. Each rat then underwent a 4-day conditioning procedure in which an active analgesic drug or its vehicle (unconditioned stimulus) was associated with the following cues (conditioned stimuli): novel testing room (environmental), vanilla scent cue (olfactory), dim incandescent lighting (visual), restraint procedure/injection (tactile), and time of day and injection-test latency (temporal). The analgesics for each experiment were as follows: Experiment 1 used 90 mg/kg gabapentin, experiment 2 used 3mg/kg loperamide hydrochloride, and experiment 3 used 6 mg/kg morphine sulfate. On the following test day, half of the animals received the opposite treatment, resulting in 4 conditioning manipulations: drug/drug, drug/vehicle, vehicle/drug, and vehicle/vehicle. Nociceptive thresholds were assessed with the mechanical paw withdrawal threshold test each day after the conditioning procedure. In all 3 experiments, no significant differences were detected on test day between control and placebo groups, indicating a lack of a conditioned placebo analgesic response. Our results contrast with prior research that implies the existence of a reliable and robust response to placebo treatment. We conclude that placebo analgesia in rats is not particularly robust and that it is difficult to achieve using conventional procedures and proper experimental design.
Collapse
Affiliation(s)
| | - Michelle M White
- Department of Psychology, University of Texas at Arlington, Arlington, TX, USA
| | - Amber L Harris
- Department of Psychology, University of Texas at Arlington, Arlington, TX, USA
| | - Perry N Fuchs
- Department of Psychology, University of Texas at Arlington, Arlington, TX, USA; Department of Biology, University of Texas at Arlington, Arlington, TX, USA.
| |
Collapse
|
41
|
|
42
|
Nolan TA, Price DD, Caudle RM, Murphy NP, Neubert JK. Placebo-induced analgesia in an operant pain model in rats. Pain 2012; 153:2009-2016. [PMID: 22871471 DOI: 10.1016/j.pain.2012.04.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/28/2012] [Accepted: 04/24/2012] [Indexed: 12/19/2022]
Abstract
Analgesia is particularly susceptible to placebo responses. Recent studies in humans have provided important insights into the neurobiology underlying placebo-induced analgesia. However, human studies provide incomplete mechanistic explanations of placebo analgesia because of limited capacity to use cellular, molecular, and genetic manipulations. To address this shortcoming, this article describes the development of a rat model of conditioned analgesia in an operant pain assay. Specifically, rats were conditioned to associate a placebo manipulation with the analgesic effect of 1mg/kg morphine (subcutaneously) on facial thermal pain. We found that conditioned (placebo) responding bore 3 of the hallmarks of placebo-induced analgesia: (1) strong interanimal variability in the response, (2) suppression by the opiate antagonist naloxone (5mg/kg subcutaneously), and (3) a positive predictive relationship between the unconditioned analgesic effect and the conditioned (placebo) effect. Because of the operant nature of the assay and the use of only a mild noxious thermal stimulus, we suggest that these results provide evidence of placebo-induced analgesia in a preclinical model that utilizes an affective behavioral end point. This finding may provide opportunities for invasive preclinical studies allowing greater understanding of placebo-induced analgesia, thus paving the way for avenues to harness its benefits.
Collapse
Affiliation(s)
- Todd A Nolan
- College of Dentistry, Department of Orthodontics, University of Florida, Gainesville, FL, USA Department of Oral Surgery, University of Florida, Gainesville, FL, USA Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|
43
|
Amnesia induced by morphine in spatial memory retrieval inhibited in morphine-sensitized rats. Eur J Pharmacol 2012; 683:132-9. [DOI: 10.1016/j.ejphar.2012.02.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 01/20/2012] [Accepted: 02/26/2012] [Indexed: 12/17/2022]
|
44
|
Halladay LR, Blair HT. The role of mu-opioid receptor signaling in the dorsolateral periaqueductal gray on conditional and unconditional responding to threatening and aversive stimuli. Neuroscience 2012; 216:82-93. [PMID: 22542677 DOI: 10.1016/j.neuroscience.2012.04.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 04/03/2012] [Accepted: 04/14/2012] [Indexed: 11/26/2022]
Abstract
Here we examined how mu-opioid receptor signaling in the periaqueductal gray (PAG) mediates conditional and unconditional responses to aversive stimuli. The mu-opioid agonist morphine (MOR) and/or the partially mu-selective antagonist naltrexone (NAL) were infused into dorsolateral PAG (dlPAG) during a fear conditioning task, in which rats were trained to fear an auditory conditional stimulus (CS) by pairing it with a unilateral eyelid shock unconditional stimulus (US). During drug-free test sessions, the CS elicited movement suppression responses (indicative of freezing) from trained rats that had not recently encountered the US. In trained rats that had recently encountered the US, the CS elicited flight behavior characterized by turning in the direction away from the eyelid where US delivery was anticipated. Infusions of MOR (30 nmol/side) into dlPAG prior to the test session did not impair CS-evoked movement suppression, but did impair CS-evoked turning behaviors. MOR infusions also reduced baseline motor movement, but US-evoked reflex movements remained largely intact. NAL was infused at two dosages, denoted 1x (26 nmol/side) and 10x (260 nmol/side). Infusions of NAL into dlPAG did not affect CS- or US-evoked behavioral responses at the 1x dosage, but impaired CS-evoked movement suppression at the 10x dosage, both in the presence and absence of MOR. When rats were co-infused with MOR and NAL, MOR-induced effects were not reversed by either dosage of NAL, and some measures of MOR-induced movement suppression were enhanced by NAL at the 1x dosage. Based on these findings, we conclude that mu-opioid receptors in dlPAG may selectively regulate descending supraspinal motor pathways that drive active movement behaviors, and that interactions between MOR and NAL in dlPAG may be more complex than simple competition for binding at the mu receptor.
Collapse
Affiliation(s)
- L R Halladay
- University of California, Los Angeles, Department of Psychology, 1285 Franz Hall, C533, Los Angeles, CA 90095-1563, USA.
| | | |
Collapse
|
45
|
Some determinants of morphine effects on intracranial self-stimulation in rats: dose, pretreatment time, repeated treatment, and rate dependence. Behav Pharmacol 2012; 22:663-73. [PMID: 21921839 DOI: 10.1097/fbp.0b013e32834aff54] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Intracranial self-stimulation (ICSS) is a procedure used to evaluate the abuse liability of drugs. The μ opioid receptor agonist morphine is an acknowledged drug of abuse, and this study examined factors that may influence expression of abuse-related morphine effects on ICSS in rats. Adult male rats were equipped with intracranial electrodes targeting the medial forebrain bundle, and 10 stimulus frequencies (56-158 Hz in 0.05 log increments) were available during each daily session under a continuous reinforcement schedule. The primary dependent variable was the ICSS rate at each frequency. Under baseline conditions, the ICSS rate increased with frequency. After acute morphine (1-10 mg/kg), rate-decreasing effects predominated at early pretreatment times (10-30 min) and rate-increasing effects predominated at later pretreatment times (100-180 min). Acute morphine effects dissipated after 300 min. Repeated morphine (3.2-18 mg/kg/day×7 days at each dose) produced tolerance to rate-decreasing effects, enhanced expression of rate-increasing effects, and enhanced rate dependency of morphine effects. Withdrawal from repeated morphine produced small but significant dose-dependent decreases in ICSS. These results show that the magnitude and valence of morphine effects on rates of ICSS in rats are strongly influenced by morphine dose and pretreatment time, history of morphine exposure, and baseline ICSS rate.
Collapse
|
46
|
Hofford RS, Wellman PJ, Eitan S. Social influences on plasma testosterone levels in morphine withdrawn adolescent mice and their drug-naïve cage-mates. Psychoneuroendocrinology 2011; 36:728-36. [PMID: 21071150 DOI: 10.1016/j.psyneuen.2010.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2010] [Revised: 08/17/2010] [Accepted: 10/12/2010] [Indexed: 10/18/2022]
Abstract
Opioid administration in males results in opioid-induced androgen deficiency which persists throughout the treatment. In adults, this quickly reverses once opioid administration is suspended. However, less is known about the duration of the effect following drug discontinuation in adolescents. Given the significant implications to sexual maturation in adolescent males, this study examined plasma testosterone levels in both morphine withdrawn mice and their drug-naïve (saline-injected) cage-mates as compared to drug-naïve mice housed physically and visually separate from the morphine-treated mice ('saline only'). Consistent with the literature, plasma testosterone levels in morphine withdrawn adults were reduced on withdrawal day 1 (WD1) and returned to baseline levels by WD9. No significant effects were observed in their saline cage-mates. In the adolescents, no significant differences were observed on WD1 between the morphine withdrawn mice, their saline cage-mates, and the saline only mice - all of which had significantly lower plasma testosterone levels than adults. By WD9, testosterone levels in the saline only adolescent mice had reached adult levels. Notably, plasma testosterone levels were reduced in both the morphine withdrawn adolescent mice and their saline cage-mates, as compared to saline only mice. The effect was not a drug effect per se, given that reduced plasma testosterone levels were not observed in individually housed morphine withdrawn mice. Moreover, our results also suggest that these social effects are not solely explained by stress. These results have numerous implications to the short term and long term health of both adolescents requiring pain management and of adolescent drug addicts.
Collapse
Affiliation(s)
- Rebecca S Hofford
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | | | | |
Collapse
|
47
|
Karami M, Zarrindast MR. Place aversion by morphine in offspring born of female morphine administered wistar rats. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2011; 10:577-84. [PMID: 24250391 PMCID: PMC3813028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This research was designed to study sexual differences in place conditioning induced by morphine in offspring born of female Wistar rats mated with drug-naïve males. Mothers were exposed to morphine during the 14(th)-16(th) days of gestational. Control dams were simply saline-injected. Female and male virgin offspring born of morphine-treated or saline-treated mothers were separately housed until become fully matured. A 3-day schedule of an unbiased conditioning procedure was used to the induce conditioning to morphine (2.5-7.5 mg/Kg, SC) in the offspring. According to the results, female offspring born of saline-administered mothers were morphine place-conditioned at lower doses of opioid (2.5 mg/Kg) in comparison to the males. An increase in locomotor activity in the females at 7.5 mg/Kg of opioid was also revealed. In contrast, administration of morphine (2.5-7.5 mg/Kg, SC), induced a significant aversion in either sexes of offspring born of morphine-exposed mothers. Moreover, female offspring of this category acquired more pronounced aversion at higher doses of morphine than males. In addition, a significant morphine-dose effect (7.5 mg/Kg, SC) on locomotor activity of these females' offspring was observed. This study may highlight sex differences in conditioning effects induced by morphine between offspring derived of morphine-treated mothers and those of saline-treated.
Collapse
Affiliation(s)
- Manizheh Karami
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran.,Basic Sciences Research Center, Shahed University, Tehran, Iran.,Corresponding author: E-mail:
| | - Mohammad Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Peri, pre and postnatal morphine exposure: exposure-induced effects and sex differences in the behavioural consequences in rat offspring. Behav Pharmacol 2010; 21:58-68. [PMID: 20038835 DOI: 10.1097/fbp.0b013e3283359f39] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This study investigated the behavioural consequences of peri, pre and postnatal morphine (MO) exposure in rats. From gestational day 1 dams were treated with either saline or MO subcutaneously once a day (5 mg/kg on the first 2 days, 10 mg/kg subsequently). Spontaneous locomotor activity in a new environment (habituation) and antinociceptive effects of MO were measured separately in male and female pups after weaning and also in late adolescence or adulthood. The rewarding effect of MO was assessed by conditioned place preference in adult animals. Both exposure-induced and sex differences were observed. A significant delay in habituation to a new environment and decreased sensitivity to the antinociceptive effect of MO were found in male offspring of MO-treated dams. In contrast, the place preference induced by MO was enhanced in the MO-exposed adult animals and this effect was more marked in females. Prenatal exposure to MO resulted in more marked changes than the postnatal exposure through maternal milk. The results indicate that a medium MO dose administered once-daily results in long-term consequences in offspring and may make them more vulnerable to MO abuse in adulthood.
Collapse
|
49
|
Circadian phase and sex effects on depressive/anxiety-like behaviors and HPA axis responses to acute stress. Physiol Behav 2009; 99:276-85. [PMID: 19932127 DOI: 10.1016/j.physbeh.2009.11.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Revised: 11/03/2009] [Accepted: 11/09/2009] [Indexed: 01/21/2023]
Abstract
Circadian dysregulation in sleep pattern, mood, and hypothalamic-pituitary-adrenal (HPA) axis activity, often occurring in a sexually dimorphic manner, are characteristics of depression. However, the inter-relationships among circadian phase, HPA function, and depressive-like behaviors are not well understood. We investigated behavioral and neuroendocrine correlates of depressive/anxiety-like responses during diurnal ('light') and nocturnal ('dark') phases of the circadian rhythm in the open field (OF), elevated plus maze (EPM), forced swim (FST), and sucrose contrast (SC) tests. Plasma corticosterone (CORT) was measured after a) acute restraint and OF testing and b) FST. Both phase and sex significantly influenced behavioral responses to stress. Males were more anxious than females on the EPM in the light but not the dark phase. Further, the open:closed arm ratio was lower in the dark for females, but not males. By contrast, in the FST, females showed more "despair" (immobility) when tested in the dark, while phase did not affect males. Acute restraint stress increased OF activity in the light, but not the dark, phase. CORT levels were increased in both sexes following the FST, and in males and light phase females post-OF. As expected, females had higher CORT levels than males, even at rest, and this effect was more pronounced in the dark phase. Together, our data highlight the sexually dimorphic influences of circadian phase and stress on behavioral and hormonal responsiveness.
Collapse
|
50
|
Halladay LR, Iñiguez SD, Furqan F, Previte MC, Chisum AM, Crawford CA. Methylphenidate potentiates morphine-induced antinociception, hyperthermia, and locomotor activity in young adult rats. Pharmacol Biochem Behav 2008; 92:190-6. [PMID: 19100281 DOI: 10.1016/j.pbb.2008.11.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 11/09/2008] [Accepted: 11/24/2008] [Indexed: 02/09/2023]
Abstract
The goal of this study was to determine if the exaggerated morphine-induced conditioned place preference (CPP) response seen in adult rats after preweanling methylphenidate exposure is unique to reward-mediated behaviors or is indicative of generalized changes in opioid-mediated behaviors. Rats were exposed to saline or methylphenidate (2.0 or 5.0 mg/kg) for 10 consecutive days starting on postnatal (PD) 11 with testing beginning on PD 60. In Experiment 1, morphine-induced (0, 2.5, 5.0 or 10.0 mg/kg) antinociception was assessed using the tail immersion and hot plate tasks. In Experiment 2, morphine-induced (0, 2.5, 5.0, or 10.0 mg/kg) hyperthermia and locomotor activity were measured. Morphine caused an increase in antinociception, with early methylphenidate (5.0 mg/kg) exposure potentiating the effects of 5.0 mg/kg morphine. Rectal temperatures were elevated after morphine, with the greatest increase occurring in male rats. Methylphenidate potentiated the hyperthermic effects of morphine (10.0 mg/kg) but only in males. Moderate doses (2.5 and 5.0 mg/kg) of morphine increased the locomotor activity of adult rats, while a higher dose (10.0 mg/kg) decreased locomotion. Interestingly, methylphenidate-pretreated females showed increased locomotor activity relative to controls. These results suggest that early methylphenidate exposure induces general changes in opioid system functioning that are not specific to reward-mediated behaviors.
Collapse
Affiliation(s)
- Lindsay R Halladay
- Department of Psychology, California State University, San Bernardino, CA 92407, USA
| | | | | | | | | | | |
Collapse
|