1
|
Wallace CW, Holleran KM, Slinkard CY, Centanni SW, Lapish CC, Jones SR. Kappa opioid receptors diminish spontaneous dopamine signals in awake mice through multiple mechanisms. Neuropharmacology 2025; 273:110458. [PMID: 40204058 DOI: 10.1016/j.neuropharm.2025.110458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/06/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
The role of the dynorphin/kappa opioid receptor (KOR) system in dopamine (DA) regulation has been extensively investigated. KOR activation reduces extracellular DA concentrations, but the exact mechanism(s) through which this is accomplished are not fully elucidated. To explore KOR influences on real-time DA fluctuations, we used the photosensor dLight1.2 with fiber photometry in the nucleus accumbens (NAc) core of freely moving male and female C57BL/6J mice. First, we established that the rise and fall of spontaneously arising DA signals were due to DA release and reuptake, respectively. Next, mice were systemically administered the KOR agonist U50,488H in the presence or absence of the KOR antagonist aticaprant. U50,488H reduced both the amplitude and width of spontaneous signals in both sexes. Further, the slope of the correlation between amplitude and width was increased, indicating that DA uptake rates were increased. U50,488H also reduced the frequency of occurrence of signals in males and females. The effects of KOR activation were stronger in males, while effects of KOR antagonism were stronger in females. Overall, KORs exerted significant inhibitory control over spontaneous DA signaling, acting through at least three mechanisms - inhibiting DA release, promoting DA transporter-mediated uptake, and reducing the frequency of signals.
Collapse
Affiliation(s)
- Conner W Wallace
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Katherine M Holleran
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Clare Y Slinkard
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Samuel W Centanni
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Christopher C Lapish
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sara R Jones
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
2
|
Vashisht A, Adamson G, Gacso Z, Slama J, Freund M, Vinod S, Sandoval N, Nachshon Z, Gubin S, Corso E, You ZB, Ranaldi R, Galaj E. Environmental enrichment attenuates reinstatement of heroin seeking and reverses heroin-induced upregulation of mesolimbic ghrelin receptors. Drug Alcohol Depend 2025; 270:112635. [PMID: 40022817 PMCID: PMC11908936 DOI: 10.1016/j.drugalcdep.2025.112635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025]
Abstract
We have shown that environmental enrichment (EE) can effectively reduce reinstatement and facilitate true abstinence in animal models of drug use. Here, we investigated whether EE is effective against reinstatement of heroin seeking in long access (LA) model, which has been argued to capture the compulsive features of human drug addiction. We also explored the neurobiology by which EE produces its anti-drug addiction effects. In particular, we focused here on the ghrelin system, which is known for its involvement in reward-motivated behaviors and upregulation following intravenous drug self-administration. Following LA to heroin, rats were housed in either non-EE or EE conditions. During extinction and cue-induced reinstatement test, EE rats showed a significant reduction in active lever responding compared to non-EE rats, suggesting that EE facilitates extinction of drug seeking and reduces the capacity of drug-associated stimuli to elicit and maintain drug seeking. Using Western Blotting, we found that rats with LA to heroin IVSA showed a significant increase in ghrelin receptor (GHS-R1a) expression in the ventral tegmental area and nucleus accumbens, the brain regions implicated in resumption of drug use . Exposure to EE attenuated heroin-induced upregulation of GHS-R1a receptor in these regions but produced no significant changes other brain regions. Our findings suggest that EE can be an effective behavioral approach to diminish drug seeking even following LA to heroin. Compulsive drug taking and seeking seem to be correlated with an upregulation of GHS-R1a expression in the limbic regions, and EE can reverse these neuroadaptations, potentially contributing to a reduction in drug seeking.
Collapse
Affiliation(s)
- Apoorva Vashisht
- Department of Biology, Graduate Center, City University of New York, NY, USA; Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - George Adamson
- Psychological and Brain Sciences, Colgate University, Hamilton, NY, USA
| | - Zuzu Gacso
- Psychological and Brain Sciences, Colgate University, Hamilton, NY, USA
| | - Joseph Slama
- Psychological and Brain Sciences, Colgate University, Hamilton, NY, USA
| | - Matthew Freund
- Psychological and Brain Sciences, Colgate University, Hamilton, NY, USA
| | - Sneha Vinod
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Natalie Sandoval
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Ziv Nachshon
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Sami Gubin
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Elizabeth Corso
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Zhi-Bing You
- Neuropsychopharmacology Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, USA
| | - Robert Ranaldi
- Department of Biology, Graduate Center, City University of New York, NY, USA; Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Ewa Galaj
- Psychological and Brain Sciences, Colgate University, Hamilton, NY, USA.
| |
Collapse
|
3
|
Fouyssac M, Hynes T, Belin‐Rauscent A, Joshi D, Belin D. Incentive Cocaine-Seeking Habits and Their Compulsive Manifestation Emerge After a Downregulation of the Dopamine Transporter in Astrocytes Across Functional Domains of the Striatum. Eur J Neurosci 2025; 61:e70054. [PMID: 40082733 PMCID: PMC11906910 DOI: 10.1111/ejn.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/03/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
The development of compulsive cue-controlled-incentive drug-seeking habits is a hallmark of substance use disorder that is predicated on an intrastriatal shift in the locus of control over behaviour from a nucleus accumbens (Nac) core-dorsomedial striatum network to a Nac core-anterior dorsolateral striatum (aDLS) network. This shift is paralleled by drug-induced (including cocaine) dopamine transporter (DAT) alterations originating in the ventral striatum that spread eventually to encompass the aDLS. Having recently shown that heroin self-administration results in a pan-striatal reduction in astrocytic DAT that precedes the development of aDLS dopamine-dependent incentive heroin-seeking habits, we tested the hypothesis that similar adaptations occur following cocaine exposure. We compared DAT protein levels in whole tissue homogenates, and in astrocytes cultured from ventral and dorsal striatal territories of drug-naïve male Sprague-Dawley rats to those of rats with a history of cocaine taking or an aDLS dopamine-dependent incentive cocaine-seeking habit. Cocaine exposure resulted in a decrease in whole tissue and astrocytic DAT across all territories of the striatum. We further demonstrated that compulsive (i.e., punishment-resistant) incentive cocaine-seeking habits were associated with a reduction in DAT mRNA levels in the Nac shell, but not the Nac core-aDLS incentive habit system. Together with the recent evidence of heroin-induced downregulation of striatal astrocytic DAT, these findings suggest that alterations in astrocytic DAT may represent a common mechanism underlying the development of compulsive incentive drug-seeking habits across drug classes.
Collapse
Affiliation(s)
| | - Tristan Hynes
- Department of PsychologyUniversity of CambridgeCambridgeUK
| | | | | | - David Belin
- Department of PsychologyUniversity of CambridgeCambridgeUK
| |
Collapse
|
4
|
Del Percio C, Lizio R, Lopez S, Noce G, Jakhar D, Carpi M, Bölükbaş B, Soricelli A, Salvatore M, Güntekin B, Yener G, Massa F, Arnaldi D, Famà F, Pardini M, Ferri R, Salerni M, Lanuzza B, Stocchi F, Vacca L, Coletti C, Marizzoni M, Taylor JP, Hanoğlu L, Helvacı Yılmaz N, Kıyı İ, Özbek-İşbitiren Y, Frisoni GB, Cuoco S, Barone P, D'Anselmo A, Bonanni L, Biundo R, D'Antonio F, Bruno G, Giubilei F, De Pandis F, Rotondo R, Antonini A, Babiloni C. Resting-state electroencephalographic rhythms depend on sex in patients with dementia due to Parkinson's and Lewy Body diseases: An exploratory study. Neurobiol Dis 2025; 206:106807. [PMID: 39855475 DOI: 10.1016/j.nbd.2025.106807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 01/05/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Parkinson's disease with dementia (PDD) and dementia with Lewy bodies (DLB) are more prevalent in males than females. Furthermore, they typically showed abnormally high delta (< 4 Hz) and low alpha (8-10 Hz) rhythms from resting-state electroencephalographic (rsEEG) activity. Here, we hypothesized that those abnormalities may depend on the patient's sex. An international database provided clinical-demographic-rsEEG datasets for cognitively unimpaired older (Healthy; N = 49; 24 females), PDD (N = 39; 13 females), and DLB (N = 38; 15 females) participants. Each group was stratified into matched female and male subgroups. The rsEEG rhythms were investigated across the individual rsEEG delta, theta, and alpha frequency bands based on the individual alpha frequency peak. The eLORETA freeware was used to estimate cortical rsEEG sources. In the Healthy group, widespread rsEEG alpha source activities were greater in the females than in the males. In the PDD group, widespread rsEEG delta source activities were lower and widespread rsEEG alpha source activities were greater in the females than in the males. In the DLB group, central-parietal rsEEG delta source activities were lower, and posterior rsEEG alpha source activities were greater in the females than in the males. These results suggest sex-dependent hormonal modulation of neuroprotective-compensatory neurophysiological mechanisms in PDD and DLB patients underlying the generation of rsEEG delta and alpha rhythms, which should be considered in the treatment of vigilance dysregulation in those patients.
Collapse
Affiliation(s)
- Claudio Del Percio
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | - Roberta Lizio
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy; Oasi Research Institute - IRCCS, Troina, Italy.
| | - Susanna Lopez
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | | | - Dharmendra Jakhar
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | - Matteo Carpi
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | - Burcu Bölükbaş
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | - Andrea Soricelli
- IRCCS Synlab SDN, Naples, Italy; Department of Medical, Movement and Well-being Sciences, University of Naples Parthenope, Naples, Italy
| | | | - Bahar Güntekin
- Department of Biophysics, School of Medicine, Istanbul Medipol University, Istanbul, Turkey; Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Görsev Yener
- Department of Neurology, Faculty of Medicine, Dokuz Eylül University, İzmir, Turkey; IBG: International Biomedicine and Genome Center, Izmir, Turkey
| | - Federico Massa
- Dipartimento di Neuroscienze, Oftalmologia, Genetica, Riabilitazione e Scienze Materno-infantili (DiNOGMI), Università di Genova, Italy; Clinica neurologica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Dario Arnaldi
- Dipartimento di Neuroscienze, Oftalmologia, Genetica, Riabilitazione e Scienze Materno-infantili (DiNOGMI), Università di Genova, Italy; Neurofisiopatologia, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Famà
- Dipartimento di Neuroscienze, Oftalmologia, Genetica, Riabilitazione e Scienze Materno-infantili (DiNOGMI), Università di Genova, Italy; Neurofisiopatologia, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Matteo Pardini
- Dipartimento di Neuroscienze, Oftalmologia, Genetica, Riabilitazione e Scienze Materno-infantili (DiNOGMI), Università di Genova, Italy; Neurofisiopatologia, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | | | | | - Fabrizio Stocchi
- IRCCS San Raffaele, Rome, Italy; Telematic University San Raffaele, Rome, Italy
| | | | | | - Moira Marizzoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - John Paul Taylor
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - Lutfu Hanoğlu
- Department of Neurology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Nesrin Helvacı Yılmaz
- Medipol University Istanbul Parkinson's Disease and Movement Disorders Center (PARMER), Istanbul, Turkey
| | - İlayda Kıyı
- Health Sciences Institute, Department of Neurosciences, Dokuz Eylül University, Izmir, Turkey
| | - Yağmur Özbek-İşbitiren
- Health Sciences Institute, Department of Neurosciences, Dokuz Eylül University, Izmir, Turkey
| | - Giovanni B Frisoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Memory Clinic and LANVIE - Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Sofia Cuoco
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Neuroscience Section, University of Salerno, Baronissi, Italy
| | - Paolo Barone
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Neuroscience Section, University of Salerno, Baronissi, Italy
| | - Anita D'Anselmo
- Department of Aging Medicine and Sciences, University "G. d'Annunzio" of Chieti-Pescara, Italy
| | - Laura Bonanni
- Department of Aging Medicine and Sciences, University "G. d'Annunzio" of Chieti-Pescara, Italy
| | - Roberta Biundo
- Department of Neuroscience, University of Padua, Padua, PD, Italy
| | - Fabrizia D'Antonio
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Bruno
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Franco Giubilei
- Department of Neuroscience, Mental Health, and Sensory Organs, Sapienza University of Rome, Rome, Italy
| | - Francesca De Pandis
- Department of Human Sciences and Promotion of the Quality of Life, University San Raffaele Roma, Italy; Hospital San Raffaele Cassino, Cassino, FR, Italy
| | | | - Angelo Antonini
- Department of Neuroscience, University of Padua, Padua, PD, Italy
| | - Claudio Babiloni
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy; Hospital San Raffaele Cassino, Cassino, FR, Italy
| |
Collapse
|
5
|
Van Zandt M, Pittenger C. Sexual dimorphism in histamine regulation of striatal dopamine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.20.595049. [PMID: 38826392 PMCID: PMC11142073 DOI: 10.1101/2024.05.20.595049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Dopamine modulation of the basal ganglia differs in males and females and is implicated in numerous neuropsychiatric conditions, including some, like Tourette Syndrome (TS) and attention deficit hyperactivity disorder (ADHD), that have marked sex differences in prevalence. Genetic studies in TS and subsequent work in animals suggest that a loss of histamine may contribute to dysregulation of dopamine. Motivated by this, we characterized the modulation of striatal dopamine by histamine, using microdialysis, targeted pharmacology, and shRNA knockdown of histamine receptors. Intracerebroventricular (ICV) histamine reduced striatal dopamine in male mice, replicating previous work. In contrast, and unexpectedly, ICV histamine increased striatal dopamine in females. ICV or targeted infusion of agonists revealed that the effect in males depends on H2R receptors in the substantia nigra pars compacta (SNc). Knockdown of H2R in SNc GABAergic neurons abrogated the effect, identifying these cells as a key locus of histamine's regulation of dopamine in males. In females, however, H2R had no discernible role; instead, H3R agonists in the striatum increased striatal dopamine. Strikingly, the effect of histamine on dopamine in females was modulated by the estrous cycle, appearing only in estrus/proestrus, when estrogen levels are high. These findings confirm the regulation of striatal dopamine by histamine but identify marked sexual dimorphism in and estrous modulation of this effect. These findings may shed light on the mechanistic underpinnings of sex differences in the striatal circuitry, and in several neuropsychiatric conditions.
Collapse
Affiliation(s)
- Meghan Van Zandt
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA, 06519
| | - Christopher Pittenger
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA, 06519
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA, 06519
- Department of Psychology, Yale School of Arts and Sciences, New Haven, USA, 06519
- Center for Brain and Mind Health, Yale University School of Medicine, New Haven, USA, 06519
- Wu-Tsai Institute, Yale University, New Haven, CT, USA, 06519
| |
Collapse
|
6
|
Fry BR, Russell N, Fex V, Mo B, Pence N, Beatty JA, Manfredsson FP, Toth BA, Burgess CR, Gershman S, Johnson AW. Devaluing memories of reward: a case for dopamine. Commun Biol 2025; 8:161. [PMID: 39900665 PMCID: PMC11790953 DOI: 10.1038/s42003-024-07440-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Midbrain dopamine cells encode differences in predictive and expected value to support learning through reward prediction error. Recent findings have questioned whether reward prediction error can fully account for dopamine function and suggest a more complex role for dopamine in encoding detailed features of the reward environment. In this series of studies, we describe a novel role for dopamine in devaluing sensory features of reward. Mesencephalic dopamine cells activated during a mediated devaluation phase were later chemogenetically reactivated. This retrieval of the devalued reward memory elicited a reduction in the hedonic evaluation of sucrose reward. Through optogenetic and chemogenetic manipulations, we confirm dopamine cells are both sufficient and necessary for mediated devaluation, and retrieval of these memories reflected dopamine release in the nucleus accumbens. Consistent with our computational modeling data, our findings indicate a critical role for dopamine in encoding predictive representations of the sensory features of reinforcement. Overall, we elucidate a novel role for dopamine function in mediated devaluation and illuminate a more elaborate framework through which dopamine encodes reinforcement signals.
Collapse
Affiliation(s)
- Benjamin R Fry
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, USA
| | - Nicolette Russell
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Victoria Fex
- Lyman Briggs College, Michigan State University, East Lansing, MI, USA
| | - Bing Mo
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Nathan Pence
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Joseph A Beatty
- Department of Physiology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Fredric P Manfredsson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Brandon A Toth
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Samuel Gershman
- Department of Psychology, Harvard University, Cambridge, MA, USA
| | - Alexander W Johnson
- Department of Psychology, Michigan State University, East Lansing, MI, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
7
|
Hagarty-Waite KA, Emmons HA, Fordahl SC, Erikson KM. The Influence of Strain and Sex on High Fat Diet-Associated Alterations of Dopamine Neurochemistry in Mice. Nutrients 2024; 16:3301. [PMID: 39408267 PMCID: PMC11479034 DOI: 10.3390/nu16193301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Objective: The objective of this study was to determine the influence of sex and strain on striatal and nucleus accumbens dopamine neurochemistry and dopamine-related behavior due to a high-saturated-fat diet (HFD). Methods: Male and female C57B6/J (B6J) and Balb/cJ (Balb/c) mice were randomly assigned to a control-fat diet (CFD) containing 10% kcal fat/g or a mineral-matched HFD containing 60% kcal fat/g for 12 weeks. Results: Intraperitoneal glucose tolerance testing (IPGTT) and elevated plus maze experiments (EPM) confirmed that an HFD produced marked blunting of glucose clearance and increased anxiety-like behavior, respectively, in male and female B6J mice. Electrically evoked dopamine release in the striatum and reuptake in the nucleus accumbens (NAc), as measured by ex vivo fast scan cyclic voltammetry, was reduced for HFD-fed B6J females. Impairment in glucose metabolism explained HFD-induced changes in dopamine neurochemistry for B6J males and, to a lesser extent, Balb/c males. The relative expressions of protein markers associated with the activation of microglia, ionized calcium binding adaptor molecule (Iba1) and cluster of differentiation molecule 11b (CD11b) in the striatum were increased due to an HFD for B6J males but were unchanged or decreased amongst HFD-fed Balb/c mice. Conclusions: Our findings demonstrate that strain and sex influence the insulin- and microglia-dependent mechanisms of alterations to dopamine neurochemistry and associated behavior due to an HFD.
Collapse
Affiliation(s)
| | | | | | - Keith M. Erikson
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (K.A.H.-W.); (H.A.E.); (S.C.F.)
| |
Collapse
|
8
|
Pedrón VT, Canero EM, Varani AP, Aon AJ, Maldonado R, Balerio GN. Baclofen prevents morphine rewarding effects and associated biochemical alterations in male and female mice. Eur J Pharmacol 2024; 979:176768. [PMID: 39002637 DOI: 10.1016/j.ejphar.2024.176768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 07/15/2024]
Abstract
Previous studies from our laboratory have shown sex differences in the behavioral, molecular, and neurochemical manifestations of morphine withdrawal and they were related to an increased sensitivity to morphine effects in males. In addition, we observed an interaction between the GABAergic and opioid systems that could also be sex-dependent. Baclofen, a GABAB receptor agonist, prevented the somatic expression and the molecular and neurochemical changes induced by morphine withdrawal syndrome in mice. On the contrary, little is known about baclofen effects in the rewarding properties of morphine in male and female mice. The present study aimed to explore the effect of baclofen (1, 2 and 3 mg/kg, i.p.) pretreatment in the rewarding effects induced by morphine (7 mg/kg, s.c.) and its effect on c-Fos and brain-derived neurotrophic factor (BDNF) expression induced by the rewarding properties of morphine in prepubertal male and female mice. Baclofen (2 mg/kg) pretreatment prevented the rewarding effects of morphine only in male mice, while baclofen (3 mg/kg) reduced these effects in both sexes. Moreover, the rewarding effects of morphine were associated with a decrease of BDNF and c-Fos expression cingulate cortex, nucleus accumbens shell, cornu ammonis 1 (CA1), and cornu ammonis 3 (CA3) areas of the hippocampus only in male mice. In addition, baclofen pretreatment prevented these changes in BDNF, but not in c-Fos expression. In conclusion, our results show that GABAB receptors have a regulatory role in the rewarding effects of morphine that could be of interest for a potential future therapeutic application in opioid use disorders.
Collapse
Affiliation(s)
- Valeria T Pedrón
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-CONICET), Junín 956, 5° Piso, C1113AAD, Buenos Aires, Argentina
| | - Eliana M Canero
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-CONICET), Junín 956, 5° Piso, C1113AAD, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Junín 956, 5° Piso, C1113AAD, Buenos Aires, Argentina
| | - Andrés P Varani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-CONICET), Junín 956, 5° Piso, C1113AAD, Buenos Aires, Argentina
| | - Amira J Aon
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-CONICET), Junín 956, 5° Piso, C1113AAD, Buenos Aires, Argentina
| | - Rafael Maldonado
- Laboratori de Neurofarmacologia, Facultat de Ciències de La Salut I de La Vida, Universitat Pompeu Fabra, C/Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Graciela N Balerio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-CONICET), Junín 956, 5° Piso, C1113AAD, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Junín 956, 5° Piso, C1113AAD, Buenos Aires, Argentina.
| |
Collapse
|
9
|
Lenoir M, Engeln M, Navailles S, Girardeau P, Ahmed SH. A large-scale c-Fos brain mapping study on extinction of cocaine-primed reinstatement. Neuropsychopharmacology 2024; 49:1459-1467. [PMID: 38664549 PMCID: PMC11251268 DOI: 10.1038/s41386-024-01867-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 07/17/2024]
Abstract
Individuals with cocaine addiction can experience many craving episodes and subsequent relapses, which represents the main obstacle to recovery. Craving is often favored when abstinent individuals ingest a small dose of cocaine, encounter cues associated with drug use or are exposed to stressors. Using a cocaine-primed reinstatement model in rat, we recently showed that cocaine-conditioned interoceptive cues can be extinguished with repeated cocaine priming in the absence of drug reinforcement, a phenomenon we called extinction of cocaine priming. Here, we applied a large-scale c-Fos brain mapping approach following extinction of cocaine priming in male rats to identify brain regions implicated in processing the conditioned interoceptive stimuli of cocaine priming. We found that cocaine-primed reinstatement is associated with increased c-Fos expression in key brain regions (e.g., dorsal and ventral striatum, several prefrontal areas and insular cortex), while its extinction mostly disengages them. Moreover, while reinstatement behavior was correlated with insular and accumbal activation, extinction of cocaine priming implicated parts of the ventral pallidum, the mediodorsal thalamus and the median raphe. These brain patterns of activation and inhibition suggest that after repeated priming, interoceptive signals lose their conditioned discriminative properties and that action-outcome associations systems are mobilized in search for new contingencies, a brain state that may predispose to rapid relapse.
Collapse
Affiliation(s)
- Magalie Lenoir
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France.
| | - Michel Engeln
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France.
| | | | - Paul Girardeau
- Univ. Bordeaux, UFR des Sciences Odontologiques, Bordeaux, France
| | - Serge H Ahmed
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France
| |
Collapse
|
10
|
Kim DH, Loke H, Thompson J, Hill R, Sundram S, Lee J. The dopamine D2-like receptor and the Y-chromosome gene, SRY, are reciprocally regulated in the human male neuroblastoma M17 cell line. Neuropharmacology 2024; 251:109928. [PMID: 38552780 DOI: 10.1016/j.neuropharm.2024.109928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/28/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Affiliation(s)
- Dong-Hyun Kim
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Hannah Loke
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
| | - James Thompson
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Rachel Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia; Mental Health Program, Monash Health, Clayton, Victoria, 3168, Australia
| | - Joohyung Lee
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia; Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, 3168, Australia.
| |
Collapse
|
11
|
Kim J, Vanrobaeys Y, Davatolhagh MF, Kelvington B, Chatterjee S, Ferri SL, Angelakos C, Mills AA, Fuccillo MV, Nickl-Jockschat T, Abel T. A chromosome region linked to neurodevelopmental disorders acts in distinct neuronal circuits in males and females to control locomotor behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594746. [PMID: 38952795 PMCID: PMC11216371 DOI: 10.1101/2024.05.17.594746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Biological sex shapes the manifestation and progression of neurodevelopmental disorders (NDDs). These disorders often demonstrate male-specific vulnerabilities; however, the identification of underlying mechanisms remains a significant challenge in the field. Hemideletion of the 16p11.2 region (16p11.2 del/+) is associated with NDDs, and mice modeling 16p11.2 del/+ exhibit sex-specific striatum-related phenotypes relevant to NDDs. Striatal circuits, crucial for locomotor control, consist of two distinct pathways: the direct and indirect pathways originating from D1 dopamine receptor (D1R) and D2 dopamine receptor (D2R) expressing spiny projection neurons (SPNs), respectively. In this study, we define the impact of 16p11.2 del/+ on striatal circuits in male and female mice. Using snRNA-seq, we identify sex- and cell type-specific transcriptomic changes in the D1- and D2-SPNs of 16p11.2 del/+ mice, indicating distinct transcriptomic signatures in D1-SPNs and D2-SPNs in males and females, with a ∼5-fold greater impact in males. Further pathway analysis reveals differential gene expression changes in 16p11.2 del/+ male mice linked to synaptic plasticity in D1- and D2-SPNs and GABA signaling pathway changes in D1-SPNs. Consistent with our snRNA-seq study revealing changes in GABA signaling pathways, we observe distinct changes in miniature inhibitory postsynaptic currents (mIPSCs) in D1- and D2-SPNs from 16p11.2 del/+ male mice. Behaviorally, we utilize conditional genetic approaches to introduce the hemideletion selectively in either D1- or D2-SPNs and find that conditional hemideletion of genes in the 16p11.2 region in D2-SPNs causes hyperactivity in male mice, but hemideletion in D1-SPNs does not. Within the striatum, hemideletion of genes in D2-SPNs in the dorsal lateral striatum leads to hyperactivity in males, demonstrating the importance of this striatal region. Interestingly, conditional 16p11.2 del/+ within the cortex drives hyperactivity in both sexes. Our work reveals that a locus linked to NDDs acts in different striatal circuits, selectively impacting behavior in a sex- and cell type-specific manner, providing new insight into male vulnerability for NDDs. Highlights - 16p11.2 hemideletion (16p11.2 del/+) induces sex- and cell type-specific transcriptomic signatures in spiny projection neurons (SPNs). - Transcriptomic changes in GABA signaling in D1-SPNs align with changes in inhibitory synapse function. - 16p11.2 del/+ in D2-SPNs causes hyperactivity in males but not females. - 16p11.2 del/+ in D2-SPNs in the dorsal lateral striatum drives hyperactivity in males. - 16p11.2 del/+ in cortex drives hyperactivity in both sexes. Graphic abstract
Collapse
|
12
|
Wallace CW, Holleran KM, Slinkard CY, Centanni SW, Jones SR. Kappa Opioid Receptors Negatively Regulate Real Time Spontaneous Dopamine Signals by Reducing Release and Increasing Uptake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578840. [PMID: 38370660 PMCID: PMC10871279 DOI: 10.1101/2024.02.05.578840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The role of the dynorphin/kappa opioid receptor (KOR) system in dopamine (DA) regulation has been extensively investigated. KOR activation reduces extracellular DA concentrations and increases DA transporter (DAT) activity and trafficking to the membrane. To explore KOR influences on real-time DA fluctuations, we used the photosensor dLight1.2 with fiber photometry in the nucleus accumbens (NAc) core of freely moving male and female C57BL/6 mice. First, we established that the rise and fall of spontaneous DA signals were due to DA release and reuptake, respectively. Then mice were systemically administered the KOR agonist U50,488H (U50), with or without pretreatment with the KOR antagonist aticaprant (ATIC). U50 reduced both the amplitude and width of spontaneous signals in males, but only reduced width in females. Further, the slope of the correlation between amplitude and width was increased in both sexes, suggesting that DA uptake rates were increased. U50 also reduced the frequency of signals in both males and females. All effects of KOR activation were stronger in males. Overall, KORs exerted significant inhibitory control over spontaneous DA signaling, acting through at least three mechanisms - inhibiting DA release, promoting DAT-mediated uptake, and reducing the frequency of signals.
Collapse
Affiliation(s)
- Conner W Wallace
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Katherine M Holleran
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Clare Y Slinkard
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Samuel W Centanni
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Sara R Jones
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
13
|
Proaño SB, Miller CK, Krentzel AA, Dorris DM, Meitzen J. Sex steroid hormones, the estrous cycle, and rapid modulation of glutamatergic synapse properties in the striatal brain regions with a focus on 17β-estradiol and the nucleus accumbens. Steroids 2024; 201:109344. [PMID: 37979822 PMCID: PMC10842710 DOI: 10.1016/j.steroids.2023.109344] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/28/2023] [Accepted: 11/15/2023] [Indexed: 11/20/2023]
Abstract
The striatal brain regions encompassing the nucleus accumbens core (NAcc), shell (NAcs) and caudate-putamen (CPu) regulate cognitive functions including motivated behaviors, habit, learning, and sensorimotor action, among others. Sex steroid hormone sensitivity and sex differences have been documented in all of these functions in both normative and pathological contexts, including anxiety, depression and addiction. The neurotransmitter glutamate has been implicated in regulating these behaviors as well as striatal physiology, and there are likewise documented sex differences in glutamate action upon the striatal output neurons, the medium spiny neurons (MSNs). Here we review the available data regarding the role of steroid sex hormones such as 17β-estradiol (estradiol), progesterone, and testosterone in rapidly modulating MSN glutamatergic synapse properties, presented in the context of the estrous cycle as appropriate. Estradiol action upon glutamatergic synapse properties in female NAcc MSNs is most comprehensively discussed. In the female NAcc, MSNs exhibit development period-specific sex differences and estrous cycle variations in glutamatergic synapse properties as shown by multiple analyses, including that of miniature excitatory postsynaptic currents (mEPSCs). Estrous cycle-differences in NAcc MSN mEPSCs can be mimicked by acute exposure to estradiol or an ERα agonist. The available evidence, or lack thereof, is also discussed concerning estrogen action upon MSN glutamatergic synapse in the other striatal regions as well as the underexplored roles of progesterone and testosterone. We conclude that there is strong evidence regarding estradiol action upon glutamatergic synapse function in female NAcs MSNs and call for more research regarding other hormones and striatal regions.
Collapse
Affiliation(s)
- Stephanie B Proaño
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Christiana K Miller
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Amanda A Krentzel
- Office of Research and Innovation, North Carolina State University, Raleigh, NC, USA
| | - David M Dorris
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - John Meitzen
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
14
|
Cubello J, Marvin E, Conrad K, Merrill AK, George JV, Welle K, Jackson BP, Chalupa D, Oberdörster G, Sobolewski M, Cory-Slechta DA. The contributions of neonatal inhalation of copper to air pollution-induced neurodevelopmental outcomes in mice. Neurotoxicology 2024; 100:55-71. [PMID: 38081392 PMCID: PMC10842733 DOI: 10.1016/j.neuro.2023.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Exposures to ambient ultrafine particle (UFP) air pollution (AP) during the early postnatal period in mice (equivalent to human third trimester brain development) produce male-biased changes in brain structure, including ventriculomegaly, reduced brain myelination, alterations in neurotransmitters and glial activation, as well as impulsive-like behavioral characteristics, all of which are also features characteristic of male-biased neurodevelopmental disorders (NDDs). The purpose of this study was to ascertain the extent to which inhaled Cu, a common contaminant of AP that is also dysregulated across multiple NDDs, might contribute to these phenotypes. For this purpose, C57BL/6J mice were exposed from postnatal days 4-7 and 10-13 for 4 hr/day to inhaled copper oxide (CuxOy) nanoparticles at an environmentally relevant concentration averaging 171.9 ng/m3. Changes in brain metal homeostasis and neurotransmitter levels were determined following termination of exposure (postnatal day 14), while behavioral changes were assessed in adulthood. CuxOy inhalation modified cortical metal homeostasis and produced male-biased disruption of striatal neurotransmitters, with marked increases in dopaminergic function, as well as excitatory/inhibitory imbalance and reductions in serotonergic function. Impulsive-like behaviors in a fixed ratio (FR) waiting-for-reward schedule and a fixed interval (FI) schedule of food reward occurred in both sexes, but more prominently in males, effects which could not be attributed to altered locomotor activity or short-term memory. Inhaled Cu as from AP exposures, at environmentally relevant levels experienced during development, may contribute to impaired brain function, as shown by its ability to disrupt brain metal homeostasis and striatal neurotransmission. In addition, its ability to evoke impulsive-like behavior, particularly in male offspring, may be related to striatal dopaminergic dysfunction that is known to mediate such behaviors. As such, regulation of air Cu levels may be protective of public health.
Collapse
Affiliation(s)
- Janine Cubello
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Elena Marvin
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Katherine Conrad
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Alyssa K Merrill
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jithin V George
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Kevin Welle
- Proteomics Core, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brian P Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - David Chalupa
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Günter Oberdörster
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
15
|
Fernandes L, Kleene R, Congiu L, Freitag S, Kneussel M, Loers G, Schachner M. CHL1 depletion affects dopamine receptor D2-dependent modulation of mouse behavior. Front Behav Neurosci 2023; 17:1288509. [PMID: 38025382 PMCID: PMC10665519 DOI: 10.3389/fnbeh.2023.1288509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The dopaminergic system plays a key role in the appropriate functioning of the central nervous system, where it is essential for emotional balance, arousal, reward, and motor control. The cell adhesion molecule close homolog of L1 (CHL1) contributes to dopaminergic system development, and CHL1 and the dopamine receptor D2 (D2R) are associated with mental disorders like schizophrenia, addiction, autism spectrum disorder and depression. Methods Here, we investigated how the interplay between CHL1 and D2R affects the behavior of young adult male and female wild-type (CHL+/+) and CHL1-deficient (CHL1-/-) mice, when D2R agonist quinpirole and antagonist sulpiride are applied. Results Low doses of quinpirole (0.02 mg/kg body weight) induced hypolocomotion of CHL1+/+ and CHL1-/- males and females, but led to a delayed response in CHL1-/- mice. Sulpiride (1 mg/kg body weight) affected locomotion of CHL1-/- females and social interaction of CHL1+/+ females as well as social interactions of CHL1-/- and CHL1+/+ males. Quinpirole increased novelty-seeking behavior of CHL1-/- males compared to CHL1+/+ males. Vehicle-treated CHL1-/- males and females showed enhanced working memory and reduced stress-related behavior. Discussion We propose that CHL1 regulates D2R-dependent functions in vivo. Deficiency of CHL1 leads to abnormal locomotor activity and emotionality, and to sex-dependent behavioral differences.
Collapse
Affiliation(s)
- Luciana Fernandes
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ludovica Congiu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Freitag
- Institut für Molekulare Neurogenetik, Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Kneussel
- Institut für Molekulare Neurogenetik, Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
16
|
Hersey M, Chen AY, Bartole MK, Anand J, Newman AH, Tanda G. An FSCV Study on the Effects of Targeted Typical and Atypical DAT Inhibition on Dopamine Dynamics in the Nucleus Accumbens Shell of Male and Female Mice. ACS Chem Neurosci 2023; 14:2802-2810. [PMID: 37466616 PMCID: PMC10766117 DOI: 10.1021/acschemneuro.3c00354] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
Understanding the neurochemistry underlying sex differences in psychostimulant use disorders (PSUD) is essential for developing related therapeutics. Many psychostimulants, like cocaine, inhibit the dopamine transporter (DAT), which is largely thought to account for actions related to their misuse and dependence. Cocaine-like, typical DAT inhibitors preferentially bind DAT in an outward-facing conformation, while atypical DAT inhibitors, like modafinil, prefer a more inward-facing DAT conformation. Modafinil and R-modafinil have emerged as potential therapeutic options for selected populations of individuals affected by PSUD. In addition, analogs of modafinil (JJC8-088 and JJC8-091) with different pharmacological profiles have been explored as potential PSUD medications in preclinical models. In this work, we employ fast scan cyclic voltammetry (FSCV) to probe nucleus accumbens shell (NAS) dopamine (DA) dynamics in C57BL/6 male and female mice. We find that cocaine slowed DA clearance in both male and female mice but produced more robust increases in evoked NAS DA in female mice. R-Modafinil produced mild increases in evoked NAS DA and slowed DA clearance across the sexes. The modafinil analog JJC8-088, a typical DAT inhibitor, produced increases in evoked NAS DA in female and male mice. Finally, JJC8-091, an atypical DAT inhibitor, produced limited increases in evoked NAS DA and slowed DA clearance in both sexes. In this work we begin to tease out how sex differences may alter the effects of DAT targeting and highlight how this may help focus research toward effective treatment options for PSUD.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
| | - Andy Y. Chen
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
| | | | - Jayati Anand
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
| | - Amy Hauck Newman
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
- Medicinal Chemistry Section, NIDA IRP, Baltimore, MD 21224
| | - Gianluigi Tanda
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
| |
Collapse
|
17
|
Hersey M, Bartole MK, Jones CS, Newman AH, Tanda G. Are There Prevalent Sex Differences in Psychostimulant Use Disorder? A Focus on the Potential Therapeutic Efficacy of Atypical Dopamine Uptake Inhibitors. Molecules 2023; 28:5270. [PMID: 37446929 PMCID: PMC10343811 DOI: 10.3390/molecules28135270] [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] [Received: 04/21/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Psychostimulant use disorders (PSUD) affect a growing number of men and women and exert sizable public health and economic burdens on our global society. Notably, there are some sex differences in the onset of dependence, relapse rates, and treatment success with PSUD observed in preclinical and clinical studies. The subtle sex differences observed in the behavioral aspects of PSUD may be associated with differences in the neurochemistry of the dopaminergic system between sexes. Preclinically, psychostimulants have been shown to increase synaptic dopamine (DA) levels and may downregulate the dopamine transporter (DAT). This effect is greatest in females during the high estradiol phase of the estrous cycle. Interestingly, women have been shown to be more likely to begin drug use at younger ages and report higher levels of desire to use cocaine than males. Even though there is currently no FDA-approved medication, modafinil, a DAT inhibitor approved for use in the treatment of narcolepsy and sleep disorders, has shown promise in the treatment of PSUD among specific populations of affected individuals. In this review, we highlight the therapeutic potential of modafinil and other atypical DAT inhibitors focusing on the lack of sex differences in the actions of these agents.
Collapse
Affiliation(s)
| | | | | | | | - Gianluigi Tanda
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA; (M.H.); (M.K.B.); (C.S.J.); (A.H.N.)
| |
Collapse
|
18
|
Davis DL, Metzger DB, Vann PH, Wong JM, Shetty RA, Forster MJ, Sumien N. Effects of chronic methamphetamine exposure on rewarding behavior and neurodegeneration markers in adult mice. Psychopharmacology (Berl) 2023; 240:1343-1358. [PMID: 37127834 DOI: 10.1007/s00213-023-06374-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 04/27/2023] [Indexed: 05/03/2023]
Abstract
Recreational and medical use of stimulants among young adults have gained popularity in the United States over the last decade and their use may increase vulnerability to brain biochemical changes and addictive behaviors. The long-term effects of chronic stimulant exposure in later adulthood have not been fully elucidated.Our study investigated whether chronic exposure to methamphetamine (METH), at a dose designed to emulate human therapeutic dosing for ADHD, would promote biochemical alterations and affect sensitivity to the rewarding effects of subsequent METH dosing.Groups of 3.5-month-old male and female C57BL/6J mice were administered non-contingent intraperitoneal injections of either saline or METH (1.4 mg/kg) twice a day for 1 month (5 days/week). METH (0.5 mg/kg)-induced conditioned place preference (CPP) was tested in mice to determine the effects of previous METH exposure on reward-related behavior. Mice were randomly assigned to Experiment I (males and females) or Experiment II (females only) in which CPP testing was respectively performed either 0.5 or 5 months after the end of METH injections, at ~5 or 10 months old respectively. The midbrain and striatum, regions involved in reward circuit, were assessed for markers associated with neurotoxicity, dopaminergic function, neuroinflammation and epigenetic changes after behavioral testing.Previous exposure to chronic METH did not have significant short-term effects on CPP response but led to a decreased CPP response in 10-month-old females. Previous exposure to METH induced some short-term changes to biochemical markers measured in a brain region and sex-dependent manner, while long-term changes were only observed with GFAP and KDM5C.In conclusion, our data suggest sex- and post-exposure duration-dependent outcomes and warrant further exploration of the long-term neurobehavioral consequences of psychostimulant use in both sexes.
Collapse
Affiliation(s)
- Delaney L Davis
- Department of Pharmacology & Neuroscience, UNT HSC, Fort Worth, Texas, USA
| | - Daniel B Metzger
- Department of Pharmacology & Neuroscience, UNT HSC, Fort Worth, Texas, USA
| | - Philip H Vann
- Department of Pharmacology & Neuroscience, UNT HSC, Fort Worth, Texas, USA
| | - Jessica M Wong
- Department of Pharmacology & Neuroscience, UNT HSC, Fort Worth, Texas, USA
| | - Ritu A Shetty
- Department of Pharmacology & Neuroscience, UNT HSC, Fort Worth, Texas, USA
| | - Michael J Forster
- Department of Pharmacology & Neuroscience, UNT HSC, Fort Worth, Texas, USA
| | - Nathalie Sumien
- Department of Pharmacology & Neuroscience, UNT HSC, Fort Worth, Texas, USA.
| |
Collapse
|
19
|
De la Luz-Cuellar YE, Coffeen U, Mercado F, Granados-Soto V. Spinal dopaminergic D1-and D2-like receptors have a sex-dependent effect in an experimental model of fibromyalgia. Eur J Pharmacol 2023; 948:175696. [PMID: 37003519 DOI: 10.1016/j.ejphar.2023.175696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/22/2023] [Accepted: 03/24/2023] [Indexed: 04/01/2023]
Abstract
There is evidence about the importance of sex in pain. The purpose of this study was to investigate the effect of sex in the antiallodynic activity of spinal dopamine D1-and D2-like receptors in a model of fibromyalgia-type pain in rats. Reserpine induced the same extent of tactile allodynia in female and male rats. Intrathecal injection of SCH- 23390 (3-30 nmol, D1-like receptor antagonist), pramipexole (0.15-15 nmol) or quinpirole (1-10 nmol D2-like receptor agonists) increased withdrawal threshold in reserpine-treated female rats. Those drugs induced a greater antiallodynic effect in female rats. Sex-difference was also observed in a nerve injury model. Ovariectomy abated the antiallodynic effect of SCH- 23390 (30 nmol) in reserpine-treated rats, while systemic reconstitution of 17β-estradiol levels or intrathecal injection estrogen receptor-α agonist protopanaxatriol in ovariectomized reserpine-treated females restored the antiallodynic effect of SCH- 23390. Intrathecal administration of ICI-182,780 (estrogen receptor-α/β antagonist) or methyl-piperidino-pyrazole hydrate (estrogen receptor-α antagonist) abated 17β-estradiol-restored antiallodynic effect of SCH- 23390 in rats. In contrast, ovariectomy slightly reduced the effect of pramipexole (15 nmol) or quinpirole (10 nmol) in reserpine-treated rats, whereas systemic reconstitution of 17β-estradiol levels did not modify the antiallodynic effect of both drugs. Combination 17β-estradiol/progesterone, but not 17β-estradiol nor progesterone alone, restored the antiallodynic effect of pramipexole and quinpirole in the rats. Mifepristone (progesterone receptor antagonist) abated 17β-estradiol + progesterone restoration of antiallodynic effect of pramipexole and quinpirole. These data suggest that the antiallodynic effect of dopamine D1-and D2-like receptors in fibromyalgia-type pain depends on spinal 17β-estradiol/estrogen receptor-α and progesterone receptors, respectively.
Collapse
|
20
|
Miller CK, Krentzel AA, Meitzen J. ERα Stimulation Rapidly Modulates Excitatory Synapse Properties in Female Rat Nucleus Accumbens Core. Neuroendocrinology 2023; 113:1140-1153. [PMID: 36746131 PMCID: PMC10623399 DOI: 10.1159/000529571] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/30/2023] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The nucleus accumbens core (NAcc) is a sexually differentiated brain region that is modulated by steroid hormones such as 17β-estradiol (estradiol), with consequential impacts on relevant motivated behaviors and disorders such as addiction, anxiety, and depression. NAcc estradiol levels naturally fluctuate, including during the estrous cycle in adult female rats, which is analogous to the menstrual cycle in adult humans. Across the estrous cycle, excitatory synapse properties of medium spiny neurons rapidly change, as indicated by analysis of miniature excitatory postsynaptic currents (mEPSCs). mEPSC frequency decreases during estrous cycle phases associated with high estradiol levels. This decrease in mEPSC frequency is mimicked by acute topical exposure to estradiol. The identity of the estrogen receptor (ER) underlying this estradiol action is unknown. Adult rat NAcc expresses three ERs, all extranuclear: membrane ERα, membrane ERβ, and GPER1. METHODS In this brief report, we take a first step toward addressing this challenge by testing whether activation of ERs via acute topical agonist application is sufficient for inducing changes in mEPSC properties recorded via whole-cell patch clamp. RESULTS An agonist of ERα induced large decreases in mEPSC frequency, while agonists of ERβ and GPER1 did not robustly modulate mEPSC properties. CONCLUSIONS These data provide evidence that activation of ERα is sufficient for inducing changes in mEPSC frequency and is a likely candidate underlying the estradiol-induced changes observed during the estrous cycle. Overall, these findings extend our understanding of the neuroendocrinology of the NAcc and implicate ERα as a primary target for future studies.
Collapse
Affiliation(s)
- Christiana K. Miller
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Amanda A. Krentzel
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
21
|
Sauton P, Jeanblanc J, Benzerouk F, Gierski F, Naassila M. Sex-specific decision-making impairments and striatal dopaminergic changes after binge drinking history in rats. Front Pharmacol 2023; 14:1076465. [PMID: 36726581 PMCID: PMC9885167 DOI: 10.3389/fphar.2023.1076465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Binge drinking (BD) is a harmful behavior for health and is a predictive factor for the development of alcohol addiction. Weak decision-making (DM) capacities could play a role in the vulnerability to BD which in turn would lead to DM impairments, thus perpetuating BD. Longitudinal preclinical studies are however lacking and necessary to understand this complex relationship. Both DM and BD are influenced by sex and involve dopamine release in the core of the nucleus accumbens, a central mechanism regulated by dopamine D2/3 autoreceptors. In this context, we used an operant self-administration procedure of BD in male and female rats, and longitudinally assessed DM capacity, memory and anxiety-like behavior. To better understand the mechanisms potentially involved in the relationship between DM and BD, ex vivo dopamine transmission was assessed short term after the end of the binge exposure in the core of the nucleus accumbens (NAc) using the fast-scan cyclic voltammetry (FSCV) technique and the D2/3 agonist quinpirole. We found important basal sex differences in DM, with female rats showing better performances at baseline. Choice processes were impaired exclusively in males after BD history, associated with a decrease in impulse control in both sexes, while memory and anxiety-like behavior were not affected. Our neurobiological results demonstrate that BD did not affect basal dopamine signaling in the NAc core, regardless of the sex, but reveal changes in the sensitivity to the inhibitory effects of quinpirole in females. DM impairments were neither associated with changes in basal dopamine signaling nor pre-synaptic D2 activity. Overall, our findings show that BD affects both DM processes and dopamine transmission in the core of the NAc in a sex-related manner, further suggesting that these effects may play a role in the vicious cycle leading to BD perpetuation and the early onset of AUD. Our results may inform novel strategies for therapeutic and prevention interventions.
Collapse
Affiliation(s)
- Pierre Sauton
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France
| | - Jerome Jeanblanc
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France
| | - Farid Benzerouk
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France,Université de Reims Champagne-Ardenne, Laboratoire Cognition, Santé, Société (C2S, EA6291), Reims, France
| | - Fabien Gierski
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France,Université de Reims Champagne-Ardenne, Laboratoire Cognition, Santé, Société (C2S, EA6291), Reims, France
| | - Mickael Naassila
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France,*Correspondence: Mickael Naassila,
| |
Collapse
|
22
|
Stewart A, Mayer FP, Gowrishankar R, Davis GL, Areal LB, Gresch PJ, Katamish RM, Peart R, Stilley SE, Spiess K, Rabil MJ, Diljohn FA, Wiggins AE, Vaughan RA, Hahn MK, Blakely RD. Behaviorally penetrant, anomalous dopamine efflux exposes sex and circuit dependent regulation of dopamine transporters. Mol Psychiatry 2022; 27:4869-4880. [PMID: 36117213 DOI: 10.1038/s41380-022-01773-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 01/19/2023]
Abstract
Virtually all neuropsychiatric disorders display sex differences in prevalence, age of onset, and/or clinical symptomology. Although altered dopamine (DA) signaling is a feature of many of these disorders, sex-dependent mechanisms uniquely responsive to DA that drive sex-dependent behaviors remain unelucidated. Previously, we established that anomalous DA efflux (ADE) is a prominent feature of the DA transporter (DAT) variant Val559, a coding substitution identified in two male-biased disorders: attention-deficit/hyperactivity disorder and autism spectrum disorder. In vivo, Val559 ADE induces activation of nigrostriatal D2-type DA autoreceptors (D2ARs) that magnifies inappropriate, nonvesicular DA release by elevating phosphorylation and surface trafficking of ADE-prone DAT proteins. Here we demonstrate that DAT Val559 mice exhibit sex-dependent alterations in psychostimulant responses, social behavior, and cognitive performance. In a search for underlying mechanisms, we discovered that the ability of ADE to elicit D2AR regulation of DAT is both sex and circuit-dependent, with dorsal striatum D2AR/DAT coupling evident only in males, whereas D2AR/DAT coupling in the ventral striatum is exclusive to females. Moreover, systemic administration of the D2R antagonist sulpiride, which precludes ADE-driven DAT trafficking, can normalize DAT Val559 behavioral changes unique to each sex and without effects on the opposite sex or wildtype mice. Our studies support the sex- and circuit dependent capacity of D2ARs to regulate DAT as a critical determinant of the sex-biased effects of perturbed DA signaling in neurobehavioral disorders. Moreover, our work provides a cogent example of how a shared biological insult drives alternative physiological and behavioral trajectories as opposed to resilience.
Collapse
Affiliation(s)
- Adele Stewart
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Felix P Mayer
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | | | - Gwynne L Davis
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Lorena B Areal
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Paul J Gresch
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Rania M Katamish
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Rodeania Peart
- Wilkes Honors College, Florida Atlantic University, Jupiter, FL, USA
| | - Samantha E Stilley
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Keeley Spiess
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Maximilian J Rabil
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | | | - Angelica E Wiggins
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Maureen K Hahn
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA. .,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA.
| |
Collapse
|
23
|
Mayberry HL, Bavley CC, Karbalaei R, Peterson DR, Bongiovanni AR, Ellis AS, Downey SH, Toussaint AB, Wimmer ME. Transcriptomics in the nucleus accumbens shell reveal sex- and reinforcer-specific signatures associated with morphine and sucrose craving. Neuropsychopharmacology 2022; 47:1764-1775. [PMID: 35190706 PMCID: PMC9372067 DOI: 10.1038/s41386-022-01289-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/16/2022]
Abstract
Incubation of craving is a well-documented phenomenon referring to the intensification of drug craving over extended abstinence. The neural adaptations that occur during forced abstinence following chronic drug taking have been a topic of intense study. However, little is known about the transcriptomic changes occurring throughout this window of time. To define gene expression changes associated with morphine consumption and extended abstinence, male and female rats underwent 10 days of morphine self-administration. Separate drug-naive rats self-administered sucrose in order to compare opioid-induced changes from those associated with natural, non-drug rewards. After one or 30 days of forced abstinence, rats were tested for craving, or nucleus accumbens shell tissue was dissected for RNA sequencing. Morphine consumption was predictive of drug seeking after extended (30 days) but not brief (1 day) abstinence in both sexes. Extended abstinence was also associated with robust sex- and reinforcer-specific changes in gene expression, suggesting sex differences underlying incubation of morphine and sucrose seeking respectively. Importantly, these changes in gene expression occurred without re-exposure to drug-paired cues, indicating that chronic morphine causes long-lasting changes in gene expression that prime the system for increased craving. These findings lay the groundwork for identifying specific therapeutic targets for curbing opioid craving without impacting the natural reward system in males and females.
Collapse
Affiliation(s)
- Hannah L Mayberry
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Charlotte C Bavley
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Reza Karbalaei
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Drew R Peterson
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Angela R Bongiovanni
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Alexandra S Ellis
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Sara H Downey
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Andre B Toussaint
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Krentzel AA, Proaño SB, Dorris DM, Setzer B, Meitzen J. The estrous cycle and 17β-estradiol modulate the electrophysiological properties of rat nucleus accumbens core medium spiny neurons. J Neuroendocrinol 2022; 34:e13122. [PMID: 35365910 PMCID: PMC9250601 DOI: 10.1111/jne.13122] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 12/03/2022]
Abstract
The nucleus accumbens core is a key nexus within the mammalian brain for integrating the premotor and limbic systems and regulating important cognitive functions such as motivated behaviors. Nucleus accumbens core functions show sex differences and are sensitive to the presence of hormones such as 17β-estradiol (estradiol) in normal and pathological contexts. The primary neuron type of the nucleus accumbens core, the medium spiny neuron (MSN), exhibits sex differences in both intrinsic excitability and glutamatergic excitatory synapse electrophysiological properties. Here, we provide a review of recent literature showing how estradiol modulates rat nucleus accumbens core MSN electrophysiology within the context of the estrous cycle. We review the changes in MSN electrophysiological properties across the estrous cycle and how these changes can be mimicked in response to exogenous estradiol exposure. We discuss in detail recent findings regarding how acute estradiol exposure rapidly modulates excitatory synapse properties in nucleus accumbens core but not caudate-putamen MSNs, which mirror the natural changes seen across estrous cycle phases. These recent insights demonstrate the strong impact of sex-specific estradiol action upon nucleus accumbens core neuron electrophysiology.
Collapse
Affiliation(s)
- Amanda A. Krentzel
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
| | - Stephanie B. Proaño
- Neurobiology LaboratoryNational Institute of Environmental Health Sciences, NIHResearch Triangle ParkNCUSA
| | - David M. Dorris
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
| | - Beverly Setzer
- Graduate Program for Neuroscience and Department of Biomedical EngineeringBoston UniversityBostonMAUSA
| | - John Meitzen
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNCUSA
- Center for Human Health and the EnvironmentNorth Carolina State UniversityRaleighNCUSA
| |
Collapse
|
25
|
Logan CN, Rojas G, Wilkinson CS, Polo Escorcia AK, Reichel CM, Peris J, Knackstedt LA. Systemic oxytocin increases glutamate efflux in the nucleus accumbens core of cocaine-experienced male and female rats but only increases dopamine efflux in males. Behav Brain Res 2022; 417:113590. [PMID: 34551348 DOI: 10.1016/j.bbr.2021.113590] [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: 06/11/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/10/2023]
Abstract
Oxytocin attenuates cocaine-seeking when administered both systemically and directly into the nucleus accumbens core. This effect is blocked by intra-accumbens antagonism of mGlu2/3 and, together with our finding that intra-accumbens oxytocin increases glutamate concentrations in this brain region, indicates that pre-synaptic regulation of glutamate release by oxytocin influences cocaine relapse. However, mGlu2/3 receptors also regulate dopamine release in the nucleus accumbens. Here we aimed to determine whether systemic oxytocin increases glutamate and dopamine concentrations in the nucleus accumbens core of cocaine-experienced and cocaine-naïve male and female rats. A subset of rats self-administered cocaine (0.5 mg/kg/infusion) and then underwent extinction training for 2-3 weeks. Rats were implanted with microdialysis probes in the accumbens core and samples were collected for a baseline period, and following saline (1 mL/kg), and oxytocin (1 mg/kg, IP) injections. Locomotion was assessed during microdialysis. In cocaine-experienced rats, oxytocin increased glutamate concentrations in the accumbens core to the same extent in males and females but only increased dopamine concentrations in male rats. Oxytocin did not alter glutamate levels in cocaine-naïve rats. Oxytocin did not produce sedation. These results extend previous findings that systemic oxytocin increases nucleus accumbens dopamine in a sex-specific manner in cocaine-experienced rats. These data are the first to find that systemic oxytocin increases nucleus accumbens glutamate after cocaine experience, providing a mechanism of action by which oxytocin attenuates the reinstatement of cocaine seeking in both male and female rats.
Collapse
Affiliation(s)
- C N Logan
- Psychology Department, University of Florida, Gainesville, FL, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA.
| | - G Rojas
- Psychology Department, University of Florida, Gainesville, FL, USA
| | - C S Wilkinson
- Psychology Department, University of Florida, Gainesville, FL, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| | | | - C M Reichel
- Neuroscience Dept., Medical University of South Carolina, Charleston SC, USA
| | - J Peris
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA; Pharmacodynamics Department, University of Florida, Gainesville, FL, USA
| | - L A Knackstedt
- Psychology Department, University of Florida, Gainesville, FL, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| |
Collapse
|
26
|
Sex Differences in Dopamine Receptors and Relevance to Neuropsychiatric Disorders. Brain Sci 2021; 11:brainsci11091199. [PMID: 34573220 PMCID: PMC8469878 DOI: 10.3390/brainsci11091199] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/03/2021] [Accepted: 09/09/2021] [Indexed: 02/06/2023] Open
Abstract
Dopamine is an important neurotransmitter that plays a key role in neuropsychiatric illness. Sex differences in dopaminergic signaling have been acknowledged for decades and have been linked to sex-specific heterogeneity in both dopamine-related behaviours as well as in various neuropsychiatric disorders. However, the overall number of studies that have evaluated sex differences in dopamine signaling, both in health and in these disorders, is low. This review will bring together what is known regarding sex differences in innate dopamine receptor expression and function, as well as highlight the known sex-specific roles of dopamine in addiction, depression, anxiety, schizophrenia, and attention deficit hyperactivity disorder. Due to differences in prognosis, diagnosis, and symptomatology between male and female subjects in disorders that involve dopamine signaling, or in responses that utilize pharmacological interventions that target dopamine receptors, understanding the fundamental sex differences in dopamine receptors is of vital importance for the personalization of therapeutic treatment strategies.
Collapse
|
27
|
Individual differences in dopamine uptake in the dorsomedial striatum prior to cocaine exposure predict motivation for cocaine in male rats. Neuropsychopharmacology 2021; 46:1757-1767. [PMID: 33953341 PMCID: PMC8357974 DOI: 10.1038/s41386-021-01009-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/27/2021] [Accepted: 03/31/2021] [Indexed: 11/08/2022]
Abstract
A major theme of addiction research has focused on the neural substrates of individual differences in the risk for addiction; however, little is known about how vulnerable populations differ from those that are relatively protected. Here, we prospectively measured dopamine (DA) neurotransmission prior to cocaine exposure to predict the onset and course of cocaine use. Using in vivo voltammetry, we first generated baseline profiles of DA release and uptake in the dorsomedial striatum (DMS) and nucleus accumbens of drug-naïve male rats prior to exposing them to cocaine using conditioned place preference (CPP) or operant self-administration. We found that the innate rate of DA uptake in the DMS strongly predicted motivation for cocaine and drug-primed reinstatement, but not CPP, responding when "price" was low, or extinction. We then assessed the impact of baseline variations in DA uptake on cocaine potency in the DMS using ex vivo voltammetry in naïve rats and in rats with DA transporter (DAT) knockdown. DA uptake in the DMS of naïve rats predicted the neurochemical response to cocaine, such that rats with innately faster rates of DA uptake demonstrated higher cocaine potency at the DAT and rats with DAT knockdown displayed reduced potency compared to controls. Together, these data demonstrate that inherent variability in DA uptake in the DMS predicts the behavioral response to cocaine, potentially by altering the apparent potency of cocaine.
Collapse
|
28
|
Mietlicki-Baase EG, Santollo J, Daniels D. Fluid intake, what's dopamine got to do with it? Physiol Behav 2021; 236:113418. [PMID: 33838203 DOI: 10.1016/j.physbeh.2021.113418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/12/2021] [Accepted: 04/05/2021] [Indexed: 12/27/2022]
Abstract
Maintaining fluid balance is critical for life. The central components that control fluid intake are only partly understood. This contribution to the collection of papers highlighting work by members of the Society for the Study of Ingestive Behavior focuses on the role that dopamine has on fluid intake and describes the roles that various bioregulators can have on thirst and sodium appetite by influencing dopamine systems in the brain. The goal of the review is to highlight areas in need of more research and to propose a framework to guide that research. We hope that this framework will inspire researchers in the field to investigate these interesting questions in order to form a more complete understanding of how fluid intake is controlled.
Collapse
Affiliation(s)
- Elizabeth G Mietlicki-Baase
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, United States; Center for Ingestive Behavior Research, University at Buffalo, State University of New York, Buffalo, NY 14260, United States
| | - Jessica Santollo
- Department of Biology, University of Kentucky, Lexington, KY 40506, United States
| | - Derek Daniels
- Center for Ingestive Behavior Research, University at Buffalo, State University of New York, Buffalo, NY 14260, United States; Department of Psychology, University at Buffalo, State University of New York, Buffalo, NY 14260, United States
| |
Collapse
|
29
|
Contribution of growth hormone secretagogue receptor (GHSR) signaling in the ventral tegmental area (VTA) to the regulation of social motivation in male mice. Transl Psychiatry 2021; 11:230. [PMID: 33879778 PMCID: PMC8058340 DOI: 10.1038/s41398-021-01350-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/19/2021] [Accepted: 03/30/2021] [Indexed: 01/22/2023] Open
Abstract
Most psychiatric disorders are characterized by deficits in the ability to interact socially with others. Ghrelin, a hormone normally associated with the regulation of glucose utilization and appetite, is also implicated in the modulation of motivated behaviors including those associated with food and sex rewards. Here we hypothesized that deficits in ghrelin receptor (growth hormone secretagogue receptor; GHSR) signaling are also associated with deficits in social motivation in male mice. To test this hypothesis, we compared social motivation in male mice lacking GHSR or mice treated with the GHSR antagonist JMV2959 with that of WT or vehicle-treated mice. GHSR signaling in dopamine cells of the ventral tegmental area (VTA) has been implicated in the control of sexual behavior, thus we further hypothesized that GHSR signaling in the VTA is important for social motivation. Thus, we conducted studies where we delivered JMV2959 to block GHSR in the VTA of mice, and studies where we rescued the expression of GHSR in the VTA of GHSR knockout (KO) mice. Mice lacking GHSR or injected with JMV2959 peripherally for 3 consecutive days displayed lower social motivation as reflected by a longer latency to approach a novel conspecific and shorter interaction time compared to WT or vehicle-treated controls. Furthermore, intra-VTA infusion of JMV2959 resulted in longer latencies to approach a novel conspecific, whereas GHSR KO mice with partial rescue of the GHSR showed decreased latencies to begin a novel social interaction. Together, these data suggest that GHSR in the VTA facilitate social approach in male mice, and GHSR-signaling deficits within the VTA result in reduced motivation to interact socially.
Collapse
|
30
|
Differential Impact of Inhibitory G-Protein Signaling Pathways in Ventral Tegmental Area Dopamine Neurons on Behavioral Sensitivity to Cocaine and Morphine. eNeuro 2021; 8:ENEURO.0081-21.2021. [PMID: 33707203 PMCID: PMC8114902 DOI: 10.1523/eneuro.0081-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/21/2022] Open
Abstract
Drugs of abuse engage overlapping but distinct molecular and cellular mechanisms to enhance dopamine (DA) signaling in the mesocorticolimbic circuitry. DA neurons of the ventral tegmental area (VTA) are key substrates of drugs of abuse and have been implicated in addiction-related behaviors. Enhanced VTA DA neurotransmission evoked by drugs of abuse can engage inhibitory G-protein-dependent feedback pathways, mediated by GABAB receptors (GABABRs) and D2 DA receptors (D2Rs). Chemogenetic inhibition of VTA DA neurons potently suppressed baseline motor activity, as well as the motor-stimulatory effect of cocaine and morphine, confirming the critical influence of VTA DA neurons and inhibitory G-protein signaling in these neurons on this addiction-related behavior. To resolve the relative influence of GABABR-dependent and D2R-dependent signaling pathways in VTA DA neurons on behavioral sensitivity to drugs of abuse, we developed a neuron-specific viral CRISPR/Cas9 approach to ablate D2R and GABABR in VTA DA neurons. Ablation of GABABR or D2R did not impact baseline physiological properties or excitability of VTA DA neurons, but it did preclude the direct somatodendritic inhibitory influence of GABABR or D2R activation. D2R ablation potentiated the motor-stimulatory effect of cocaine in male and female mice, whereas GABABR ablation selectively potentiated cocaine-induced activity in male subjects only. Neither D2R nor GABABR ablation impacted morphine-induced motor activity. Collectively, our data show that cocaine and morphine differ in the extent to which they engage inhibitory G-protein-dependent feedback pathways in VTA DA neurons and highlight key sex differences that may impact susceptibility to various facets of addiction.
Collapse
|
31
|
Dynamic control of the dopamine transporter in neurotransmission and homeostasis. NPJ Parkinsons Dis 2021; 7:22. [PMID: 33674612 PMCID: PMC7935902 DOI: 10.1038/s41531-021-00161-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/08/2021] [Indexed: 01/31/2023] Open
Abstract
The dopamine transporter (DAT) transports extracellular dopamine into the intracellular space contributing to the regulation of dopamine neurotransmission. A reduction of DAT density is implicated in Parkinson's disease (PD) by neuroimaging; dopamine turnover is dopamine turnover is elevated in early symptomatic PD and in presymptomatic individuals with monogenic mutations causal for parkinsonism. As an integral plasma membrane protein, DAT surface expression is dynamically regulated through endocytic trafficking, enabling flexible control of dopamine signaling in time and space, which in turn critically modulates movement, motivation and learning behavior. Yet the cellular machinery and functional implications of DAT trafficking remain enigmatic. In this review we summarize mechanisms governing DAT trafficking under normal physiological conditions and discuss how PD-linked mutations may disturb DAT homeostasis. We highlight the complexity of DAT trafficking and reveal DAT dysregulation as a common theme in genetic models of parkinsonism.
Collapse
|
32
|
Effect of Chronic Methylphenidate Treatment in a Female Experimental Model of Parkinsonism. Neurotox Res 2021; 39:667-676. [PMID: 33666887 DOI: 10.1007/s12640-021-00347-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/11/2021] [Accepted: 02/28/2021] [Indexed: 10/22/2022]
Abstract
Methylphenidate (MPH) is the most commonly prescribed drug for the treatment of ADHD in males and females. However, a majority of previous studies investigated the effect of MPH in only males, and little is known regarding consequences of female exposure to MPH. This is unfortunate because the few studies that have been conducted indicate that females have a greater sensitivity to MPH. Previous research in male mice has shown that chronic exposure to MPH causes dopaminergic neurons within the nigrostriatal pathway to be more sensitive to the Parkinsonian toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). However, estrogen has been shown to protect dopaminergic neurons from MPTP neurotoxicity. Therefore, in this study, we test the hypothesis that chronic MPH exposure in female mice will render dopaminergic neurons in the nigrostriatal pathway more sensitive to MPTP, and that estrogen may play a protective role. Interestingly, proestrus females exhibited greater sensitivity to MPTP, with significantly reduced dopaminergic neurons in the SN and significant increases in DA quinone production. Chronic MPH exposure contributed to GSH depletion, but surprisingly, it did not increase dopamine quinone levels or dopaminergic cell loss. There were no significant differences in anestrus animals, with the exception of a depletion in GSH seen when animals received chronic high-dose (10 mg/kg) MPH followed by MPTP. Thus, estrogen may actually sensitize neurons to MPTP in this model, and chronic MPH may contribute to GSH depletion within the striatum. This study provides insight into how chronic psychostimulant use may affect males and females differently.
Collapse
|
33
|
Costa KM, Schenkel D, Roeper J. Sex-dependent alterations in behavior, drug responses and dopamine transporter expression in heterozygous DAT-Cre mice. Sci Rep 2021; 11:3334. [PMID: 33558587 PMCID: PMC7870653 DOI: 10.1038/s41598-021-82600-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 01/19/2021] [Indexed: 01/30/2023] Open
Abstract
Heterozygous mice that express Cre-recombinase under the dopamine transporter promoter (DAT-Cre knock in mice, or KI) are widely used for targeting midbrain dopamine neurons, under the assumption that their constitutive physiology is not affected. We report here that these mice display striking sex-dependent behavioral and molecular differences in relation to wildtypes (WT). Male and female KI mice were constitutively hyperactive, and male KI mice showed attenuated hyperlocomotor responses to amphetamine. In contrast, female KIs displayed a marked reduction in locomotion ("calming" effect) in response to the same dose of amphetamine. Furthermore, male and female DAT-Cre KI mice showed opposing differences in reinforcement learning, with females showing faster conditioning and males showing slower extinction. Other behavioral variables, including working memory and novelty preference, were not changed compared to WT. These effects were paralleled by differences in striatal DAT expression that disproportionately affected female KI mice. Our findings reveal clear limitations of the DAT-Cre line that must be considered when using this model.
Collapse
Affiliation(s)
- Kauê Machado Costa
- grid.7839.50000 0004 1936 9721Institute of Neurophysiology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany ,grid.94365.3d0000 0001 2297 5165Present Address: National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224 USA
| | - Daniela Schenkel
- grid.7839.50000 0004 1936 9721Institute of Neurophysiology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Jochen Roeper
- grid.7839.50000 0004 1936 9721Institute of Neurophysiology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
34
|
Zachry JE, Nolan SO, Brady LJ, Kelly SJ, Siciliano CA, Calipari ES. Sex differences in dopamine release regulation in the striatum. Neuropsychopharmacology 2021; 46:491-499. [PMID: 33318634 PMCID: PMC8027008 DOI: 10.1038/s41386-020-00915-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 01/05/2023]
Abstract
The mesolimbic dopamine system-which originates in the ventral tegmental area and projects to the striatum-has been shown to be involved in the expression of sex-specific behavior and is thought to be a critical mediator of many psychiatric diseases. While substantial work has focused on sex differences in the anatomy of dopamine neurons and relative dopamine levels between males and females, an important characteristic of dopamine release from axon terminals in the striatum is that it is rapidly modulated by local regulatory mechanisms independent of somatic activity. These processes can occur via homosynaptic mechanisms-such as presynaptic dopamine autoreceptors and dopamine transporters-as well as heterosynaptic mechanisms, such as retrograde signaling from postsynaptic cholinergic and GABAergic systems, among others. These regulators serve as potential targets for the expression of sex differences in dopamine regulation in both ovarian hormone-dependent and independent fashions. This review describes how sex differences in microcircuit regulatory mechanisms can alter dopamine dynamics between males and females. We then describe what is known about the hormonal mechanisms controlling/regulating these processes. Finally, we highlight the missing gaps in our knowledge of these systems in females. Together, a more comprehensive and mechanistic understanding of how sex differences in dopamine function manifest will be particularly important in developing evidence-based therapeutics that target this system and show efficacy in both sexes.
Collapse
Affiliation(s)
- Jennifer E. Zachry
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
| | - Suzanne O. Nolan
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
| | - Lillian J. Brady
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
| | - Shannon J. Kelly
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
| | - Cody A. Siciliano
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232 USA
| | - Erin S. Calipari
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN 37232 USA
| |
Collapse
|
35
|
Krentzel AA, Willett JA, Johnson AG, Meitzen J. Estrogen receptor alpha, G-protein coupled estrogen receptor 1, and aromatase: Developmental, sex, and region-specific differences across the rat caudate-putamen, nucleus accumbens core and shell. J Comp Neurol 2020; 529:786-801. [PMID: 32632943 DOI: 10.1002/cne.24978] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022]
Abstract
Sex steroid hormones such as 17β-estradiol (estradiol) regulate neuronal function by binding to estrogen receptors (ERs), including ERα and GPER1, and through differential production via the enzyme aromatase. ERs and aromatase are expressed across the nervous system, including in the striatal brain regions. These regions, comprising the nucleus accumbens core, shell, and caudate-putamen, are instrumental for a wide-range of functions and disorders that show sex differences in phenotype and/or incidence. Sex-specific estrogen action is an integral component for generating these sex differences. A distinctive feature of the striatal regions is that in adulthood neurons exclusively express membrane but not nuclear ERs. This long-standing finding dominates models of estrogen action in striatal regions. However, the developmental etiology of ER and aromatase cellular expression in female and male striatum is unknown. This omission in knowledge is important to address, as developmental stage influences cellular estrogenic mechanisms. Thus, ERα, GPER1, and aromatase cellular immunoreactivity was assessed in perinatal, prepubertal, and adult female and male rats. We tested the hypothesis that ERα, GPER1, and aromatase exhibits sex, region, and age-specific differences, including nuclear expression. ERα exhibits nuclear expression in all three striatal regions before adulthood and disappears in a region- and sex-specific time-course. Cellular GPER1 expression decreases during development in a region- but not sex-specific time-course, resulting in extranuclear expression by adulthood. Somatic aromatase expression presents at prepuberty and increases by adulthood in a region- but not sex-specific time-course. These data indicate that developmental period exerts critical sex-specific influences on striatal cellular estrogenic mechanisms.
Collapse
Affiliation(s)
- Amanda A Krentzel
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA.,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, USA
| | - Jaime A Willett
- Department of Neuroscience, Albert Einstein College of Medicine, New York, New York, USA
| | - Ashlyn G Johnson
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia, USA
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA.,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, USA.,Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
36
|
McDougall SA, Apodaca MG, Park GI, Teran A, Baum TJ, Montejano NR. MK801-induced locomotor activity in preweanling and adolescent male and female rats: role of the dopamine and serotonin systems. Psychopharmacology (Berl) 2020; 237:2469-2483. [PMID: 32445054 PMCID: PMC7354898 DOI: 10.1007/s00213-020-05547-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 05/05/2020] [Indexed: 01/01/2023]
Abstract
RATIONALE MK801, like other NMDA receptor open-channel blockers (e.g., ketamine and phencyclidine), increases the locomotor activity of rats and mice. Whether this behavioral effect ultimately relies on monoamine neurotransmission is of dispute. OBJECTIVE The purpose of this study was to determine whether these psychopharmacological effects and underlying neural mechanisms vary according to sex and age. METHODS Across four experiments, male and female preweanling and adolescent rats were pretreated with vehicle, the monoamine-depleting agent reserpine (1 or 5 mg/kg), the dopamine (DA) synthesis inhibitor ∝-methyl-DL-p-tyrosine (AMPT), the serotonin (5-HT) synthesis inhibitor 4-chloro-DL-phenylalanine methyl ester hydrochloride (PCPA), or both AMPT and PCPA. The locomotor activity of preweanling and adolescent rats was then measured after saline or MK801 (0.3 mg/kg) treatment. RESULTS As expected, MK801 increased the locomotor activity of all age groups and both sexes, but the stimulatory effects were significantly less pronounced in male adolescent rats. Preweanling rats and adolescent female rats were more sensitive to the effects of DA and 5-HT synthesis inhibitors, as AMPT and PCPA caused only small reductions in the MK801-induced locomotor activity of male adolescent rats. Co-administration of AMPT+PCPA or high-dose reserpine (5 mg/kg) treatment substantially reduced MK801-induced locomotor activity in both age groups and across both sexes. CONCLUSIONS These results, when combined with other recent studies, show that NMDA receptor open-channel blockers cause pronounced age-dependent behavioral effects that can vary according to sex. The neural changes underlying these sex and age differences appear to involve monoamine neurotransmission.
Collapse
Affiliation(s)
- Sanders A McDougall
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA.
| | - Matthew G Apodaca
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| | - Ginny I Park
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| | - Angie Teran
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| | - Timothy J Baum
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| | - Nazaret R Montejano
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| |
Collapse
|
37
|
Nolan SO, Zachry JE, Johnson AR, Brady LJ, Siciliano CA, Calipari ES. Direct dopamine terminal regulation by local striatal microcircuitry. J Neurochem 2020; 155:475-493. [PMID: 32356315 DOI: 10.1111/jnc.15034] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023]
Abstract
Regulation of axonal dopamine release by local microcircuitry is at the hub of several biological processes that govern the timing and magnitude of signaling events in reward-related brain regions. An important characteristic of dopamine release from axon terminals in the striatum is that it is rapidly modulated by local regulatory mechanisms. These processes can occur via homosynaptic mechanisms-such as presynaptic dopamine autoreceptors and dopamine transporters - as well heterosynaptic mechanisms such as retrograde signaling from postsynaptic cholinergic and dynorphin systems, among others. Additionally, modulation of dopamine release via diffusible messengers, such as nitric oxide and hydrogen peroxide, allows for various metabolic factors to quickly and efficiently regulate dopamine release and subsequent signaling. Here we review how these mechanisms work in concert to influence the timing and magnitude of striatal dopamine signaling, independent of action potential activity at the level of dopaminergic cell bodies in the midbrain, thereby providing a parallel pathway by which dopamine can be modulated. Understanding the complexities of local regulation of dopamine signaling is required for building comprehensive frameworks of how activity throughout the dopamine system is integrated to drive signaling and control behavior.
Collapse
Affiliation(s)
- Suzanne O Nolan
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jennifer E Zachry
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Amy R Johnson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Lillian J Brady
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN TN, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN TN, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.,Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
38
|
Egenrieder L, Mitricheva E, Spanagel R, Noori HR. No basal or drug‐induced sex differences in striatal dopaminergic levels: a cluster and meta‐analysis of rat microdialysis studies. J Neurochem 2019; 152:482-492. [DOI: 10.1111/jnc.14911] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Lisamon Egenrieder
- Institute of Psychopharmacology Central Institute of Mental Health Medical Faculty Mannheim University of Heidelberg Mannheim Germany
| | | | - Rainer Spanagel
- Institute of Psychopharmacology Central Institute of Mental Health Medical Faculty Mannheim University of Heidelberg Mannheim Germany
| | - Hamid R. Noori
- Institute of Psychopharmacology Central Institute of Mental Health Medical Faculty Mannheim University of Heidelberg Mannheim Germany
- Max Planck Institute for Biological Cybernetics Tübingen Germany
| |
Collapse
|
39
|
Schacht JP, Anton RF, McNamara PJ, Im Y, King AC. The dopamine transporter VNTR polymorphism moderates the relationship between acute response to alcohol and future alcohol use disorder symptoms. Addict Biol 2019; 24:1109-1118. [PMID: 30230123 DOI: 10.1111/adb.12676] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 07/23/2018] [Accepted: 07/27/2018] [Indexed: 12/19/2022]
Abstract
Alcohol use disorder (AUD) is a genetically influenced disease with peak onset in young adulthood. Identification of factors that predict whether AUD symptoms will diminish or persist after young adulthood is a critical public health need. King and colleagues previously reported that acute response to alcohol predicted future AUD symptom trajectory. Genes associated with brain dopamine signaling, which underlies alcohol's rewarding effects, might influence this finding. This study analyzed whether variation at a variable number tandem repeat polymorphism in DAT1/SLC6A3, the gene encoding the dopamine transporter, moderated the predictive relationships between acute response to alcohol and future AUD symptoms among participants enrolled in the Chicago Social Drinking Project (first two cohorts). Heavy-drinking young adults (N = 197) completed an alcohol challenge, in which acute response (liking, wanting, stimulation, and sedation) was measured. Alcohol use disorder symptoms were assessed over the following 6 years. DAT1 genotype significantly moderated the interactions between follow-up time and alcohol liking (P = 0.006) and wanting (P = 0.006) in predicting future AUD symptoms. These predictive effects were strongest among participants who carried the DAT1 9-repeat allele, previously associated with enhanced striatal dopamine tone relative to the 10-repeat allele. Exploratory analyses indicated that DAT1 effects on the relationship between alcohol liking and AUD symptoms appeared stronger for females (n = 79) than males (n = 118) (P = 0.0496). These data suggest that heavy-drinking DAT1 9-repeat allele carriers who display high alcohol-induced reward in young adulthood may be predisposed to persistent AUD symptoms and support combining genotypic and phenotypic information to predict future AUD risk.
Collapse
Affiliation(s)
- Joseph P. Schacht
- Department of Psychiatry and Behavioral Sciences; Medical University of South Carolina; Charleston South Carolina USA
| | - Raymond F. Anton
- Department of Psychiatry and Behavioral Sciences; Medical University of South Carolina; Charleston South Carolina USA
| | - Patrick J. McNamara
- Department of Psychiatry and Behavioral Neuroscience; University of Chicago; Chicago Illinois USA
| | - Yeongbin Im
- Department of Psychiatry and Behavioral Sciences; Medical University of South Carolina; Charleston South Carolina USA
| | - Andrea C. King
- Department of Psychiatry and Behavioral Neuroscience; University of Chicago; Chicago Illinois USA
| |
Collapse
|
40
|
Borland JM, Aiani LM, Norvelle A, Grantham KN, O’Laughlin K, Terranova JI, Frantz KJ, Albers HE. Sex-dependent regulation of social reward by oxytocin receptors in the ventral tegmental area. Neuropsychopharmacology 2019; 44:785-792. [PMID: 30467338 PMCID: PMC6372681 DOI: 10.1038/s41386-018-0262-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/22/2018] [Accepted: 10/27/2018] [Indexed: 01/26/2023]
Abstract
Social reward is critical for social relationships, and yet we know little about the characteristics of social interactions that are rewarding or the neural mechanisms underlying that reward. Here, we investigate the sex-dependent role of oxytocin receptors within the ventral tegmental area (VTA) in mediating the magnitude and valence of social reward. Operant and classical conditioning tests were used to measure social reward associated with same-sex social interactions. The effects of oxytocin, selective oxytocin receptor agonists, antagonists, and vehicle injected into the VTA on social reward was determined in male and female Syrian hamsters. The colocalization of FOS and oxytocin in sites that project to the VTA following social interaction was also determined. Females find same-sex social interactions more rewarding than males and activation of oxytocin receptors in the VTA is critical for social reward in females, as well as males. These studies provide support for the hypothesis that there is an inverted U relationship between the duration of social interaction and social reward, mediated by oxytocin; and that in females the dose-response relationship is initiated at lower doses compared with males. Same-sex social interaction is more rewarding in females than in males, and an inverted U relationship mediated by oxytocin may have a critical role in assigning positive and negative valence to social stimuli. Understanding these sex differences in social reward processing may be essential for understanding the sex differences in the prevalence of many psychiatric disorders and the development of gender-specific treatments of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Johnathan M. Borland
- 0000 0004 1936 7400grid.256304.6Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA USA
| | - Lauren M. Aiani
- 0000 0004 1936 7400grid.256304.6Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA USA
| | - Alisa Norvelle
- 0000 0004 1936 7400grid.256304.6Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA USA
| | - Kymberly N. Grantham
- 0000 0004 1936 7400grid.256304.6Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA USA
| | - Kylie O’Laughlin
- 0000 0004 1936 7400grid.256304.6Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA USA
| | - Joseph I. Terranova
- 0000 0004 1936 7400grid.256304.6Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA USA
| | - Kyle J. Frantz
- 0000 0004 1936 7400grid.256304.6Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA USA
| | - H. Elliott Albers
- 0000 0004 1936 7400grid.256304.6Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA USA
| |
Collapse
|
41
|
Andric Petrovic S, Jerotic S, Mihaljevic M, Pavlovic Z, Ristic I, Soldatovic I, Maric NP. Sex differences in facial emotion recognition in health and psychotic disorders. Cogn Neuropsychiatry 2019; 24:108-122. [PMID: 30789053 DOI: 10.1080/13546805.2019.1582411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Previous studies examining sex-differences in facial emotion recognition (FER) in psychosis yielded inconsistent results. Although females are considered to be superior in FER in health, it remains unclear whether the specific sex-difference is present in psychosis. We aimed to examine whether women and men differ in FER ability in health and in psychosis, and to explore potential sex differences in the illness' effects on FER. METHODS Remitted psychotic patients and controls were assessed using the CANTAB Emotion Recognition Task (ERT) examining accuracies/response latencies in identifying basic emotional expressions. General linear model was performed to assess the effects of group, sex and their interactions on ERT performance. RESULTS Healthy females showed FER advantage in comparison to healthy males, while the aforementioned sex-difference was not observed in remitted psychotic patients. Our results also demonstrated the existence of overall FER deficit in psychosis in comparison to healthy controls, as well as the differential illness' effects on the recognition accuracy of facial expression of anger in males and females-suggesting that females with psychotic disorders undergo more profound deterioration of FER ability than their male counterparts. CONCLUSION The assessment of sex-differences in FER and other important features of psychosis is important for better understanding of its neurobiological basis and for the development of targeted treatments for improved functioning.
Collapse
Affiliation(s)
| | - Stefan Jerotic
- a Clinic for Psychiatry, Clinical Center of Serbia , Belgrade , Serbia
| | - Marina Mihaljevic
- a Clinic for Psychiatry, Clinical Center of Serbia , Belgrade , Serbia.,b School of Medicine , University of Belgrade , Belgrade , Serbia
| | - Zorana Pavlovic
- a Clinic for Psychiatry, Clinical Center of Serbia , Belgrade , Serbia.,b School of Medicine , University of Belgrade , Belgrade , Serbia
| | - Ivan Ristic
- b School of Medicine , University of Belgrade , Belgrade , Serbia
| | - Ivan Soldatovic
- b School of Medicine , University of Belgrade , Belgrade , Serbia
| | - Nadja P Maric
- a Clinic for Psychiatry, Clinical Center of Serbia , Belgrade , Serbia.,b School of Medicine , University of Belgrade , Belgrade , Serbia
| |
Collapse
|
42
|
Borland JM, Rilling JK, Frantz KJ, Albers HE. Sex-dependent regulation of social reward by oxytocin: an inverted U hypothesis. Neuropsychopharmacology 2019; 44:97-110. [PMID: 29968846 PMCID: PMC6235847 DOI: 10.1038/s41386-018-0129-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/10/2018] [Accepted: 06/15/2018] [Indexed: 12/17/2022]
Abstract
The rewarding properties of social interactions are essential for the expression of social behavior and the development of adaptive social relationships. Here, we review sex differences in social reward, and more specifically, how oxytocin (OT) acts in the mesolimbic dopamine system (MDS) to mediate the rewarding properties of social interactions in a sex-dependent manner. Evidence from rodents and humans suggests that same-sex social interactions may be more rewarding in females than in males. We propose that there is an inverted U relationship between OT dose, social reward, and neural activity within structures of the MDS in both males and females, and that this dose-response relationship is initiated at lower doses in females than males. As a result, depending on the dose of OT administered, OT could reduce social reward in females, while enhancing it in males. Sex differences in the neural mechanisms regulating social reward may contribute to sex differences in the incidence of a large number of psychiatric and neurodevelopmental disorders. This review addresses the potential significance of a sex-dependent inverted U dose-response function for OT's effects on social reward and in the development of gender-specific therapies for these disorders.
Collapse
Affiliation(s)
- Johnathan M Borland
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - James K Rilling
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
- Anthropology, Emory University, Atlanta, GA, USA
- Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
- Center for Translational and Social Neuroscience, Emory University, Atlanta, GA, USA
| | - Kyle J Frantz
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - H Elliott Albers
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA.
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
43
|
Females are less sensitive than males to the motivational- and dopamine-suppressing effects of kappa opioid receptor activation. Neuropharmacology 2018; 146:231-241. [PMID: 30528327 DOI: 10.1016/j.neuropharm.2018.12.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 11/21/2022]
Abstract
The neuropeptide dynorphin (DYN) activates kappa opioid receptors (KORs) in the brain to produce depressive-like states and decrease motivation. KOR-mediated suppression of dopamine release in the nucleus accumbens (NAc) is considered one underlying mechanism. We previously showed that, regardless of estrous cycle stage, female rats are less sensitive than males to KOR agonist-mediated decreases in motivation to respond for brain stimulation reward, measured with intracranial self-stimulation (ICSS). However, the explicit roles of KORs, circulating gonadal hormones, and their interaction with dopamine signaling in motivated behavior are not known. As such, we measured the effects of the KOR agonist U50,488 on ICSS stimulation thresholds before and after gonadectomy (or sham surgery). We found that ovariectomized females remained less sensitive than sham or castrated males to KOR-mediated decreases in brain stimulation reward, indicating that circulating gonadal hormones do not play a role. We used qRT-PCR to examine whether sex differences in gene expression in limbic brain regions are associated with behavioral sex differences. We found no sex differences in Pdyn or Oprk1 mRNA in the NAc and ventral tegmental area (VTA), but tyrosine hydroxylase (Th) mRNA was significantly higher in female compared to male VTA. To further explore sex-differences in KOR-mediated suppression of dopamine, we used fast scan cyclic voltammetry (FSCV) and demonstrated that U50,488 was less effective in suppressing evoked NAc dopamine release in females compared to males. These data raise the possibility that females are protected from KOR-mediated decreases in motivation by an increased capacity to produce and release dopamine.
Collapse
|
44
|
Marino RA, Levy R. Differential effects of D1 and D2 dopamine agonists on memory, motivation, learning and response time in non-human primates. Eur J Neurosci 2018; 49:199-214. [PMID: 30326151 DOI: 10.1111/ejn.14208] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/18/2018] [Indexed: 11/29/2022]
Abstract
Dopamine (DA) plays a critical role in cognition, motivation and information processing. DA action has been shown to both improve and/or impair cognition across different receptor types, species, subjects and tasks. This complex relationship has been described as an inverted U-shaped function and may be due to the differential effects of DA receptor activation in the striatum and prefrontal cortex. We have investigated the effects of selective DA agonists on cognitive performance in healthy monkeys using a touch screen running tasks from the CAmbridge Neuropsychological Test Automated Battery (CANTAB). One of two DA agonist drugs or placebo was administered prior to each daily CANTAB session: Dihydrexidine hydrochloride (selective D1 agonist, 0.4-0.9 mg/kg), or sumanirole maleate (selective D2 agonist 0.05-0.3 mg/kg). Three CANTAB tasks were tested: (a) "self-ordered sequential search task" which tested spatial working memory, (b) "reversal learning task," which tested association learning, cognitive flexibility and attention and (c) "visually guided reaching task," which tested reaction time and accuracy. At high dosages, the D2 agonist improved spatial working memory performance, while impairing reversal learning and slowing reach response latency. No consistent cognitive effects were observed with the D1 agonist across the dosages tested. A significant decrease in trial completion rate was observed at the higher dosages of both the D1 and D2 agonists which were consistent with decreased motivation. These results are consistent with task-specific effects of a D2 agonist as well as dose specific insensitivities of a D1 agonist on cognitive and motor behaviors in a healthy monkey.
Collapse
Affiliation(s)
- Robert A Marino
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada.,Department of Surgery, Kingston General Hospital, Kingston, Ontario, Canada
| | - Ron Levy
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada.,Department of Surgery, Kingston General Hospital, Kingston, Ontario, Canada
| |
Collapse
|
45
|
CRF modulation of central monoaminergic function: Implications for sex differences in alcohol drinking and anxiety. Alcohol 2018; 72:33-47. [PMID: 30217435 DOI: 10.1016/j.alcohol.2018.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/03/2018] [Accepted: 01/19/2018] [Indexed: 01/06/2023]
Abstract
Decades of research have described the importance of corticotropin-releasing factor (CRF) signaling in alcohol addiction, as well as in commonly co-expressed neuropsychiatric diseases, including anxiety and mood disorders. However, CRF signaling can also acutely regulate binge alcohol consumption, anxiety, and affect in non-dependent animals, possibly via modulation of central monoaminergic signaling. We hypothesize that basal CRF tone is particularly high in animals and humans with an inherent propensity for high anxiety and alcohol consumption, and thus these individuals are at increased risk for the development of alcohol use disorder and comorbid neuropsychiatric diseases. The current review focuses on extrahypothalamic CRF circuits, particularly those stemming from the bed nucleus of the stria terminalis (BNST), found to play a role in basal phenotypes, and examines whether the intrinsic hyperactivity of these circuits is sufficient to escalate the expression of these behaviors and steepen the trajectory of development of disease states. We focus our efforts on describing CRF modulation of biogenic amine neuron populations that have widespread projections to the forebrain to modulate behaviors, including alcohol and drug intake, stress reactivity, and anxiety. Further, we review the known sex differences and estradiol modulation of these neuron populations and CRF signaling at their synapses to address the question of whether females are more susceptible to the development of comorbid addiction and stress-related neuropsychiatric diseases because of hyperactive extrahypothalamic CRF circuits compared to males.
Collapse
|
46
|
Stolf AR, Cupertino RB, Müller D, Sanvicente-Vieira B, Roman T, Vitola ES, Grevet EH, von Diemen L, Kessler FHP, Grassi-Oliveira R, Bau CHD, Rovaris DL, Pechansky F, Schuch JB. Effects of DRD2 splicing-regulatory polymorphism and DRD4 48 bp VNTR on crack cocaine addiction. J Neural Transm (Vienna) 2018; 126:193-199. [PMID: 30367264 DOI: 10.1007/s00702-018-1946-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/17/2018] [Indexed: 11/24/2022]
Abstract
There is evidence that dopamine receptors D2 (DRD2) and D4 (DRD4) polymorphisms may influence substance use disorders (SUD) susceptibility both individually and through their influence in the formation of DRD2-DRD4 heteromers. The dopaminergic role on the vulnerability to addiction appears to be influenced by sex. A cross-sectional study with 307 crack cocaine addicts and 770 controls was conducted. The influence of DRD2 rs2283265 and DRD4 48 bp VNTR in exon 3 variants, as well as their interaction on crack cocaine addiction susceptibility and severity were evaluated in women and men separately. An association between the DRD2 T allele and crack cocaine addiction was found in women. In this same group, interaction analysis demonstrated that the presence of DRD2-T allele and concomitant absence of DRD4-7R allele were associated with risk for crack cocaine addiction. No influence of DRD2 and DRD4 variants was observed in men regarding addiction severity. This study reinforces the role of dopaminergic genes in externalizing behaviors, especially the influence of DRD2-DRD4 interaction on SUD. This is the fourth sample that independently associated the DRD2-DRD4 interaction with SUD itself or related disorders. In addition, our findings point out to a potential difference of dopaminergic neurotransmission across sex influencing addiction susceptibility.
Collapse
Affiliation(s)
- Anderson R Stolf
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Renata B Cupertino
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Diana Müller
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Breno Sanvicente-Vieira
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tatiana Roman
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo S Vitola
- ADHD Outpatient Program, Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Eugenio H Grevet
- ADHD Outpatient Program, Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lisia von Diemen
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Felix H P Kessler
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rodrigo Grassi-Oliveira
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Claiton H D Bau
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,ADHD Outpatient Program, Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Diego L Rovaris
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Flavio Pechansky
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jaqueline B Schuch
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil. .,Laboratory of Immunosenescence, Graduate Program in Biomedical Gerontology, Pontifícia Universidade Católica do Rio Grande do Sul, Av. Ipiranga, 6681, prédio 81, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil.
| |
Collapse
|
47
|
Modeling drug addiction in females: how internal state and environmental context facilitate vulnerability. Curr Opin Behav Sci 2018. [DOI: 10.1016/j.cobeha.2018.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Krolick KN, Zhu Q, Shi H. Effects of Estrogens on Central Nervous System Neurotransmission: Implications for Sex Differences in Mental Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 160:105-171. [PMID: 30470289 PMCID: PMC6737530 DOI: 10.1016/bs.pmbts.2018.07.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nearly one of every five US individuals aged 12 years old or older lives with certain types of mental disorders. Men are more likely to use various types of substances, while women tend to be more susceptible to mood disorders, addiction, and eating disorders, all of which are risks associated with suicidal attempts. Fundamental sex differences exist in multiple aspects of the functions and activities of neurotransmitter-mediated neural circuits in the central nervous system (CNS). Dysregulation of these neural circuits leads to various types of mental disorders. The potential mechanisms of sex differences in the CNS neural circuitry regulating mood, reward, and motivation are only beginning to be understood, although they have been largely attributed to the effects of sex hormones on CNS neurotransmission pathways. Understanding this topic is important for developing prevention and treatment of mental disorders that should be tailored differently for men and women. Studies using animal models have provided important insights into pathogenesis, mechanisms, and new therapeutic approaches of human diseases, but some concerns remain to be addressed. The purpose of this chapter is to integrate human and animal studies involving the effects of the sex hormones, estrogens, on CNS neurotransmission, reward processing, and associated mental disorders. We provide an overview of existing evidence for the physiological, behavioral, cellular, and molecular actions of estrogens in the context of controlling neurotransmission in the CNS circuits regulating mood, reward, and motivation and discuss related pathology that leads to mental disorders.
Collapse
Affiliation(s)
- Kristen N Krolick
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Qi Zhu
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Haifei Shi
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States; Cellular, Molecular and Structural Biology, Miami University, Oxford, OH, United States.
| |
Collapse
|
49
|
Donovan CH, Wong SA, Randolph SH, Stark RA, Gibb RL, Gruber AJ. Sex differences in rat decision-making: The confounding role of extraneous feeder sampling between trials. Behav Brain Res 2018; 342:62-69. [PMID: 29355674 DOI: 10.1016/j.bbr.2018.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/08/2018] [Accepted: 01/16/2018] [Indexed: 01/19/2023]
Abstract
Although male and female rats appear to perform differently in some tasks, a clear picture of sex differences in decision-making has yet to develop. This is in part due to significant variability arising from differences in strains and tasks. The aim of this study was to characterize the effects of sex on specific response elements in a reinforcement learning task so as to help identify potential explanations for this variability. We found that the primary difference between sexes was the propensity to approach feeders out of the task context. This extraneous feeder sampling affects choice on subsequent trials in both sexes by promoting a lose-shift response away from the last feeder sampled. Female rats, however, were more likely to engage in this extraneous feeder sampling, and therefore exhibited a greater rate of this effect. Once trials following extraneous sampling were removed, there were no significant sex differences in any of the tested measures. These data suggest that feeder approach outside of the task context, which is often not recorded, could produce a confound in sex-based differences of reinforcement sensitivity in some tasks.
Collapse
Affiliation(s)
- Clifford H Donovan
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Scott A Wong
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Sienna H Randolph
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Rachel A Stark
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Robbin L Gibb
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Aaron J Gruber
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada.
| |
Collapse
|
50
|
Cao J, Willett JA, Dorris DM, Meitzen J. Sex Differences in Medium Spiny Neuron Excitability and Glutamatergic Synaptic Input: Heterogeneity Across Striatal Regions and Evidence for Estradiol-Dependent Sexual Differentiation. Front Endocrinol (Lausanne) 2018; 9:173. [PMID: 29720962 PMCID: PMC5915472 DOI: 10.3389/fendo.2018.00173] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/03/2018] [Indexed: 12/21/2022] Open
Abstract
Steroid sex hormones and biological sex influence how the brain regulates motivated behavior, reward, and sensorimotor function in both normal and pathological contexts. Investigations into the underlying neural mechanisms have targeted the striatal brain regions, including the caudate-putamen, nucleus accumbens core (AcbC), and shell. These brain regions are of particular interest to neuroendocrinologists given that they express membrane-associated but not nuclear estrogen receptors, and also the well-established role of the sex steroid hormone 17β-estradiol (estradiol) in modulating striatal dopamine systems. Indeed, output neurons of the striatum, the medium spiny neurons (MSNs), exhibit estradiol sensitivity and sex differences in electrophysiological properties. Here, we review sex differences in rat MSN glutamatergic synaptic input and intrinsic excitability across striatal regions, including evidence for estradiol-mediated sexual differentiation in the nucleus AcbC. In prepubertal animals, female MSNs in the caudate-putamen exhibit a greater intrinsic excitability relative to male MSNs, but no sex differences are detected in excitatory synaptic input. Alternatively, female MSNs in the nucleus AcbC exhibit increased excitatory synaptic input relative to male MSNs, but no sex differences in intrinsic excitability were detected. Increased excitatory synaptic input onto female MSNs in the nucleus AcbC is abolished after masculinizing estradiol or testosterone exposure during the neonatal critical period. No sex differences are detected in MSNs in prepubertal nucleus accumbens shell. Thus, despite possessing the same neuron type, striatal regions exhibit heterogeneity in sex differences in MSN electrophysiological properties, which likely contribute to the sex differences observed in striatal function.
Collapse
Affiliation(s)
- Jinyan Cao
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, United States
| | - Jaime A. Willett
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, United States
- Graduate Program in Physiology, North Carolina State University, Raleigh, NC, United States
| | - David M. Dorris
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, United States
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- *Correspondence: John Meitzen,
| |
Collapse
|