1
|
de Guglielmo G, Carrette L, Kallupi M, Brennan M, Boomhower B, Maturin L, Conlisk D, Sedighim S, Tieu L, Fannon MJ, Martinez AR, Velarde N, Othman D, Sichel B, Ramborger J, Lau J, Kononoff J, Kimbrough A, Simpson S, Smith LC, Shankar K, Bonnet-Zahedi S, Sneddon EA, Avelar A, Plasil SL, Mosquera J, Crook C, Chun L, Vang A, Milan KK, Schweitzer P, Lin B, Peng B, Chitre AS, Polesskaya O, Solberg Woods LC, Palmer AA, George O. Large-scale characterization of cocaine addiction-like behaviors reveals that escalation of intake, aversion-resistant responding, and breaking-points are highly correlated measures of the same construct. eLife 2024; 12:RP90422. [PMID: 39484794 DOI: 10.7554/elife.90422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
Addiction is commonly characterized by escalation of drug intake, compulsive drug seeking, and continued use despite harmful consequences. However, the factors contributing to the transition from moderate drug use to these problematic patterns remain unclear, particularly regarding the role of sex. Many preclinical studies have been limited by small sample sizes, low genetic diversity, and restricted drug access, making it challenging to model significant levels of intoxication or dependence and translate findings to humans. To address these limitations, we characterized addiction-like behaviors in a large sample of >500 outbred heterogeneous stock (HS) rats using an extended cocaine self-administration paradigm (6 hr/daily). We analyzed individual differences in escalation of intake, progressive ratio (PR) responding, continued use despite adverse consequences (contingent foot shocks), and irritability-like behavior during withdrawal. Principal component analysis showed that escalation of intake, progressive ratio responding, and continued use despite adverse consequences loaded onto a single factor that was distinct from irritability-like behaviors. Categorizing rats into resilient, mild, moderate, and severe addiction-like phenotypes showed that females exhibited higher addiction-like behaviors, with a lower proportion of resilient individuals compared to males. These findings suggest that, in genetically diverse rats with extended drug access, escalation of intake, continued use despite adverse consequences, and PR responding are highly correlated measures of a shared underlying construct. Furthermore, our results highlight sex differences in resilience to addiction-like behaviors.
Collapse
Affiliation(s)
- Giordano de Guglielmo
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Lieselot Carrette
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Marsida Kallupi
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Molly Brennan
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Brent Boomhower
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Lisa Maturin
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Dana Conlisk
- Department of Neuroscience, The Scripps Research Institute, La Jolla, San Diego, United States
| | - Sharona Sedighim
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Lani Tieu
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - McKenzie J Fannon
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Angelica R Martinez
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Nathan Velarde
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Dyar Othman
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Benjamin Sichel
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Jarryd Ramborger
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Justin Lau
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Jenni Kononoff
- Department of Neuroscience, The Scripps Research Institute, La Jolla, San Diego, United States
| | - Adam Kimbrough
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Sierra Simpson
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Lauren C Smith
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
- Department of Neuroscience, The Scripps Research Institute, La Jolla, San Diego, United States
| | - Kokila Shankar
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
- Department of Neuroscience, The Scripps Research Institute, La Jolla, San Diego, United States
| | - Selene Bonnet-Zahedi
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
- Institut de Neurosciences de la Timone, Aix-Marseille Université, Marseille, France
| | - Elizabeth A Sneddon
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Alicia Avelar
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
- Department of Neuroscience, The Scripps Research Institute, La Jolla, San Diego, United States
| | - Sonja Lorean Plasil
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Joseph Mosquera
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Caitlin Crook
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Lucas Chun
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Ashley Vang
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Kristel K Milan
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Paul Schweitzer
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Bonnie Lin
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Beverly Peng
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Apurva S Chitre
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| | - Oksana Polesskaya
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
- Institute for Genomic Medicine, University of California, San Diego, La Jolla, United States
| | - Leah C Solberg Woods
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, United States
| | - Abraham A Palmer
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
- Institute for Genomic Medicine, University of California, San Diego, La Jolla, United States
| | - Olivier George
- Department of Psychiatry, University of California, San Diego, La Jolla, United States
| |
Collapse
|
2
|
Sardari M, Mohammadpourmir F, Hosseinzadeh Sahafi O, Rezayof A. Neuronal biomarkers as potential therapeutic targets for drug addiction related to sex differences in the brain: Opportunities for personalized treatment approaches. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111068. [PMID: 38944334 DOI: 10.1016/j.pnpbp.2024.111068] [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: 11/25/2023] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024]
Abstract
Biological sex disparities manifest at various stages of drug addiction, including craving, substance abuse, abstinence, and relapse. These discrepancies are underpinned by notable distinctions in neurobiological substrates, encompassing brain structures, functions, and neurotransmitter systems implicated in drug addiction. Neuronal biomarkers, such as neurotransmitters, signaling proteins, and genes may be associated with the diagnosis, prognosis, and treatment outcomes in both biological sexes afflicted by drug abuse. Sex differences in the neural reward system, mainly through dopaminergic transmission during drug abuse, can be attributed to modifications in neurotransmitter systems and signaling pathways. This results in distinct patterns of neural activation and responsiveness to addictive substances in males and females. Sex hormones, the estrus/menstrual cycle, and cerebral neurochemistry contribute to the progression of psychological and physiological dependence in both male and female individuals grappling with addiction. Moreover, the alteration of sex hormone balance and neurotransmitter release plays a pivotal role in substance use disorders, subsequently modulating cognitive functions pertinent to reward, including memory formation, decision-making, and locomotor activity. Comparative investigations reveal distinctions in brain region volume, gene expression, neuronal firing, and circuitry in substance use disorders affecting individuals of both biological sexes. This review examines prevalent substance use disorders to elucidate the impact of sex hormones as therapeutic biomarkers on the mesocorticolimbic neurotransmitter systems via diverse mechanisms within the addicted brain. We underscore the imperative necessity of considering these variations to gain a deeper comprehension of addiction mechanisms and potentially discern sex-specific neuronal biomarkers for tailored therapeutic interventions.
Collapse
Affiliation(s)
- Maryam Sardari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Farina Mohammadpourmir
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Oveis Hosseinzadeh Sahafi
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
3
|
O'Connor AM, Hagenauer MH, Thew Forrester LC, Maras PM, Arakawa K, Hebda-Bauer EK, Khalil H, Richardson ER, Rob FI, Sannah Y, Watson SJ, Akil H. Adolescent environmental enrichment induces social resilience and alters neural gene expression in a selectively bred rodent model with anxious phenotype. Neurobiol Stress 2024; 31:100651. [PMID: 38933284 PMCID: PMC11201356 DOI: 10.1016/j.ynstr.2024.100651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 04/10/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Stress is a major influence on mental health status; the ways that individuals respond to or copes with stressors determine whether they are negatively affected in the future. Stress responses are established by an interplay between genetics, environment, and life experiences. Psychosocial stress is particularly impactful during adolescence, a critical period for the development of mood disorders. In this study we compared two established, selectively-bred Sprague Dawley rat lines, the "internalizing" bred Low Responder (bLR) line versus the "externalizing" bred High Responder (bHR) line, to investigate how genetic temperament and adolescent environment impact future responses to social interactions and psychosocial stress, and how these determinants of stress response interact. Male bLR and bHR rats were exposed to social and environmental enrichment in adolescence prior to experiencing social defeat and were then assessed for social interaction and anxiety-like behavior. Adolescent enrichment caused rats to display more social interaction, as well as nominally less social avoidance, less submission during defeat, and resilience to the effects of social stress on corticosterone, in a manner that seemed more notable in bLRs. For bHRs, enrichment also caused greater aggression during a neutral social encounter and nominally during defeat, and decreased anxiety-like behavior. To explore the neurobiology underlying the development of social resilience in the anxious phenotype bLRs, RNA-seq was conducted on the hippocampus and nucleus accumbens, two brain regions that mediate stress regulation and social behavior. Gene sets previously associated with stress, social behavior, aggression and exploratory activity were enriched with differential expression in both regions, with a particularly large effect on gene sets that regulate social behaviors. Our findings provide further evidence that adolescent enrichment can serve as an inoculating experience against future stressors. The ability to induce social resilience in a usually anxious line of animals by manipulating their environment has translational implications, as it underscores the feasibility of intervention strategies targeted at genetically vulnerable adolescent populations.
Collapse
Affiliation(s)
| | - Megan Hastings Hagenauer
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Liam Cannon Thew Forrester
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Pamela M. Maras
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Keiko Arakawa
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Elaine K. Hebda-Bauer
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Huzefa Khalil
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Evelyn R. Richardson
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Farizah I. Rob
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Yusra Sannah
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Stanley J. Watson
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Huda Akil
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| |
Collapse
|
4
|
O'Connor AM, Hagenauer MH, Forrester LCT, Maras PM, Arakawa K, Hebda-Bauer EK, Khalil H, Richardson ER, Rob FI, Sannah Y, Watson SJ, Akil H. Adolescent environmental enrichment induces social resilience and alters neural gene expression in a selectively bred rodent model with anxious phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.03.560702. [PMID: 38645129 PMCID: PMC11030238 DOI: 10.1101/2023.10.03.560702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Stress is a major influence on mental health status; the ways that individuals respond to or copes with stressors determine whether they are negatively affected in the future. Stress responses are established by an interplay between genetics, environment, and life experiences. Psychosocial stress is particularly impactful during adolescence, a critical period for the development of mood disorders. In this study we compared two established, selectively-bred Sprague Dawley rat lines, the "internalizing" bred Low Responder (bLR) line versus the "externalizing" bred High Responder (bHR) line, to investigate how genetic temperament and adolescent environment impact future responses to social interactions and psychosocial stress, and how these determinants of stress response interact. Male bLR and bHR rats were exposed to social and environmental enrichment in adolescence prior to experiencing social defeat and were then assessed for social interaction and anxiety-like behavior. Adolescent enrichment caused rats to display more social interaction, as well as nominally less social avoidance, less submission during defeat, and resilience to the effects of social stress on corticosterone, in a manner that seemed more notable in bLRs. For bHRs, enrichment also caused greater aggression during a neutral social encounter and nominally during defeat, and decreased anxiety-like behavior. To explore the neurobiology underlying the development of social resilience in the anxious phenotype bLRs, RNA-seq was conducted on the hippocampus and nucleus accumbens, two brain regions that mediate stress regulation and social behavior. Gene sets previously associated with stress, social behavior, aggression and exploratory activity were enriched with differential expression in both regions, with a particularly large effect on gene sets that regulate social behaviors. Our findings provide further evidence that adolescent enrichment can serve as an inoculating experience against future stressors. The ability to induce social resilience in a usually anxious line of animals by manipulating their environment has translational implications, as it underscores the feasibility of intervention strategies targeted at genetically vulnerable adolescent populations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Huda Akil
- Univ. of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
5
|
Zakiniaeiz Y, Gueorguieva R, Peltier MR, Verplaetse TL, Roberts W, McKee SA, Cosgrove KP. Sex steroid hormone levels associated with dopamine D 2/3 receptor availability in people who smoke cigarettes. Front Behav Neurosci 2023; 17:1192740. [PMID: 37358969 PMCID: PMC10288103 DOI: 10.3389/fnbeh.2023.1192740] [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: 03/23/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction Sex differences exist in tobacco smoking. Women have greater difficulty quitting smoking than men. Tobacco smoking is driven by the reinforcing effects of nicotine, the primary addictive component in cigarettes. Nicotine binds to nicotinic acetylcholine receptors, facilitating dopamine release in striatal and cortical brain regions. Dysregulated dopamine D2/3 receptor signaling in the dorsolateral prefrontal cortex (dlPFC) is associated with cognitive deficits such as impairments in attention, learning, and inhibitory control that impede quit attempts. Sex steroid hormones, such as estradiol and progesterone, influence drug-taking behaviors, through dopaminergic actions, suggesting that their influence may explain sex differences in tobacco smoking. The goal of this study was to relate dlPFC dopamine metrics to sex steroid hormone levels in people who smoke and healthy controls. Methods Twenty-four (12 women) people who smoke cigarettes and 25 sex- and age-matched controls participated in two same-day [11C]FLB457 positron emission tomography scans, one before and one after amphetamine administration. D2R availability (BPND) at baseline and after amphetamine administration was calculated. On the same day, plasma samples were collected for the analysis of sex steroid hormone levels: estradiol, progesterone, and free testosterone. Results Women who smoke had trending lower levels of estradiol than their sex-matched counterparts. Men who smoke had higher levels of estradiol and trending higher levels of free testosterone than their sex-matched counterparts. Among women only, lower estradiol levels were significantly associated with lower pre-amphetamine dlPFC BPND. Discussion/conclusion This study demonstrated that lower estradiol levels are associated with lower dlPFC D2R availability in women which may underlie difficulty resisting smoking.
Collapse
Affiliation(s)
- Yasmin Zakiniaeiz
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
| | - Ralitza Gueorguieva
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT, United States
| | - MacKenzie R. Peltier
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
- Psychology Service, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Terril L. Verplaetse
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
| | - Walter Roberts
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
| | - Sherry A. McKee
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
| | - Kelly P. Cosgrove
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
- Yale Positron Emission Tomography (PET) Center, School of Medicine, Yale University, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
6
|
Johnson CS, Mermelstein PG. The interaction of membrane estradiol receptors and metabotropic glutamate receptors in adaptive and maladaptive estradiol-mediated motivated behaviors in females. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:33-91. [PMID: 36868633 DOI: 10.1016/bs.irn.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Estrogen receptors were initially identified as intracellular, ligand-regulated transcription factors that result in genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor α and estrogen receptor β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) can rapidly alter cellular excitability and gene expression, particularly through the phosphorylation of CREB. A principal mechanism of neuronal mER action has been shown to occur through glutamate-independent transactivation of metabotropic glutamate receptors (mGlu), which elicits multiple signaling outcomes. The interaction of mERs with mGlu has been shown to be important in many diverse functions in females, including driving motivated behaviors. Experimental evidence suggests that a large part of estradiol-induced neuroplasticity and motivated behaviors, both adaptive and maladaptive, occurs through estradiol-dependent mER activation of mGlu. Herein we will review signaling through estrogen receptors, both "classical" nuclear receptors and membrane-bound receptors, as well as estradiol signaling through mGlu. We will focus on how the interactions of these receptors and their downstream signaling cascades are involved in driving motivated behaviors in females, discussing a representative adaptive motivated behavior (reproduction) and maladaptive motivated behavior (addiction).
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
7
|
de Guglielmo G, Iemolo A, Nur A, Turner A, Montilla-Perez P, Martinez A, Crook C, Roberts A, Telese F. Reelin deficiency exacerbates cocaine-induced hyperlocomotion by enhancing neuronal activity in the dorsomedial striatum. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12828. [PMID: 35906757 PMCID: PMC9744517 DOI: 10.1111/gbb.12828] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022]
Abstract
The Reln gene encodes for the extracellular glycoprotein Reelin, which regulates several brain functions from development to adulthood, including neuronal migration, dendritic growth and branching and synapse formation and plasticity. Human studies have implicated Reelin signaling in several neurodevelopmental and psychiatric disorders. Mouse studies using the heterozygous Reeler (HR) mice have shown that reduced levels of Reln expression are associated with deficits in learning and memory and increased disinhibition. Although these traits are relevant to substance use disorders, the role of Reelin in cellular and behavioral responses to addictive drugs remains largely unknown. Here, we compared HR mice to wild-type (WT) littermate controls to investigate whether Reelin signaling contributes to the hyperlocomotor and rewarding effects of cocaine. After a single or repeated cocaine injections, HR mice showed enhanced cocaine-induced locomotor activity compared with WT controls. This effect persisted after withdrawal. In contrast, Reelin deficiency did not induce cocaine sensitization, and did not affect the rewarding effects of cocaine measured in the conditioned place preference assay. The elevated cocaine-induced hyperlocomotion in HR mice was associated with increased protein Fos expression in the dorsal medial striatum (DMS) compared with WT. Lastly, we performed an RNA fluorescent in situ hybridization experiment and found that Reln was highly co-expressed with the Drd1 gene, which encodes for the dopamine receptor D1, in the DMS. These findings show that Reelin signaling contributes to the locomotor effects of cocaine and improve our understanding of the neurobiological mechanisms underlying the cellular and behavioral effects of cocaine.
Collapse
Affiliation(s)
- Giordano de Guglielmo
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Attilio Iemolo
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Aisha Nur
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Andrew Turner
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | | | - Angelica Martinez
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Caitlin Crook
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Amanda Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, California, USA
| | - Francesca Telese
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
8
|
Involvement of the ghrelin system in the maintenance and reinstatement of cocaine-motivated behaviors: a role of adrenergic action at peripheral β1 receptors. Neuropsychopharmacology 2022; 47:1449-1460. [PMID: 34923576 PMCID: PMC9206024 DOI: 10.1038/s41386-021-01249-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/22/2021] [Accepted: 11/28/2021] [Indexed: 12/30/2022]
Abstract
Cocaine addiction is a significant medical and public concern. Despite decades of research effort, development of pharmacotherapy for cocaine use disorder remains largely unsuccessful. This may be partially due to insufficient understanding of the complex biological mechanisms involved in the pathophysiology of this disorder. In the present study, we show that: (1) elevation of ghrelin by cocaine plays a critical role in maintenance of cocaine self-administration and cocaine-seeking motivated by cocaine-conditioned stimuli; (2) acquisition of cocaine-taking behavior is associated with the acquisition of stimulatory effects of cocaine by cocaine-conditioned stimuli on ghrelin secretion, and with an upregulation of ghrelin receptor mRNA levels in the ventral tegmental area (VTA); (3) blockade of ghrelin signaling by pretreatment with JMV2959, a selective ghrelin receptor antagonist, dose-dependently inhibits reinstatement of cocaine-seeking triggered by either cocaine or yohimbine in behaviorally extinguished animals with a history of cocaine self-administration; (4) JMV2959 pretreatment also inhibits brain stimulation reward (BSR) and cocaine-potentiated BSR maintained by optogenetic stimulation of VTA dopamine neurons in DAT-Cre mice; (5) blockade of peripheral adrenergic β1 receptors by atenolol potently attenuates the elevation in circulating ghrelin induced by cocaine and inhibits cocaine self-administration and cocaine reinstatement triggered by cocaine. These findings demonstrate that the endogenous ghrelin system plays an important role in cocaine-related addictive behaviors and suggest that manipulating and targeting this system may be viable for mitigating cocaine use disorder.
Collapse
|
9
|
Melis MR, Sanna F, Argiolas A. Dopamine, Erectile Function and Male Sexual Behavior from the Past to the Present: A Review. Brain Sci 2022; 12:brainsci12070826. [PMID: 35884633 PMCID: PMC9312911 DOI: 10.3390/brainsci12070826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Early and recent studies show that dopamine through its neuronal systems and receptor subtypes plays different roles in the control of male sexual behavior. These studies show that (i) the mesolimbic/mesocortical dopaminergic system plays a key role in the preparatory phase of sexual behavior, e.g., in sexual arousal, motivation and reward, whereas the nigrostriatal system controls the sensory-motor coordination necessary for copulation, (ii) the incertohypothalamic system is involved in the consummatory aspects of sexual behavior (penile erection and copulation), but evidence for its role in sexual motivation is also available, (iii) the pro-sexual effects of dopamine occur in concert with neural systems interconnecting the hypothalamus and preoptic area with the spinal cord, ventral tegmental area and other limbic brain areas and (iv) D2 and D4 receptors play a major role in the pro-sexual effects of dopamine. Despite some controversy, increases or decreases, respectively, of brain dopamine activity induced by drugs or that occur physiologically, usually improves or worsens, respectively, sexual activity. These findings suggest that an altered central dopaminergic tone plays a role in mental pathologies characterized by aberrant sexual behavior, and that pro-erectile D4 receptor agonists may be considered a new strategy for the treatment of erectile dysfunction in men.
Collapse
|
10
|
Glover ME, Unroe KA, Moughnyeh MM, McCoy C, Kerman IA, Clinton SM. Structural and metabolic activity differences in serotonergic cell groups in a rat model of individual differences of emotionality and stress reactivity. Neurosci Lett 2022; 784:136752. [PMID: 35753615 DOI: 10.1016/j.neulet.2022.136752] [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: 05/28/2021] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 11/27/2022]
Abstract
Serotonin regulates a diverse set of functions, including emotional behavior, cognition, sociability, appetite, and sleep. Serotonin is also a key trophic factor that shapes neurodevelopmental processes. Genetic and environmental factors that drive individual differences in the serotonergic system have the capacity to impact brain structure and behavior, and likely contribute to pathophysiological processes involved in neuropsychiatric disorders. Using adult rats selectively bred for low novelty exploration (Low Responders, LR), we previously demonstrated pronounced increases in the levels of their anxiety- and depression- relevant behaviors as compared to the selectively bred High Novelty Responder (HR) rats. These behavioral differences were accompanied by alterations in the expression of genes that regulate serotonin synthesis in the brainstem, and its signaling in the forebrain. The present study extends these observations with a focus on the organization and the metabolism of brainstem serotonin cell groups that provide serotonergic innervation of the hippocampus and other limbic regions of male HR/LR rats. Using design-based stereology, we found the median raphe (MnR) in adult male LR rats contains increased number of serotonergic neurons as compared to the HRs. This is preceded by an increase in the metabolic activity of the caudal dorsal raphe (DRC) and the intrafascicular DR (DRI) during early postnatal development. These findings suggest that structural and functional differences in the raphe-limbic projections shape behavioral inhibition throughout the lifespan.
Collapse
Affiliation(s)
| | - Keaton A Unroe
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA; Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | | | - Chelsea McCoy
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | - Ilan A Kerman
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA; Behavioral Health Service Line, Veterans Affairs Pittsburgh Health System, Pittsburgh, PA 15240, USA
| | | |
Collapse
|
11
|
Clinton SM, Unroe KA, Shupe EA, McCoy CR, Glover ME. Resilience to Stress: Lessons from Rodents about Nature versus Nurture. Neuroscientist 2022; 28:283-298. [PMID: 33567987 PMCID: PMC11092422 DOI: 10.1177/1073858421989357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Individual differences in human temperament influence how we respond to stress and can confer vulnerability (or resilience) to emotional disorders. For example, high levels of behavioral inhibition in children predict increased risk of mood and anxiety disorders in later life. The biological underpinnings of temperament are unknown, although improved understanding can offer insight into the pathogenesis of emotional disorders. Our laboratory has used a rat model of temperamental differences to study neurodevelopmental factors that lead to a highly inhibited, stress vulnerable phenotype. Selective breeding for high versus low behavioral response to novelty created two rat strains that exhibit dramatic behavior differences over multiple domains relevant to emotional disorders. Low novelty responder (bLR) rats exhibit high levels of behavioral inhibition, passive stress coping, anhedonia, decreased sociability and vulnerability to chronic stress compared to high novelty responders (bHRs). On the other hand, bHRs exhibit high levels of behavioral dis-inhibition, active coping, and aggression. This review article summarizes our work with the bHR/bLR model showing the developmental emergence of the bHR/bLR phenotypes, the role the environment plays in shaping it, and the involvement of epigenetic processes such as DNA methylation that mediate differences in emotionality and stress reactivity.
Collapse
Affiliation(s)
- Sarah M. Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Keaton A. Unroe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Elizabeth A. Shupe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Chelsea R. McCoy
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Matthew E. Glover
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| |
Collapse
|
12
|
Chang SE, Krueger LD, Flagel SB. Investigating individual differences in opioid-taking and opioid-seeking behavior in male rats. Psychopharmacology (Berl) 2022; 239:1065-1080. [PMID: 34981179 DOI: 10.1007/s00213-021-06023-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 11/04/2021] [Indexed: 12/27/2022]
Abstract
RATIONALE Understanding the behavioral and neurobiological factors that render some individuals more susceptible than others to opioid addiction will be critical in combatting the opioid crisis. OBJECTIVE The purpose of the current study was to determine if behavioral traits associated with an increased likelihood to take and seek cocaine are the same traits that render one more susceptible to opioid-taking and opioid-seeking behavior. Individual differences in the acquisition of remifentanil self-administration and subsequent cue-induced reinstatement of remifentanil-seeking behavior were investigated using two animal models: the high-responder (HR)/low-responder (LR) and sign-tracker (ST)/goal-tracker (GT) models. Relative to LR rats, HR rats show increased novelty-induced locomotion or "sensation-seeking" behavior, and are more likely to acquire cocaine-taking behavior and do so at a faster rate. Relative to GT rats, ST rats attribute greater incentive motivational value to reward cues and are more likely to exhibit reinstatement of cocaine-seeking behavior. RESULTS In contrast to previous work using cocaine, we did not observe individual differences with respect to the acquisition of remifentanil self-administration- or cue-induced reinstatement of remifentanil-seeking behavior within the context of either the HR/LR or ST/GT model. Thus, neither the sensation-seeking trait nor the propensity to attribute incentive motivational value to reward cues predicts remifentanil-taking or remifentanil-seeking behavior. CONCLUSIONS These findings suggest that different traits may confer the initiation of opioid- vs. cocaine-taking behavior, and the propensity to relapse to opioid- vs. cocaine-seeking. Additional studies are needed to identify which neurobehavioral constructs confer liability to opioid use and relapse.
Collapse
Affiliation(s)
- Stephen E Chang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Lauren D Krueger
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA.,Office of Comparative Medicine, University of Utah, Salt Lake City, UT, USA
| | - Shelly B Flagel
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA. .,Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Even‐Chen O, Herburg L, Kefalakes E, Urshansky N, Grothe C, Barak S. FGF2 is an endogenous regulator of alcohol reward and consumption. Addict Biol 2022; 27:e13115. [PMID: 34796591 DOI: 10.1111/adb.13115] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 11/30/2022]
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing disorder, characterized by escalating alcohol drinking and loss of control, with very limited available treatments. We recently reported that the expression of fibroblast growth factor 2 (Fgf2) is increased in the striatum of rodents following long-term excessive alcohol drinking and that the systemic or intra-striatal administration of recombinant FGF2 increases alcohol consumption. Here, we set out to determine whether the endogenous FGF2 plays a role in alcohol drinking and reward, by testing the behavioural phenotype of Fgf2 knockout mice. We found that Fgf2 deficiency resulted in decreased alcohol consumption when tested in two-bottle choice procedures with various alcohol concentrations. Importantly, these effects were specific for alcohol, as a natural reward (sucrose) or water consumption was not affected by Fgf2 deficiency. In addition, Fgf2 knockout mice failed to show alcohol-conditioned place preference (CPP) but showed normal fear conditioning, suggesting that deletion of the growth factor reduces alcohol's rewarding properties. Finally, Fgf2 knockout mice took longer to recover from the loss of righting reflex and showed higher blood alcohol concentrations when challenged with an intoxicating alcohol dose, suggesting that their ethanol metabolism might be affected. Together, our results show that the endogenous FGF2 plays a critical role in alcohol drinking and reward and indicate that FGF2 is a positive regulator of alcohol-drinking behaviours. Our findings suggest that FGF2 is a potential biomarker for problem alcohol drinking and is a potential target for pharmacotherapy development for AUD.
Collapse
Affiliation(s)
- Oren Even‐Chen
- School of Psychological Sciences Tel Aviv University Tel Aviv 69978 Israel
| | - Leonie Herburg
- Institute of Neuroanatomy and Cell Biology Hannover Medical School Carl‐Neuberg‐Straße 1 Hanover 30625 Germany
- Center for Systems Neuroscience (ZSN) Hannover Germany
| | - Ekaterini Kefalakes
- Institute of Neuroanatomy and Cell Biology Hannover Medical School Carl‐Neuberg‐Straße 1 Hanover 30625 Germany
- Center for Systems Neuroscience (ZSN) Hannover Germany
| | - Nataly Urshansky
- School of Psychological Sciences Tel Aviv University Tel Aviv 69978 Israel
| | - Claudia Grothe
- Institute of Neuroanatomy and Cell Biology Hannover Medical School Carl‐Neuberg‐Straße 1 Hanover 30625 Germany
- Center for Systems Neuroscience (ZSN) Hannover Germany
| | - Segev Barak
- School of Psychological Sciences Tel Aviv University Tel Aviv 69978 Israel
- Sagol School of Neuroscience Tel Aviv University Tel Aviv 69978 Israel
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences Tel Aviv University Tel Aviv 69978 Israel
| |
Collapse
|
14
|
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
|
15
|
Quigley JA, Logsdon MK, Graham BC, Beaudoin KG, Becker JB. Activation of G protein-coupled estradiol receptor 1 in the dorsolateral striatum enhances motivation for cocaine and drug-induced reinstatement in female but not male rats. Biol Sex Differ 2021; 12:46. [PMID: 34391470 PMCID: PMC8364009 DOI: 10.1186/s13293-021-00389-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/24/2021] [Indexed: 01/06/2023] Open
Abstract
Background Estradiol potentiates drug-taking behaviors, including motivation to self-administer cocaine and reinstatement of drug-seeking after extinction in females, but not males. The dorsolateral stratum (DLS) is a region of the brain implicated in mediating drug-seeking behaviors and, more specifically, is a target brain area to study how estradiol regulates these behaviors. The estradiol receptors α, β, and G protein-coupled estradiol receptor 1 (GPER1) are all present in the DLS. In this study, the effects of activating GPER1 in the DLS on drug-seeking are investigated. Methods Gonad-intact male and female rats were trained to self-administer cocaine (0.4 mg/kg/inf) on a fixed-ratio 1 schedule of reinforcement. For 4 weeks, animals underwent testing on a progressive ratio schedule of reinforcement to determine their motivation to attain cocaine. Halfway through progressive ratio testing, a selective agonist targeting GPER1 (G1) was administered intra-DLS to determine the contribution of GPER1 activation on motivation for cocaine. The effects of intra-DLS GPER1 activation on drug-induced reinstatement after extinction were subsequently determined. Results Activation of GPER1, via intra-DLS G1 administration, potentiated females’ motivation to self-administer cocaine. There was no effect of prior G1 treatment on extinction of cocaine-taking in females; however, G1 treatment resulted in greater drug-induced reinstatement (10 mg/kg cocaine, i.p.). There were no effects of intra-DLS GPER1 activation observed on motivation for cocaine or cocaine-induced reinstatement of responding in males. Conclusions These results support the conclusion that activation of GPER1 in the DLS enhances cocaine-seeking behaviors for female, but not male rats.
Collapse
Affiliation(s)
- Jacqueline A Quigley
- Psychology Department, University of Michigan, Ann Arbor, MI, 48109, USA. .,Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Molly K Logsdon
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Brianna C Graham
- Psychology Department, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kendra G Beaudoin
- Psychology Department, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jill B Becker
- Psychology Department, University of Michigan, Ann Arbor, MI, 48109, USA.,Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
16
|
Glover ME, Cohen JL, Singer JR, Sabbagh MN, Rainville JR, Hyland MT, Morrow CD, Weaver CT, Hodes GE, Kerman IA, Clinton SM. Examining the Role of Microbiota in Emotional Behavior: Antibiotic Treatment Exacerbates Anxiety in High Anxiety-Prone Male Rats. Neuroscience 2021; 459:179-197. [PMID: 33540050 PMCID: PMC7965353 DOI: 10.1016/j.neuroscience.2021.01.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023]
Abstract
Intestinal microbiota are essential for healthy gastrointestinal function and also broadly influence brain function and behavior, in part, through changes in immune function. Gastrointestinal disorders are highly comorbid with psychiatric disorders, although biological mechanisms linking these disorders are poorly understood. The present study utilized rats bred for distinct emotional behavior phenotypes to examine relationships between emotionality, the microbiome, and immune markers. Prior work showed that Low Novelty Responder (LR) rats exhibit high levels of anxiety- and depression-related behaviors as well as myriad neurobiological differences compared to High Novelty Responders (HRs). Here, we hypothesized that the divergent HR/LR phenotypes are accompanied by changes in fecal microbiome composition. We used next-generation sequencing to assess the HR/LR microbiomes and then treated adult HR/LR males with an antibiotic cocktail to test whether it altered behavior. Given known connections between the microbiome and immune system, we also analyzed circulating cytokines and metabolic factors to determine relationships between peripheral immune markers, gut microbiome components, and behavioral measures. There were no baseline HR/LR microbiome differences, and antibiotic treatment disrupted the microbiome in both HR and LR rats. Antibiotic treatment exacerbated aspects of HR/LR behavior, increasing LRs' already high levels of anxiety-like behavior while reducing passive stress coping in both strains. Our results highlight the importance of an individual's phenotype to their response to antibiotics, contributing to the understanding of the complex interplay between gut microbes, immune function, and an individual's emotional phenotype.
Collapse
Affiliation(s)
- M E Glover
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| | - J L Cohen
- Department of Psychiatry, University of California, San Francisco, CA, USA
| | - J R Singer
- MD/PhD Medical Scientist Training Program, University of Alabama-Birmingham, Birmingham, AL, USA
| | - M N Sabbagh
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - J R Rainville
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - M T Hyland
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - C D Morrow
- Department of Cell, Developmental, and Integrative Biology, University of Alabama-Birmingham, Birmingham, AL, USA
| | - C T Weaver
- Department of Pathology, University of Alabama-Birmingham, Birmingham, AL, USA
| | - G E Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Ilan A Kerman
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; Behavioral Health Service Line, Veterans Affairs Pittsburgh Health System, Pittsburgh, PA, USA
| | - S M Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
17
|
Doyle MR, Sulima A, Rice KC, Collins GT. MDPV self-administration in female rats: influence of reinforcement history. Psychopharmacology (Berl) 2021; 238:735-744. [PMID: 33236170 PMCID: PMC7914194 DOI: 10.1007/s00213-020-05726-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/16/2020] [Indexed: 11/26/2022]
Abstract
RATIONALE A subset of male rats that self-administer 3,4-methylenedioxypyrovalerone (MDPV) have unusually high levels of drug intake; however, factor(s) that influence this behavior (e.g., reinforcement history and sex) are unknown. OBJECTIVES Characterize the reinforcing potency and effectiveness of MDPV in female rats to determine whether (1) a subset of females also develop high levels of MDPV self-administration (i.e., a high-responder phenotype) and (2) the degree to which the high-responder phenotype is influenced by various reinforcement histories (i.e., responding for cocaine or food). METHODS Female Sprague Dawley rats initially responded for MDPV (0.032 mg/kg/infusion), cocaine (0.32 mg/kg/infusion), or food (45-mg grain pellet) under fixed ratio (FR) 1 and FR5 schedules of reinforcement. After 20 sessions, the cocaine- and food-history rats responded for MDPV for 20 additional sessions. Dose-response curves for MDPV were generated under FR5 and progressive ratio (PR) schedules of reinforcement. RESULTS A subset of rats responding for MDPV developed high levels of MDPV intake. A history of responding for cocaine, but not food, inhibited the development of high levels of MDPV intake. Large individual differences were observed in the level of self-administration when MDPV was available under an FR5, but not PR, schedule of reinforcement. CONCLUSIONS MDPV functions as a powerful reinforcer in female rats, as has been previously reported in male rats. The substantial variability in MDPV self-administration between subjects may be related to individual differences in human drug-taking behavior.
Collapse
Affiliation(s)
- Michelle R Doyle
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr - MC 7764, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Gregory T Collins
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr - MC 7764, San Antonio, TX, 78229, USA.
- South Texas Veterans Health Care System, San Antonio, TX, USA.
| |
Collapse
|
18
|
Clinton SM, Shupe EA, Glover ME, Unroe KA, McCoy CR, Cohen JL, Kerman IA. Modeling heritability of temperamental differences, stress reactivity, and risk for anxiety and depression: Relevance to research domain criteria (RDoC). Eur J Neurosci 2021; 55:2076-2107. [PMID: 33629390 PMCID: PMC8382785 DOI: 10.1111/ejn.15158] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/29/2021] [Accepted: 02/20/2021] [Indexed: 01/04/2023]
Abstract
Animal models provide important tools to study biological and environmental factors that shape brain function and behavior. These models can be effectively leveraged by drawing on concepts from the National Institute of Mental Health Research Domain Criteria (RDoC) Initiative, which aims to delineate molecular pathways and neural circuits that underpin behavioral anomalies that transcend psychiatric conditions. To study factors that contribute to individual differences in emotionality and stress reactivity, our laboratory utilized Sprague-Dawley rats that were selectively bred for differences in novelty exploration. Selective breeding for low versus high locomotor response to novelty produced rat lines that differ in behavioral domains relevant to anxiety and depression, particularly the RDoC Negative Valence domains, including acute threat, potential threat, and loss. Bred Low Novelty Responder (LR) rats, relative to their High Responder (HR) counterparts, display high levels of behavioral inhibition, conditioned and unconditioned fear, avoidance, passive stress coping, anhedonia, and psychomotor retardation. The HR/LR traits are heritable, emerge in the first weeks of life, and appear to be driven by alterations in the developing amygdala and hippocampus. Epigenomic and transcriptomic profiling in the developing and adult HR/LR brain suggest that DNA methylation and microRNAs, as well as differences in monoaminergic transmission (dopamine and serotonin in particular), contribute to their distinct behavioral phenotypes. This work exemplifies ways that animal models such as the HR/LR rats can be effectively used to study neural and molecular factors driving emotional behavior, which may pave the way toward improved understanding the neurobiological mechanisms involved in emotional disorders.
Collapse
Affiliation(s)
- Sarah M Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Elizabeth A Shupe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Matthew E Glover
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Keaton A Unroe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Chelsea R McCoy
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Joshua L Cohen
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Ilan A Kerman
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.,Behavioral Health Service Line, Veterans Affairs Pittsburgh Health System, Pittsburgh, PA, USA
| |
Collapse
|
19
|
Abstract
There are sex differences in the development of cocaine addiction. For example, the time that it takes for women from initial use to addiction is significantly shorter than for men. Thus, understanding why females are more vulnerable to cocaine addiction will provide insights into sex differences in the mechanisms underlying cocaine addiction. This study aimed to determine how cocaine demand intensity and elasticity might differ between sexes. In addition, the impact of estrous cycle and cocaine intake on demand was investigated. Male and female rats were trained to self-administer 0.125 mg of cocaine intravenously under a chained schedule in daily 2-h sessions for 2 weeks, and then, the cocaine demand function was determined with a modified within-session threshold procedure. Following the test, the rats began to self-administer a higher dose of cocaine (0.25 mg) to increase the cocaine intake. The demand function was then similarly determined in the same rats after 2 weeks of cocaine self-administration of the higher dose. No sex differences were found in either demand intensity or elasticity. Neither did the level of cocaine intake have an impact on demand. The demand elasticity, but not intensity, was significantly lower during proestrus/estrus compared with diestrus. These data suggest that the faster transition to cocaine addiction in women cannot be explained by sex differences in the demand for cocaine and such a demand may change during different phases of estrus cycle.
Collapse
Affiliation(s)
| | | | - Meiyun Fan
- Department of Pathology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
20
|
Carroll ME, Zlebnik NE, Holtz NA. Preference for Palatable Food, Impulsivity, and Relation to Drug Addiction in Rats. NEUROMETHODS 2021. [DOI: 10.1007/978-1-0716-0924-8_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Eck SR, Bangasser DA. The effects of early life stress on motivated behaviors: A role for gonadal hormones. Neurosci Biobehav Rev 2020; 119:86-100. [PMID: 33022296 PMCID: PMC7744121 DOI: 10.1016/j.neubiorev.2020.09.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/22/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
Motivated behaviors are controlled by the mesocorticolimbic dopamine (DA) system, consisting of projections from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) and prefrontal cortex (PFC), with input from structures including the medial preoptic area (mPOA). Sex differences are present in this circuit, and gonadal hormones (e.g., estradiol and testosterone) are important for regulating DA transmission. Early life stress (ELS) also regulates the mesocorticolimbic DA system. ELS modifies motivated behaviors and the underlying DA circuitry, increasing risk for disorders such as substance use disorder, major depression, and schizophrenia. ELS has been shown to change gonadal hormone signaling in both sexes. Thus, one way that ELS could impact mesocorticolimbic DA is by altering the efficacy of gonadal hormones. This review provides evidence for this idea by integrating the gonadal hormone, motivation, and ELS literature to argue that ELS alters gonadal hormone signaling to impact motivated behavior. We also discuss the importance of these effects in the context of understanding risk and treatments for psychiatric disorders in men and women.
Collapse
Affiliation(s)
- Samantha R Eck
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA.
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| |
Collapse
|
22
|
Bongiovanni AR, Peer K, Carpenter RE, Ellis AS, Duggan MR, Parikh V, Wimmer ME. Aging reduces the sensitivity to the reinforcing efficacy of morphine. Neurobiol Aging 2020; 97:28-32. [PMID: 33120086 DOI: 10.1016/j.neurobiolaging.2020.09.020] [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: 05/20/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 11/25/2022]
Abstract
The US geriatric population is growing and using more opioids than ever before. The purpose of this study was to determine whether aging influenced the reinforcing efficacy of morphine in male and female rats using a rodent intravenous self-administration paradigm. Male and female aged (20-24 months) and young (2-4 months) Wistar rats were tested at 2 doses of morphine (0.75 mg/kg/infusion and 0.25 mg/kg/infusion). During 10 days of self-administration, aged rats took significantly less morphine than their younger counterparts at the 0.25 mg/kg/infusion dose. Aged males also earned significantly fewer infusions on a progressive ration reinforcement schedule at this dose, suggesting that the reinforcing efficacy of morphine is decreased for this group at this dose. These effects dissipated when a separate group of animals had access to the 0.75 mg/kg/infusion dose for both sexes. Our results indicate that morphine is less reinforcing at lower doses in aged male, but not female rats. This research has potential clinical implications for the chronic treatments involving opioids in aged individuals.
Collapse
Affiliation(s)
- Angela R Bongiovanni
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Kyle Peer
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Rachel E Carpenter
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Alexandra S Ellis
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Michael R Duggan
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Kantak KM, Gauthier JM, Mathieson E, Knyazhanskaya E, Rodriguez-Echemendia P, Man HY. Sex differences in the effects of a combined behavioral and pharmacological treatment strategy for cocaine relapse prevention in an animal model of cue exposure therapy. Behav Brain Res 2020; 395:112839. [PMID: 32750464 DOI: 10.1016/j.bbr.2020.112839] [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: 04/27/2020] [Revised: 07/05/2020] [Accepted: 07/26/2020] [Indexed: 10/23/2022]
Abstract
Brief interventions of environmental enrichment (EE) or the glycine transporter-1 inhibitor Org24598 administered with cocaine-cue extinction training were shown previously to inhibit reacquisition of cocaine self-administration in male rats trained to self-administer a moderate 0.3 mg/kg dose of cocaine. Determining how EE and Org24598 synergize in combination in an animal model of cue exposure therapy is novel. Important changes made in this investigation were increasing the cocaine training dose to 1.0 mg/kg and determining sex differences. Adult male and female rats self-administering 1.0 mg/kg cocaine for 35-40 daily sessions exhibited an addiction-like phenotype under a second-order schedule of cocaine delivery and cue presentation. Rats next underwent 6 weekly extinction training sessions for which treatments consisted of EE or NoEE and Vehicle or Org24598 (3.0 mg/kg in males; 3.0 or 7.5 mg/kg in females). Rats then were tested for reacquisition of cocaine self-administration for 15 daily sessions. In males, the combined EE +3.0 mg/kg Org24598 treatment facilitated extinction learning and inhibited reacquisition of cocaine self-administration to a greater extent than no treatment and to individual EE or 3.0 mg/kg Org24598 treatments. In females, EE +7.5 mg/kg Org24598 facilitated extinction learning, but did not inhibit reacquisition of cocaine self-administration. Thus, there were sex differences in the ability of EE + Org24598 administered in conjunction with extinction training to inhibit cocaine relapse in rats exhibiting an addiction-like phenotype. These findings suggest that this multimodal treatment approach might be a feasible option during cue exposure therapy in cocaine-dependent men, but not women.
Collapse
Affiliation(s)
- Kathleen M Kantak
- Department of Psychological and Brain Sciences, Boston University, Boston, USA; Center for Systems Neuroscience, Boston University, Boston, USA.
| | - Jamie M Gauthier
- Department of Psychological and Brain Sciences, Boston University, Boston, USA
| | - Elon Mathieson
- Department of Psychological and Brain Sciences, Boston University, Boston, USA
| | | | | | - Heng-Ye Man
- Department of Biology, Boston University, Boston, USA; Center for Systems Neuroscience, Boston University, Boston, USA
| |
Collapse
|
24
|
Intermittent access cocaine self-administration produces psychomotor sensitization: effects of withdrawal, sex and cross-sensitization. Psychopharmacology (Berl) 2020; 237:1795-1812. [PMID: 32206828 PMCID: PMC7244391 DOI: 10.1007/s00213-020-05500-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/28/2020] [Indexed: 02/05/2023]
Abstract
RATIONALE With repeated administration, the psychomotor activating effects of drugs such as cocaine or amphetamine can change in very different ways-showing sensitization or tolerance-depending on whether they are administered more or less intermittently. This behavioral plasticity is thought to reflect, at least in part, changes in dopamine (DA) neurotransmission, and therefore, may provide insights into the development of substance use disorders. Indeed, the most widely used preclinical model of cocaine addiction, which involves Long Access (LgA) self-administration procedures, is reported to produce tolerance to cocaine's psychomotor activating effects and effects on DA activity. In contrast, Intermittent Access (IntA) cocaine self-administration is more effective than LgA in producing addiction-like behavior, but sensitizes DA neurotransmission. There is, however, very little information concerning the effects of IntA experience on the psychomotor activating effects of cocaine. OBJECTIVE The objective of this study was to determine whether IntA experience produces psychomotor sensitization with similar characteristics to that produced by the intermittent, noncontingent administration of cocaine. RESULTS IntA to cocaine did indeed produce psychomotor sensitization that (1) was greater after a long (30 days) vs. short (1 day) period of withdrawal, (2) was greater in females than males, and (3) resulted in cross-sensitization to another psychomotor stimulant drug, amphetamine. CONCLUSION The tolerance sometimes associated with LgA cocaine self-administration has been cited in support of the idea that, in addiction, drug-seeking and drug-taking is motivated to overcome a DA deficiency and associated anhedonia. In contrast, the neurobehavioral sensitization associated with IntA cocaine self-administration favors an incentive-sensitization view.
Collapse
|
25
|
Distinct relationships between risky decision making and cocaine self-administration under short- and long-access conditions. Prog Neuropsychopharmacol Biol Psychiatry 2020; 98:109791. [PMID: 31676462 PMCID: PMC7375467 DOI: 10.1016/j.pnpbp.2019.109791] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/01/2019] [Accepted: 10/16/2019] [Indexed: 11/23/2022]
Abstract
Substance use is strongly associated with impaired decision making, with cocaine use particularly linked to elevated risky and impulsive choice. It is not clear, however, whether such maladaptive decision making is a consequence of cocaine use or instead precedes and predisposes individuals to cocaine use. The current study was designed to specifically address the latter possibility with respect to risky choice in both male and female rats. Rats were first trained in a "Risky Decision-making Task" (RDT), in which they made discrete choices between a small, "safe" food reward and a large, "risky" food reward accompanied by increasing probabilities of mild footshock punishment. After reaching stable performance, rats underwent jugular catheter surgery followed by either short-access cocaine self-administration sessions (2 h, 0.5 mg/kg/infusion) for 5 days or long-access cocaine self-administration sessions (6 h, 0.5 mg/kg/infusion) for 14 days. Under short-access conditions, there was no relationship between risk preference and changes in cocaine intake over time, but greater risk aversion in females predicted greater overall cocaine intake. Under long-access conditions, heightened risk taking predicted greater escalation of cocaine intake over the course of self-administration, supporting the notion that pre-existing risk-taking behavior predicts cocaine intake. Collectively, results from these experiments have implications for understanding and identifying pre-existing vulnerabilities to substance use, which may lead to strategies to prevent development of substance use disorders.
Collapse
|
26
|
Castro-Zavala A, Martín-Sánchez A, Valverde O. Sex differences in the vulnerability to cocaine's addictive effects after early-life stress in mice. Eur Neuropsychopharmacol 2020; 32:12-24. [PMID: 31918976 DOI: 10.1016/j.euroneuro.2019.12.112] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/13/2019] [Accepted: 12/13/2019] [Indexed: 01/28/2023]
Abstract
Even though men are more likely to use drugs, women tend to progress faster from drug use to drug abuse, especially in the case of psychostimulants such as cocaine. Preclinical studies evaluating the differences in cocaine self-administration (SA) between sexes are contradictory. While some have shown no between-sex differences, others have reported female rodents to acquire higher percentages of cocaine SA criteria. Furthermore, early-life adversity is a risk factor for substance-use disorder and clinical evidence showed that women who have experienced childhood adversity are more likely to use drugs in comparison with males. However, the molecular differences between sexes as a consequence of early-life adversity or cocaine consumption have scarcely been explored. The aim of our study was to evaluate the differences in the expression of the GluA1, GluA2 subunits of AMPA receptors, pCREB and CREB in male and female mice exposed to maternal separation with early weaning (MSEW). Moreover, we evaluated the effects of cocaine SA in both sexes during adulthood, and the possible changes in GluA1, GluA2, pCREB and CREB expressions. Our results showed a higher acquisition percentage in females and an MSEW-induced increase in cocaine-seeking solely in males. Additionally, we observed sex differences in GluA1, GluA2, CREB and pCREB levels in the NAc and the VTA. The present results displayed changes in molecules that play a crucial role in the regulation of the rewarding effects of cocaine, helping to elucidate the mechanisms involved in the progression from cocaine use to cocaine abuse in both females and males.
Collapse
Affiliation(s)
- Adriana Castro-Zavala
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Ana Martín-Sánchez
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Dr. Aiguader 88, Barcelona 08003, Spain; Neuroscience Research Program, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Dr. Aiguader 88, Barcelona 08003, Spain; Neuroscience Research Program, IMIM-Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
27
|
Nawarawong NN, Olsen CM. Within-animal comparisons of novelty and cocaine neuronal ensemble overlap in the nucleus accumbens and prefrontal cortex. Behav Brain Res 2020; 379:112275. [PMID: 31614186 DOI: 10.1016/j.bbr.2019.112275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/28/2019] [Accepted: 10/01/2019] [Indexed: 11/29/2022]
Abstract
Novelty seeking is a personality trait associated with an increased vulnerability for substance abuse. In rodents, elevated novelty seeking has been shown to be a predictor for elevated drug self-administration and compulsive use. While previous studies have shown that both novelty and drugs of abuse have actions within similar mesocorticolimbic regions, little is known as to whether the same neural ensembles are engaged by these two stimuli. Using the TetTag mouse model and Fos immunohistochemistry, we measured neurons engaged by novelty and acute cocaine exposure, respectively in the prefrontal cortex (PFC) and nucleus accumbens (NAc). While there was no significant impact of novelty exposure on the size of the EGFP+ ensemble, we found that cocaine engaged significantly more Fos+ neurons in the NAc, while stress increased the size of the Fos+ ensemble in the PFC. Analysis of ensemble reactivation was specific to the emotional valence of the second stimuli. We found that a greater proportion of the EGFP+ ensemble was reactivated in the groups that paired novelty with a positive (cocaine) or neutral (saline) experience in the NAc, while the novelty/stress paired groups exhibited significantly less ensemble overlap in the PFC. However, only in the NAc shell was this increase in ensemble overlap specific to those exposed to both novelty and cocaine. This suggests that the NAc shell, but not the NAc core or PFC, may play an important role in general reward processing by engaging a similar network of neurons.
Collapse
Affiliation(s)
- Natalie N Nawarawong
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Christopher M Olsen
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
28
|
McCoy CR, Sabbagh MN, Huaman JP, Pickrell AM, Clinton SM. Oxidative metabolism alterations in the emotional brain of anxiety-prone rats. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109706. [PMID: 31330216 PMCID: PMC6708503 DOI: 10.1016/j.pnpbp.2019.109706] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/31/2019] [Accepted: 07/17/2019] [Indexed: 10/26/2022]
Abstract
Mood disorders such as anxiety and depression are heterogeneous disorders with many sufferers unresponsive to current pharmacological treatments. Individual differences in temperament represent one factor that may underlie symptom heterogeneity, so understanding its biological underpinnings can help pave the way to personalized therapies and improved patient outcomes. The present study uses a rodent model of temperamental differences to examine whether individual differences in emotional behavior phenotypes correspond to altered limbic brain cellular metabolism, an indicator of neuronal activity. The model uses two selectively bred rat lines - high novelty responder rats (HRs) that show highly exploratory behavior in a novel environment, active coping style and resilience to chronic mild stress compared to low novelty responder rats (LRs), which are inhibited in novel environments, display passive coping style, and are susceptible to chronic stress. Utilizing transcriptome data from a prior study in adult HR/LR rats, we first show that a preponderance of genes differing in the HR vs. LR hippocampus and amygdala are involved in cellular metabolism. This led us to then ask if oxygen consumption was altered in isolated mitochondria of the hippocampus and amygdala of HR/LR rats; here we found increased oxygen consumption reserve capacity in LR amygdala. Our last experiment examined activity of cytochrome c oxidase (COX), an enzyme responsible for ATP production and correlate of metabolic activity, in several brain regions of HR/LR rats. We found that LRs displayed higher COX activity in the dentate gyrus, prefrontal cortex, and dorsal raphe compared to HRs, with no significant HR/LR difference in nuclei of the amygdala. Correlational analyses of COX activity across brain regions suggested divergent connectivity between the prefrontal cortex, amygdala, hippocampus, and dorsal raphe of HR vs. LR rats. Together these studies point to altered cellular metabolism in the limbic brain of LR/HR animals, which may reflect altered neural circuitry that drives their divergent behavioral profiles.
Collapse
Affiliation(s)
| | | | | | | | - Sarah M. Clinton
- Corresponding author at: Integrated Life Sciences Building (ILSB), 1981 Kraft Drive, Blacksburg, VA, , Phone: (540) 231-5946
| |
Collapse
|
29
|
Klintsova AY, Hamilton DA, Mooney SM, Petrenko CL. Proceedings of the 2018 annual meeting of the Fetal Alcohol Spectrum Disorders study group. Alcohol 2019; 81:47-55. [PMID: 31173861 DOI: 10.1016/j.alcohol.2019.05.005] [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: 04/18/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 10/26/2022]
Abstract
The 2018 Fetal Alcohol Spectrum Disorders Study Group (FASDSG) meeting was entitled "Sex Differences and Vulnerability." The theme reflected the ongoing NIH initiative to address sex differences in both clinical and preclinical research. The first keynote speaker, Jill Becker, Ph.D., addressed sex differences in addiction in preclinical studies. The second keynote speaker, Meeyoung Min, Ph.D., discussed effects of gender on adolescent outcomes in poly-drug exposed children. The conference presented updates from three government agencies, a discussion panel of new data on FASD prevalence, and short presentations by junior and senior investigators showcasing late-breaking FASD research. The conference was capped by the presentation of Dr. Sarah Mattson, Ph.D., the recipient of the 2018 Henry Rosett award for career-long contributions to the field.
Collapse
|
30
|
Even-Chen O, Barak S. Inhibition of FGF Receptor-1 Suppresses Alcohol Consumption: Role of PI3 Kinase Signaling in Dorsomedial Striatum. J Neurosci 2019; 39:7947-7957. [PMID: 31375540 PMCID: PMC6774404 DOI: 10.1523/jneurosci.0805-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/20/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022] Open
Abstract
Excessive alcohol intake leads to mesostriatal neuroadaptations, and to addiction phenotypes. We recently found in rodents that alcohol increases fibroblast growth factor 2 (FGF2) expression in the dorsomedial striatum (DMS), which promotes alcohol consumption. Here, we show that systemic or intra-DMS blockade of the FGF2 receptor, FGF receptor-1 (FGFR1), suppresses alcohol consumption, and that the effects of FGF2-FGFR1 on alcohol drinking are mediated via the phosphoinositide 3 kinase (PI3K) signaling pathway. Specifically, we found that sub-chronic alcohol treatment (7 d × 2.5 g/kg, i.p.) increased Fgfr1 mRNA expression in the dorsal hippocampus and dorsal striatum. However, prolonged and excessive voluntary alcohol consumption in a two-bottle choice procedure increased Fgfr1 expression selectively in DMS. Importantly, systemic administration of the FGFR1 inhibitor PD173074 to mice, as well as its infusion into the DMS of rats, decreased alcohol consumption and preference, with no effects on natural reward consumption. Finally, inhibition of the PI3K, but not of the mitogen-activated protein kinase (MAPK) signaling pathway, blocked the effects of FGF2 on alcohol intake and preference. Our results suggest that activation of FGFR1 by FGF2 in the DMS leads to activation of the PI3K signaling pathway, which promotes excessive alcohol consumption, and that inhibition of FGFR1 may provide a novel therapeutic target for alcohol use disorder.SIGNIFICANCE STATEMENT Long-term alcohol consumption causes neuroadaptations in the mesostriatal reward system, leading to addiction-related behaviors. We recently showed that alcohol upregulates the expression of fibroblast growth factor 2 (FGF2) in dorsomedial striatum (DMS) or rats and mice, and in turn, FGF2 increases alcohol consumption. Here, we show that long-term alcohol intake also increases the expression of the FGF2 receptor, FGFR1 in the DMS. Importantly, inhibition of FGFR1 activity by a selective receptor antagonist reduces alcohol drinking, when given systemically or directly into the DMS. We further show that the effects of FGF2-FGFR1 on alcohol drinking are mediated via activation of the PI3K intracellular signaling pathway, providing an insight on the mechanism for this effect.
Collapse
Affiliation(s)
| | - Segev Barak
- School of Psychological Sciences, and
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| |
Collapse
|
31
|
Widman AJ, Cohen JL, McCoy CR, Unroe KA, Glover ME, Khan AU, Bredemann T, McMahon LL, Clinton SM. Rats bred for high anxiety exhibit distinct fear-related coping behavior, hippocampal physiology, and synaptic plasticity-related gene expression. Hippocampus 2019; 29:939-956. [PMID: 30994250 DOI: 10.1002/hipo.23092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/24/2019] [Accepted: 04/01/2019] [Indexed: 12/29/2022]
Abstract
The hippocampus is essential for learning and memory but also regulates emotional behavior. We previously identified the hippocampus as a major brain region that differs in rats bred for emotionality differences. Rats bred for low novelty response (LRs) exhibit high levels of anxiety- and depression-like behavior compared to high novelty responder (HR) rats. Manipulating the hippocampus of high-anxiety LR rats improves their behavior, although no work to date has examined possible HR/LR differences in hippocampal synaptic physiology. Thus, the current study examined hippocampal slice electrophysiology, dendritic spine density, and transcriptome profiling in HR/LR hippocampus, and compared performance on three hippocampus-dependent tasks: The Morris water maze, contextual fear conditioning, and active avoidance. Our physiology experiments revealed increased long-term potentiation (LTP) at CA3-CA1 synapses in HR versus LR hippocampus, and Golgi analysis found an increased number of dendritic spines in basal layer of CA1 pyramidal cells in HR versus LR rats. Transcriptome data revealed glutamate neurotransmission as the top functional pathway differing in the HR/LR hippocampus. Our behavioral experiments showed that HR/LR rats exhibit similar learning and memory capability in the Morris water maze, although the groups differed in fear-related tasks. LR rats displayed greater freezing behavior in the fear-conditioning task, and HR/LR rats adopted distinct behavioral strategies in the active avoidance task. In the active avoidance task, HRs avoided footshock stress by pressing a lever when presented with a warning cue; LR rats, on the other hand, waited until footshocks began before pressing the lever to stop them. Taken together, these findings concur with prior observations of HR rats generally exhibiting active stress coping behavior while LRs exhibit reactive coping. Overall, our current findings coupled with previous work suggest that HR/LR differences in stress reactivity and stress coping may derive, at least in part, from differences in the developing and adult hippocampus.
Collapse
Affiliation(s)
- Allie J Widman
- Department of Cellular, Developmental and Integrative Biology, University of Alabama, Birmingham, Alabama
| | - Joshua L Cohen
- Medical Scientist Training Program (MSTP), University of Alabama, Birmingham, Alabama
| | - Chelsea R McCoy
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Keaton A Unroe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia
| | - Matthew E Glover
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Anas U Khan
- Department of Cellular, Developmental and Integrative Biology, University of Alabama, Birmingham, Alabama
| | - Teruko Bredemann
- Department of Cellular, Developmental and Integrative Biology, University of Alabama, Birmingham, Alabama
| | - Lori L McMahon
- Department of Cellular, Developmental and Integrative Biology, University of Alabama, Birmingham, Alabama
| | - Sarah M Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| |
Collapse
|
32
|
Sex differences in incentive-sensitization produced by intermittent access cocaine self-administration. Psychopharmacology (Berl) 2019; 236:625-639. [PMID: 30368583 PMCID: PMC6401254 DOI: 10.1007/s00213-018-5091-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/21/2018] [Indexed: 10/28/2022]
Abstract
RATIONALE Intermittent Access (IntA) cocaine self-administration, which models intermittent patterns of cocaine use in humans during the transition to addiction, is especially effective in producing incentive-sensitization and other addiction-like behavior in male rats. However, female rats show more robust psychomotor sensitization than males, and following initial use, women develop problematic patterns of drug use more readily than men. We hypothesized, therefore, that female rats might be more susceptible to the incentive-sensitization produced by IntA experience. OBJECTIVE To assess changes in motivation for cocaine, using a behavioral economic indicator of cocaine demand ("elasticity" of demand curves), and other addiction-like behavior, as a function of IntA cocaine self-administration experience in male and female rats. RESULTS IntA experience produced a progressive increase in motivation for cocaine in both males and females, as indicated by a decrease in the elasticity of cocaine demand curves, and this persisted undiminished following 14 days of abstinence. However, IntA produced a more rapid and greater increase in motivation for cocaine (incentive-sensitization) in females than males. Females also consumed more cocaine than males, although this did not predict changes in motivation. On the other hand, there were no sex differences in the preferred level of cocaine consumption when cost was low (Q0), nor in cocaine- or cue-induced reinstatement of drug-seeking behavior. CONCLUSIONS The observation that females are more susceptible to incentive-sensitization when intermittently exposed to cocaine may provide a mechanism for the more rapid development of problematic drug use in females ("telescoping effect") reported in clinical studies.
Collapse
|
33
|
Rats selectively bred for showing divergent behavioral traits in response to stress or novelty or spontaneous yawning with a divergent frequency show similar changes in sexual behavior: the role of dopamine. Rev Neurosci 2018; 30:427-454. [DOI: 10.1515/revneuro-2018-0058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/26/2018] [Indexed: 02/07/2023]
Abstract
Abstract
Sexual behavior plays a fundamental role for reproduction in mammals and other animal species. It is characterized by an anticipatory and a consummatory phase, and several copulatory parameters have been identified in each phase, mainly in rats. Sexual behavior varies significantly across rats even when they are of the same strain and reared under identical conditions. This review shows that rats of the same strain selectively bred for showing a divergent behavioral trait when exposed to stress or novelty (i.e. Roman high and low avoidance rats, bred for their different avoidance response to the shuttle box, and high and low novelty exploration responders rats, bred for their different exploratory response to a novel environment) or a spontaneous behavior with divergent frequency (i.e. low and high yawning frequency rats, bred for their divergent yawning frequency) show similar differences in sexual behavior, mainly in copulatory pattern, but also in sexual motivation. As shown by behavioral pharmacology and intracerebral microdialysis experiments carried out mainly in Roman rats, these sexual differences may be due to a more robust dopaminergic tone present in the mesocorticolimbic dopaminergic system of one of the two sub-lines (e.g. high avoidance, high novelty exploration, and low yawning rat sub-lines). Thus, differences in genotype and/or in prenatal/postnatal environment lead not only to individual differences in temperament and environmental/emotional reactivity but also in sexual behavior. Because of the highly conserved mechanisms controlling reproduction in mammals, this may occur not only in rats but also in humans.
Collapse
|
34
|
Even-Chen O, Barak S. The role of fibroblast growth factor 2 in drug addiction. Eur J Neurosci 2018; 50:2552-2561. [PMID: 30144335 DOI: 10.1111/ejn.14133] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/08/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022]
Abstract
Fibroblast growth factor 2 (FGF2) is a member of the FGF-family, which consists of 22 members, with four known FGF receptors (five in humans). Over the last 30 years, FGF2 has been extensively studied for its role in cell proliferation, differentiation, growth, survival and angiogenesis during development, as well as for its role in adult neurogenesis and regenerative plasticity. Over the past decade, FGF2 has been implicated in learning and memory, as well as in several neuropsychiatric disorders, including anxiety, stress, depression and drug addiction. In this review, we present accumulating evidence indicating the involvement of FGF2 in neuroadaptations caused by drugs of abuse, namely, amphetamine, cocaine, nicotine and alcohol. Moreover, evidence suggests that FGF2 is a positive regulator of alcohol and drug-related behaviors. Thus, although additional studies are yet required, we suggest that reducing FGF2 activity may provide a novel therapeutic approach for substance use disorders.
Collapse
Affiliation(s)
- Oren Even-Chen
- School of Psychological Sciences, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Segev Barak
- School of Psychological Sciences, Tel Aviv University, 69978, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
35
|
A paternal methyl donor depleted diet leads to increased anxiety- and depression-like behavior in adult rat offspring. Biosci Rep 2018; 38:BSR20180730. [PMID: 29945927 PMCID: PMC6153370 DOI: 10.1042/bsr20180730] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022] Open
Abstract
Epigenetic mechanisms such as DNA methylation elicit lasting changes in gene expression and likely mediate gene-environment interactions that shape brain development, behavior, and emotional health. Myriad environmental factors influence DNA methylation, including methyl donor content in the paternal diet, could influence methylation in offspring via changes in the paternal germ line. The present study examines the effects of paternal methyl donor dietary deficiency on offspring's emotional behaviors, including anxiety, social interaction, and depression-like behavior. We previously found that rats bred to display high levels of anxiety- and depression-like behavior exhibit diminished DNA methylation in the amygdala. We also observed that depleting dietary methyl donor content exacerbated the rats' already high levels of anxiety- and depression-like behavior. Here we sought to determine whether paternal dietary methyl donor depletion elicits intergenerational effects on first generation (F1) offspring's behavior (potentially triggering a similar increase in anxiety- and/or depression-like behavior). Thus, adult male rats prone to high anxiety/depression-like behavior, were fed either a methyl donor depleted (DEP) or control (CON) diet for 5 weeks prior to mating. They were paired with females and resultant F1 male offspring were subjected to a behavioral test battery in adulthood. F1-DEP offspring showed a similar behavioral profile to the F0 males, including greater depression-like behavior in the forced swim test (FST) and increased anxiety-like behavior in the open field test (OFT). Future work will interrogate molecular changes in the brains of F1 offspring that mediate these intergenerational effects of paternal methyl donor dietary content on offspring emotional behavior.
Collapse
|
36
|
Swalve N, Smethells JR, Younk R, Mitchell J, Dougen B, Carroll ME. Sex-specific attenuation of impulsive action by progesterone in a go/no-go task for cocaine in rats. Psychopharmacology (Berl) 2018; 235:135-143. [PMID: 29018893 PMCID: PMC5892199 DOI: 10.1007/s00213-017-4750-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/20/2017] [Indexed: 12/14/2022]
Abstract
RATIONALE Previous work indicated that progesterone (PRO) reduced impulsive choice for cocaine in female but not male rats (Smethells et al. Psychopharmacology 233:2999-3008, 2016). Impulsive action, typically measured by responding for a reinforcer during a signaled period of nonavailability of natural reinforcers, predicts initiation and escalation of drug use in animals and humans. The present study examined impulsive action for cocaine using PRO in male and female rats trained on a go/no-go task. OBJECTIVE Rats were trained on a go/no-go task to respond for cocaine infusions (0.4 mg/kg/inf). During the "go" component, responding was reinforced on a VI 30-s schedule, whereas during the "no-go" component, withholding a response was reinforced on a differential reinforcement of other behavior (DRO) 30-s schedule. A response during the no-go component resets the DRO timer and served as a measure of impulsive action. After baseline responding was established, rats were pretreated with vehicle (VEH) or PRO (0.5 mg/kg), and DRO resets and responding during the go component for cocaine were compared in males vs. females. RESULTS DRO resets were significantly lower following PRO treatment compared to VEH in female, but not male, rats. Response rates and overall infusions during the go component were not significantly altered by PRO in either females or males. CONCLUSION Treatment with PRO resulted in a sex-specific reduction in impulsive action for cocaine, while not affecting cocaine self-administration.
Collapse
Affiliation(s)
- Natashia Swalve
- Department of Psychology, Alma College, Alma, MI, 48801, USA
| | - John R Smethells
- Department of Medicine, Minneapolis Medical Research Foundation, Minneapolis, MN, 55414, USA
| | - Rebecca Younk
- Department of Psychiatry, University of Minnesota, MMC 392 UMHC, Minneapolis, MN, 55455, USA
| | - Jared Mitchell
- Department of Psychiatry, University of Minnesota, MMC 392 UMHC, Minneapolis, MN, 55455, USA
| | - Ben Dougen
- Department of Psychiatry, University of Minnesota, MMC 392 UMHC, Minneapolis, MN, 55455, USA
| | - Marilyn E Carroll
- Department of Psychiatry, University of Minnesota, MMC 392 UMHC, Minneapolis, MN, 55455, USA.
| |
Collapse
|
37
|
Becker JB, McClellan ML, Reed BG. Sex differences, gender and addiction. J Neurosci Res 2017; 95:136-147. [PMID: 27870394 DOI: 10.1002/jnr.23963] [Citation(s) in RCA: 335] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/17/2016] [Accepted: 09/19/2016] [Indexed: 12/13/2022]
Abstract
This review discusses alcohol and other forms of drug addiction as both a sociocultural and biological phenomenon. Sex differences and gender are not solely determined by biology, nor are they entirely sociocultural. The interactions among biological, environmental, sociocultural, and developmental influences result in phenotypes that may be more masculine or more feminine. These gender-related sex differences in the brain can influence the responses to drugs of abuse, progressive changes in the brain after exposure to drugs of abuse and whether addiction results from drug-taking experiences. In addition, the basic laboratory evidence for sex differences is discussed within the context of four types of sex/gender differences. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jill B Becker
- Department of Psychology and the Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109
| | - Michele L McClellan
- Department of History and the Residential College, University of Michigan, Ann Arbor, MI, 48109
| | - Beth Glover Reed
- School of Social Work and the Department of Women's Studies, University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
38
|
Barker JM, Taylor JR. Sex differences in incentive motivation and the relationship to the development and maintenance of alcohol use disorders. Physiol Behav 2017; 203:91-99. [PMID: 28974459 DOI: 10.1016/j.physbeh.2017.09.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/08/2017] [Accepted: 09/30/2017] [Indexed: 10/18/2022]
Abstract
Despite considerable evidence of higher rates of alcohol use disorders (AUDs) in men than in women, there is a dearth of research into the underlying causes of this disparity. As the gap in high risk drinking between men and women closes, it is critical to disentangle the biological factors that may place men and women at different risk for the development of AUDs as well as AUD-associated health problems. While sex differences in alcohol drinking have been reported in animal models and in human alcoholics, it increasingly seems that consummatory behavior may be dissociated from propensity toward inflexible and cue-elicited drug seeking and taking that characterize alcohol use disorders. While much of this work was initially performed in males a growing, yet limited, body of literature suggests that there are sex differences in both cue reactivity, and further, the relationship between cue reactivity and the maintenance of addictive behavior, indicating that males may be at greater risk for the development of a subset of addiction-related behaviors independent of alcohol consumption. Here, we will review the current literature on sex effects on the relationship between incentive motivation and addictive behavior and discuss unanswered questions that we expect will inform the development of individualized and sex-specific treatment and prevention strategies for AUDs. We believe that a greater understanding of how sex interacts with in cue reactivity to independently mediate the drug taking and risk for the development of uncontrolled drug or alcohol-seeking and -taking will inform the development of individualized treatment and prevention strategies for addiction.
Collapse
Affiliation(s)
- Jacqueline M Barker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, United States.
| | - Jane R Taylor
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, United States
| |
Collapse
|
39
|
Abstract
Women exhibit more rapid escalation from casual drug taking to addiction, exhibit a greater withdrawal response with abstinence, and tend to exhibit greater vulnerability than men in terms of treatment outcome. In rodents, short-term estradiol intake in female rats enhances acquisition and escalation of drug taking, motivation for drugs of abuse, and relapse-like behaviors. There is also a sex difference in the dopamine response in the nucleus accumbens. Ovariectomized female rats exhibit a smaller initial dopamine increase after cocaine treatment than castrated males. Estradiol treatment of ovariectomized female rats enhances stimulated dopamine release in the dorsolateral striatum, but not in the nucleus accumbens, resulting in a sex difference in the balance between these two dopaminergic projections. In the situation where drug-taking behavior becomes habitual, dopamine release has been reported to be enhanced in the dorsolateral striatum and attenuated in the nucleus accumbens. The sex difference in the balance between these neural systems is proposed to underlie sex differences in addiction.
Collapse
Affiliation(s)
- Jill B Becker
- Department of Psychology, Molecular and Behavioral Neuroscience Institute, University of Michigan
| |
Collapse
|
40
|
Even-Chen O, Sadot-Sogrin Y, Shaham O, Barak S. Fibroblast Growth Factor 2 in the Dorsomedial Striatum Is a Novel Positive Regulator of Alcohol Consumption. J Neurosci 2017; 37:8742-8754. [PMID: 28821667 PMCID: PMC6596666 DOI: 10.1523/jneurosci.0890-17.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/16/2017] [Accepted: 07/31/2017] [Indexed: 12/13/2022] Open
Abstract
Repeated alcohol intake leads to mesostriatal neuroadaptations, resulting in drinking escalation and addiction phenotypes. Fibroblast growth factor 2 (FGF2) has been shown to interact with the mesostriatal dopaminergic system, and has been implicated in the actions of psychostimulants in the brain, and in several psychiatric disorders. Here, we report on a positive regulatory feedback loop of alcohol and FGF2 in rodent models. Specifically, we found that acute alcohol exposure (2.5 g/kg, i.p.) increased the mRNA expression of Fgf2 in the dorsal hippocampus, nucleus accumbens, and dorsal striatum. Longer alcohol exposure (7 d × 2.5 g/kg, i.p.) restricted these increases to the dorsal striatum, and the latter effect was blocked by the dopamine D2-like receptor antagonist haloperidol. Voluntary prolonged and excessive alcohol consumption in a 2-bottle choice procedure increased Fgf2 expression selectively in dorsomedial striatum (DMS) of both mice and rats. Importantly, we found that systemic administration of recombinant FGF2 (rFGF2) in mice, or rFGF2 infusion into the dorsal striatum or DMS of rats, increased alcohol consumption and preference, with no similar effects on saccharin or sucrose consumption. Finally, we found that inhibition of the endogenous FGF2 function in the DMS, by an anti-FGF2 neutralizing antibody, suppressed alcohol consumption and preference. Together, our results suggest that alcohol consumption increases the expression of Fgf2 in the DMS, and that striatal FGF2 promotes alcohol consumption, suggesting that FGF2 in the DMS is a positive regulator of alcohol drinking.SIGNIFICANCE STATEMENT Long-term alcohol intake may lead to neuroadaptations in the mesostriatal reward system, resulting in addiction phenotypes. Fibroblast growth factor 2 (FGF2) is crucial for the development and maintenance of the mesostriatal dopaminergic system. Here, we provide evidence for the involvement of FGF2 in alcohol-drinking behaviors. We show that alcohol increases Fgf2 expression in the dorsal striatum, an effect mediated via dopamine D2-like receptors. Importantly, we show that infusion of recombinant FGF2 into the dorsomedial striatum increases alcohol consumption, whereas inhibiting the endogenous FGF2 function suppresses consumption. Thus, FGF2 is an alcohol-responsive gene constituting a positive regulatory feedback loop with alcohol. This loop leads to facilitation of alcohol consumption, marking FGF2 as a potential new therapeutic target for alcohol addiction.
Collapse
Affiliation(s)
| | - Yossi Sadot-Sogrin
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | | | - Segev Barak
- School of Psychological Sciences and
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| |
Collapse
|
41
|
Datta U, Martini M, Sun WL. Sex Differences in the Motivational Contrast between Sucrose and Cocaine in Rats. JOURNAL OF DRUG DESIGN AND RESEARCH 2017; 4:1042. [PMID: 34622250 PMCID: PMC8494449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
There are sex differences in the vulnerability to cocaine abuse and addiction. Understanding the differences is critical for developing the sex-tailored prevention and treatment strategies. Cocaine addiction is characterized by the pathological motivation for cocaine accompanied by the diminished motivation for natural rewards. Thus, the motivational impact of cocaine relative to natural rewards likely determines the attractiveness of cocaine and likely plays a role in the vulnerability to cocaine abuse and addiction. This study aimed to determine whether the relative magnitudes or contrast of the motivational impact between cocaine and sucrose is different between sexes. To this end, cocaine-naïve out bred Wistar rats were trained to self-administer sucrose pellets and the motivation for different amounts of sucrose was then determined as the breakpoints under the progressive-ratio schedule of reinforcement. Following the sucrose tests, the same rats were trained to self-administer cocaine and the motivation for different doses of cocaine was similarly measured. For the female rats, the motivation was also measured during the diestrus and proestrus/estrus, respectively, to determine the impact of the estrous cycle on the motivational effects of cocaine and sucrose. The differences between the breakpoints of cocaine and sucrose were significantly larger in the males. The enhanced motivational contrast may contribute to the increased vulnerability to recreational cocaine abuse and addiction in the males.
Collapse
Affiliation(s)
| | | | - Wen Lin Sun
- Corresponding author Wen Lin Sun, Department of Pharmacology, University of Tennessee Health Science Center, 71 S. Manassas, Rm 205, Memphis, TN 38103, USA, Tel: 1-901-448-1354; Fax: 1-901-448-7206;
| |
Collapse
|
42
|
McCoy CR, Jackson NL, Day J, Clinton SM. Genetic predisposition to high anxiety- and depression-like behavior coincides with diminished DNA methylation in the adult rat amygdala. Behav Brain Res 2016; 320:165-178. [PMID: 27965039 DOI: 10.1016/j.bbr.2016.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/22/2016] [Accepted: 12/07/2016] [Indexed: 12/17/2022]
Abstract
Understanding biological mechanisms that shape vulnerability to emotional dysfunction is critical for elucidating the neurobiology of psychiatric illnesses like anxiety and depression. To elucidate molecular and epigenetic alterations in the brain that contribute to individual differences in emotionality, our laboratory utilized a rodent model of temperamental differences. Rats bred for low response to novelty (Low Responders, LRs) are inhibited in novel situations and display high anxiety, helplessness, and diminished sociability compared to High Novelty Responder (HR) rats. Our current transcriptome profiling experiment identified widespread gene expression differences in the amygdala of adult HR/LR rats; we hypothesize that HR/LR gene expression and downstream behavioral differences stem from distinct epigenetic (specifically DNA methylation) patterning in the HR/LR brain. Although we found similar levels of DNA methyltransferase proteins in the adult HR/LR amygdala, next-generation sequencing analysis of the methylome revealed 793 differentially methylated genomic sites between the groups. Most of the differentially methylated sites were hypermethylated in HR versus LR, so we next tested the hypothesis that enhancing DNA methylation in LRs would improve their anxiety/depression-like phenotype. We found that increasing DNA methylation in LRs (via increased dietary methyl donor content) improved their anxiety-like behavior and decreased their typically high levels of Forced Swim Test (FST) immobility; however, dietary methyl donor depletion exacerbated LRs' high FST immobility. These data are generally consistent with findings in depressed patients showing that treatment with DNA methylation-promoting agents improves depressive symptoms, and highlight epigenetic mechanisms that may contribute to individual differences in risk for emotional dysfunction.
Collapse
Affiliation(s)
- Chelsea R McCoy
- School of Neuroscience, Virginia Tech University, Blacksburg, VA 24060, USA
| | - Nateka L Jackson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham AL, USA
| | - Jeremy Day
- Department of Neurobiology, University of Alabama at Birmingham AL, USA
| | - Sarah M Clinton
- School of Neuroscience, Virginia Tech University, Blacksburg, VA 24060, USA.
| |
Collapse
|
43
|
Arenas MC, Aguilar MA, Montagud-Romero S, Mateos-García A, Navarro-Francés CI, Miñarro J, Rodríguez-Arias M. Influence of the Novelty-Seeking Endophenotype on the Rewarding Effects of Psychostimulant Drugs in Animal Models. Curr Neuropharmacol 2016; 14:87-100. [PMID: 26391743 PMCID: PMC4787288 DOI: 10.2174/1570159x13666150921112841] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/20/2015] [Accepted: 09/19/2015] [Indexed: 12/16/2022] Open
Abstract
Novelty seeking (NS), defined as a tendency to pursue novel and intense emotional sensations and experiences, is one of the most relevant individual factors predicting drug use among humans. High novelty seeking (HNS) individuals present an increased risk of drug use compared to low novelty seekers. The NS endophenotype may explain some of the differences observed among individuals exposed to drugs of abuse in adolescence. However, there is little research about the particular response of adolescents to drugs of abuse in function of this endophenotype, and the data that do exist are inconclusive. The present work reviews the literature regarding the influence of NS on psychostimulant reward, with particular focus on adolescent subjects. First, the different animal models of NS and the importance of this endophenotype in adolescence are discussed. Later, studies that have used the most common animal models of reward (self-administration, conditioned place preference paradigms) to evaluate how the NS trait influences the rewarding effects of psychostimulants are reviewed. Finally, possible explanations for the enhanced risk of developing substance dependence among HNS individuals are discussed. In conclusion, the studies referred to in this review show that the HNS trait is associated with: (1) increased initial sensitivity to the rewarding effects of psychostimulants, (2) a higher level of drug craving when the subject is exposed to the environmental cues associated with the drug, and (3) enhanced long-term vulnerability to relapse to drug consumption after prolonged abstinence.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marta Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Avda. Blasco Ibañez, 21, 46010, Valencia, Spain.
| |
Collapse
|
44
|
Abstract
The purpose of this review is to discuss ways to think about and study sex differences in preclinical animal models. We use the framework of addiction, in which animal models have excellent face and construct validity, to illustrate the importance of considering sex differences. There are four types of sex differences: qualitative, quantitative, population, and mechanistic. A better understanding of the ways males and females can differ will help scientists design experiments to characterize better the presence or absence of sex differences in new phenomena that they are investigating. We have outlined major quantitative, population, and mechanistic sex differences in the addiction domain using a heuristic framework of the three established stages of the addiction cycle: binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation. Female rats, in general, acquire the self-administration of drugs and alcohol more rapidly, escalate their drug taking with extended access more rapidly, show more motivational withdrawal, and (where tested in animal models of "craving") show greater reinstatement. The one exception is that female rats show less motivational withdrawal to alcohol. The bases for these quantitative sex differences appear to be both organizational, in that estradiol-treated neonatal animals show the male phenotype, and activational, in that the female phenotype depends on the effects of gonadal hormones. In animals, differences within the estrous cycle can be observed but are relatively minor. Such hormonal effects seem to be most prevalent during the acquisition of drug taking and less influential once compulsive drug taking is established and are linked largely to progesterone and estradiol. This review emphasizes not only significant differences in the phenotypes of females and males in the domain of addiction but emphasizes the paucity of data to date in our understanding of those differences.
Collapse
Affiliation(s)
- Jill B Becker
- Molecular & Behavioral Neuroscience Institute, Department of Psychiatry, Department of Psychology, University of Michigan, Ann Arbor, Michigan (J.B.B.); and Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (G.F.K.)
| | - George F Koob
- Molecular & Behavioral Neuroscience Institute, Department of Psychiatry, Department of Psychology, University of Michigan, Ann Arbor, Michigan (J.B.B.); and Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (G.F.K.)
| |
Collapse
|
45
|
Fattore L, Melis M. Sex differences in impulsive and compulsive behaviors: a focus on drug addiction. Addict Biol 2016; 21:1043-51. [PMID: 26935237 DOI: 10.1111/adb.12381] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 12/19/2022]
Abstract
Sex differences in inhibition and self-regulation at a behavioral level have been widely described. From an evolutionary point of view, the different selection pressures placed on male and female hominids led them to differ in their behavioral strategies that allowed our species to survive during natural selection processes. These differences reflect changes in neural and structural plasticity that might be the core of sex differences, and of the susceptibility towards one psychiatric condition rather than another. The goal of the present review is to summarize current evidence for such a dichotomy in impulsive and compulsive behavior with a focus on drug addiction. Sex-dependent differences in drug abuse and dependence will be examined in the context of pathophysiological regulation of impulse and motivation by neuromodulators (i.e. gonadal hormones) and neurotransmitters (i.e. dopamine). Advances in the understanding of the sex differences in the capability to control impulses and motivational states is key for the determination of efficacious biologically based intervention and prevention strategies for several neuropsychiatric disorders where loss of impulse control and compulsivity are the core symptoms.
Collapse
Affiliation(s)
- Liana Fattore
- Institute of Neuroscience-Cagliari National Research Council of Italy; Cittadella Universitaria di Monserrato; Italy
- Centre of Excellence ‘Neurobiology of Dependence’; University of Cagliari; Italy
| | - Miriam Melis
- Centre of Excellence ‘Neurobiology of Dependence’; University of Cagliari; Italy
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology; Cittadella Universitaria di Monserrato, University of Cagliari; Italy
| |
Collapse
|
46
|
Model-Free Temporal-Difference Learning and Dopamine in Alcohol Dependence: Examining Concepts From Theory and Animals in Human Imaging. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2016; 1:401-410. [DOI: 10.1016/j.bpsc.2016.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/09/2016] [Accepted: 06/14/2016] [Indexed: 02/04/2023]
|
47
|
Wingo T, Nesil T, Choi JS, Li MD. Novelty Seeking and Drug Addiction in Humans and Animals: From Behavior to Molecules. J Neuroimmune Pharmacol 2016; 11:456-70. [PMID: 26481371 PMCID: PMC4837094 DOI: 10.1007/s11481-015-9636-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 10/13/2015] [Indexed: 12/21/2022]
Abstract
Global treatment of drug addiction costs society billions of dollars annually, but current psychopharmacological therapies have not been successful at desired rates. The increasing number of individuals suffering from substance abuse has turned attention to what makes some people more vulnerable to drug addiction than others. One personality trait that stands out as a contributing factor is novelty seeking. Novelty seeking, affected by both genetic and environmental factors, is defined as the tendency to desire novel stimuli and environments. It can be measured in humans through questionnaires and in rodents using behavioral tasks. On the behavioral level, both human and rodent studies demonstrate that high novelty seeking can predict the initiation of drug use and a transition to compulsive drug use and create a propensity to relapse. These predictions are valid for several drugs of abuse, such as alcohol, nicotine, cocaine, amphetamine, and opiates. On the molecular level, both novelty seeking and addiction are modulated by the central reward system in the brain. Dopamine is the primary neurotransmitter involved in the overlapping neural substrates of both parameters. In sum, the novelty-seeking trait can be valuable for predicting individual vulnerability to drug addiction and for generating successful treatment for patients with substance abuse disorders.
Collapse
Affiliation(s)
- Taylor Wingo
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, 450 Ray C Hunt Drive, Suite G-170, Charlottesville, VA, 22903, USA
| | - Tanseli Nesil
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, 450 Ray C Hunt Drive, Suite G-170, Charlottesville, VA, 22903, USA
| | - Jung-Seok Choi
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, 450 Ray C Hunt Drive, Suite G-170, Charlottesville, VA, 22903, USA
- Department of Psychiatry, SMG-SNU Boramae Medical Center and Department of Psychiatry and Behavioral Science, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ming D Li
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, 450 Ray C Hunt Drive, Suite G-170, Charlottesville, VA, 22903, USA.
| |
Collapse
|
48
|
Goulopoulou S, McCarthy CG, Webb RC. Toll-like Receptors in the Vascular System: Sensing the Dangers Within. Pharmacol Rev 2016; 68:142-67. [PMID: 26721702 DOI: 10.1124/pr.114.010090] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Toll-like receptors (TLRs) are components of the innate immune system that respond to exogenous infectious ligands (pathogen-associated molecular patterns, PAMPs) and endogenous molecules that are released during host tissue injury/death (damage-associated molecular patterns, DAMPs). Interaction of TLRs with their ligands leads to activation of downstream signaling pathways that induce an immune response by producing inflammatory cytokines, type I interferons (IFN), and other inflammatory mediators. TLR activation affects vascular function and remodeling, and these molecular events prime antigen-specific adaptive immune responses. Despite the presence of TLRs in vascular cells, the exact mechanisms whereby TLR signaling affects the function of vascular tissues are largely unknown. Cardiovascular diseases are considered chronic inflammatory conditions, and accumulating data show that TLRs and the innate immune system play a determinant role in the initiation and development of cardiovascular diseases. This evidence unfolds a possibility that targeting TLRs and the innate immune system may be a novel therapeutic goal for these conditions. TLR inhibitors and agonists are already in clinical trials for inflammatory conditions such as asthma, cancer, and autoimmune diseases, but their study in the context of cardiovascular diseases is in its infancy. In this article, we review the current knowledge of TLR signaling in the cardiovascular system with an emphasis on atherosclerosis, hypertension, and cerebrovascular injury. Furthermore, we address the therapeutic potential of TLR as pharmacological targets in cardiovascular disease and consider intriguing research questions for future study.
Collapse
Affiliation(s)
- Styliani Goulopoulou
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - Cameron G McCarthy
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - R Clinton Webb
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| |
Collapse
|
49
|
Swalve N, Smethells JR, Zlebnik NE, Carroll ME. Sex differences in reinstatement of cocaine-seeking with combination treatments of progesterone and atomoxetine. Pharmacol Biochem Behav 2016; 145:17-23. [PMID: 27003832 DOI: 10.1016/j.pbb.2016.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/15/2016] [Accepted: 03/17/2016] [Indexed: 12/29/2022]
Abstract
Two repurposed medications have been proposed to treat cocaine abuse. Progesterone, a gonadal hormone, and atomoxetine, a medication commonly used to treat attention deficit/hyperactivity disorder, have both been separately shown to reduce cocaine self-administration and reinstatement (i.e., relapse). The goal of the present study was to examine sex differences in the individual effects of PRO and ATO as well as the combination PRO+ATO treatment on cocaine (COC), caffeine (CAF), and/or cue-primed reinstatement of cocaine-seeking. Adult male and female Wistar rats lever-pressed under a FR 1 schedule for cocaine infusions (0.4mg/kg/inf). After 14 sessions of stable responding in daily 2-h sessions, rats underwent a 21-day extinction period when no drug or drug-related stimuli were present. Rats were then separated into four groups that received PRO (0.5mg/kg) alone (PRO+SAL), ATO (1.5mg/kg) alone (VEH+ATO), control (VEH+SAL) or combination (PRO+ATO) treatments prior to the reinstatement condition. Reinstatement of cocaine-seeking to cues and/or drug injections of cocaine or caffeine was tested after extinction. During maintenance, females self-administered more cocaine than males, but no sex differences were seen during extinction. Females showed greater cocaine-seeking than males after a CAF priming injection. Individual treatment with ATO did not decrease reinstatement under any priming condition; however, the combination treatment decreased cocaine-seeking under the COC+CUES priming condition in males, and both PRO alone and the combination treatment decreased cocaine-seeking in the CAF+CUES condition in females. Overall, PRO alone was only effective in reducing reinstatement in females, while the combination treatment was consistently effective in reducing reinstatement in both sexes.
Collapse
Affiliation(s)
- Natashia Swalve
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - John R Smethells
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Natalie E Zlebnik
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Marilyn E Carroll
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
50
|
Sex differences in the acquisition and maintenance of cocaine and nicotine self-administration in rats. Psychopharmacology (Berl) 2016; 233:1005-13. [PMID: 26685990 PMCID: PMC4761341 DOI: 10.1007/s00213-015-4183-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/03/2015] [Indexed: 10/22/2022]
Abstract
RATIONALE Consistent sex differences are observed in human drug addiction, with females often exceeding males on drug intake. However, there is still a need for animal models for some aspects of addiction such as acquisition of drug self-administration and the subsequent development of drug-seeking. OBJECTIVES The present study examined sex differences in the acquisition and maintenance of self-administration of two widely used stimulants, cocaine and nicotine. METHODS Male and female rats self-administered cocaine (0.4 mg/kg/infusion) or nicotine (0.03 mg/kg/infusion) daily under a fixed-ratio 1 (FR 1) schedule until acquisition criteria were met (maximum of 30 sessions). The self-administration criterion for cocaine was ≥20 infusions in a 2-h session and ≥5 infusions in a 1-h session for nicotine. Sex differences were assessed by examining the percentage of rats that met acquisition criteria, the number of sessions to meet criteria, and the number of infusions earned during the maintenance phase. RESULTS A significantly higher percentage of male rats acquired both cocaine and nicotine self-administration than females, and males met acquisition criteria in fewer sessions. However, after criteria were met, females self-administered more cocaine than males during the first 5 days of maintenance. There were no sex differences in nicotine infusions post-acquisition. CONCLUSIONS Differences in acquisition amongst sexes can reveal factors that are integral to initiation of drug use, an often overlooked phase of drug addiction.
Collapse
|