1
|
Lewitus VJ, Kim J, Blackwell KT. Sex and estradiol effects in the rodent dorsal striatum. Eur J Neurosci 2024; 60:6962-6986. [PMID: 39573926 DOI: 10.1111/ejn.16607] [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: 07/01/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
17β-Estradiol (E2) is a sex hormone that acts on many brain regions to produce changes in neuronal activity and learning. A key brain region sensitive to E2 is the dorsal striatum (also called caudate-putamen), which controls motor behaviour, goal-directed learning and habit learning. In adult rodents, oestrogen receptors (ERs) in the dorsal striatum are localized to the plasma membrane and include ERα, ERβ and G protein-coupled ER (GPER). E2, either naturally produced or exogenously applied, may influence neuronal excitability, basal synaptic transmission and long-term synaptic potentiation. These effects may be due to direct action on signalling pathways or may be due to changes in dopamine availability. In particular, estradiol influences dopamine release, dopamine receptor expression and dopamine transporter expression. We review the cellular effects that E2 has in the dorsal striatum, distinguishing between exogenously applied E2 and the oestrous cycle, as well as its influence on dorsal striatal-dependent motor and learning behaviour.
Collapse
|
2
|
Van Zandt M, Flanagan D, Pittenger C. Sex differences in the distribution and density of regulatory interneurons in the striatum. Front Cell Neurosci 2024; 18:1415015. [PMID: 39045533 PMCID: PMC11264243 DOI: 10.3389/fncel.2024.1415015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Dysfunction of the cortico-basal circuitry - including its primary input nucleus, the striatum - contributes to neuropsychiatric disorders, such as autism and Tourette Syndrome (TS). These conditions show marked sex differences, occurring more often in males than in females. Regulatory interneurons, such as cholinergic interneurons (CINs) and parvalbumin-expressing GABAergic fast spiking interneurons (FSIs), are implicated in human neuropsychiatric disorders such as TS, and ablation of these interneurons produces relevant behavioral pathology in male mice, but not in females. Here we investigate sex differences in the density and distribution of striatal interneurons. Methods We use stereological quantification of CINs, FSIs, and somatostatin-expressing (SOM) GABAergic interneurons in the dorsal striatum (caudate-putamen) and the ventral striatum (nucleus accumbens) in male and female mice. Results Males have a higher density of CINs than females, especially in the dorsal striatum; females have equal distribution between dorsal and ventral striatum. FSIs showed similar distributions, with a greater dorsal-ventral density gradient in males than in females. SOM interneurons were denser in the ventral than in the dorsal striatum, with no sex differences. Discussion These sex differences in the density and distribution of FSIs and CINs may contribute to sex differences in basal ganglia function, particularly in the context of psychopathology.
Collapse
Affiliation(s)
- Meghan Van Zandt
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Deirdre Flanagan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Christopher Pittenger
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, United States
- Department of Psychology, Yale School of Arts and Sciences, New Haven, CT, United States
- Center for Brain and Mind Health, Yale University School of Medicine, New Haven, CT, United States
- Wu-Tsai Institute, Yale University, New Haven, CT, United States
| |
Collapse
|
3
|
Copenhaver AE, LeGates TA. Sex-Specific Mechanisms Underlie Long-Term Potentiation at Hippocampus→Medium Spiny Neuron Synapses in the Medial Shell of the Nucleus Accumbens. J Neurosci 2024; 44:e0100242024. [PMID: 38806250 PMCID: PMC11223474 DOI: 10.1523/jneurosci.0100-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
Sex differences have complicated our understanding of the neurobiological basis of many behaviors that are key for survival. As such, continued elucidation of the similarities and differences between sexes is necessary to gain insight into brain function and vulnerability. The connection between the hippocampus (Hipp) and nucleus accumbens (NAc) is a crucial site where modulation of neuronal activity mediates reward-related behavior. Our previous work demonstrated that long-term potentiation (LTP) of Hipp→NAc synapses is rewarding, and mice can establish learned associations between LTP of these synapses and the contextual environment in which LTP occurred. Here, we investigated sex differences in the mechanisms underlying Hipp→NAc LTP using whole-cell electrophysiology and pharmacology. We observed similarities in basal synaptic strength between males and females and found that LTP occurs postsynaptically with similar magnitudes in both sexes. However, key sex differences emerged as LTP in males required NMDA receptors (NMDAR), whereas LTP in females utilized an NMDAR-independent mechanism involving L-type voltage-gated Ca2+ channels (VGCCs) and estrogen receptor α (ERα). We also uncovered sex-similar features as LTP in both sexes depended on CaMKII activity and occurred independently of dopamine-1 receptor (D1R) activation. Our results have elucidated sex-specific molecular mechanisms for LTP in an integral pathway that mediates reward-related behaviors, emphasizing the importance of considering sex as a variable in mechanistic studies. Continued characterization of sex-specific mechanisms underlying plasticity will offer novel insight into the neurophysiological basis of behavior, with significant implications for understanding how diverse processes mediate behavior and contribute to vulnerability to developing psychiatric disorders.
Collapse
Affiliation(s)
- Ashley E Copenhaver
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
| | - Tara A LeGates
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| |
Collapse
|
4
|
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
|
5
|
Johnson CS, Chapp AD, Lind EB, Thomas MJ, Mermelstein PG. Sex differences in mouse infralimbic cortex projections to the nucleus accumbens shell. Biol Sex Differ 2023; 14:87. [PMID: 38082417 PMCID: PMC10712109 DOI: 10.1186/s13293-023-00570-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The nucleus accumbens (NAc) is an important region in motivation and reward. Glutamatergic inputs from the infralimbic cortex (ILC) to the shell region of the NAc (NAcSh) have been implicated in driving the motivation to seek reward through repeated action-based behavior. While this has primarily been studied in males, observed sex differences in motivational circuitry and behavior suggest that females may be more sensitive to rewarding stimuli. These differences have been implicated for the observed vulnerability in women to substance use disorders. METHODS We used an optogenetic self-stimulation task in addition to ex vivo electrophysiological recordings of NAcSh neurons in mouse brain slices to investigate potential sex differences in ILC-NAcSh circuitry in reward-seeking behavior. Glutamatergic neurons in the ILC were infected with an AAV delivering DNA encoding for channelrhodopsin. Entering the designated active corner of an open field arena resulted in photostimulation of the ILC terminals in the NAcSh. Self-stimulation occurred during two consecutive days of testing over three consecutive weeks: first for 10 Hz, then 20 Hz, then 30 Hz. Whole-cell recordings of medium spiny neurons in the NAcSh assessed both optogenetically evoked local field potentials and intrinsic excitability. RESULTS Although both sexes learned to seek the active zone, within the first day, females entered the zone more than males, resulting in a greater amount of photostimulation. Increasing the frequency of optogenetic stimulation amplified female reward-seeking behavior. Males were less sensitive to ILC stimulation, with higher frequencies and repeated days required to increase male reward-seeking behavior. Unexpectedly, ex vivo optogenetic local field potentials in the NAcSh were greater in slices from male animals. In contrast, female medium-spiny neurons (MSNs) displayed significantly greater intrinsic neuronal excitability. CONCLUSIONS Taken together, these data indicate that there are sex differences in the motivated behavior driven by glutamate within the ILC-NAcSh circuit. Though glutamatergic signaling was greater in males, heightened intrinsic excitability in females appears to drive this sex difference.
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
| | - Andrew D Chapp
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
| | - Erin B Lind
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
| | - Mark J Thomas
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
| | - Paul G Mermelstein
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA.
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
6
|
Knouse MC, Deutschmann AU, Nenov MN, Wimmer ME, Briand LA. Sex differences in pre- and post-synaptic glutamate signaling in the nucleus accumbens core. Biol Sex Differ 2023; 14:52. [PMID: 37596655 PMCID: PMC10439632 DOI: 10.1186/s13293-023-00537-4] [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: 03/07/2023] [Accepted: 08/08/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Glutamate signaling within the nucleus accumbens underlies motivated behavior and is involved in psychiatric disease. Although behavioral sex differences in these processes are well-established, the neural mechanisms driving these differences are largely unexplored. In these studies, we examine potential sex differences in synaptic plasticity and excitatory transmission within the nucleus accumbens core. Further understanding of baseline sex differences in reward circuitry will shed light on potential mechanisms driving behavioral differences in motivated behavior and psychiatric disease. METHODS Behaviorally naïve adult male and female Long-Evans rats, C57Bl/6J mice, and constitutive PKMζ knockout mice were killed and tissue containing the nucleus accumbens core was collected for ex vivo slice electrophysiology experiments. Electrophysiology recordings examined baseline sex differences in synaptic plasticity and transmission within this region and the potential role of PKMζ in long-term depression. RESULTS Within the nucleus accumbens core, both female mice and rats exhibit higher AMPA/NMDA ratios compared to male animals. Further, female mice have a larger readily releasable pool of glutamate and lower release probability compared to male mice. No significant sex differences were detected in spontaneous excitatory postsynaptic current amplitude or frequency. Finally, the threshold for induction of long-term depression was lower for male animals than females, an effect that appears to be mediated, in part, by PKMζ. CONCLUSIONS We conclude that there are baseline sex differences in synaptic plasticity and excitatory transmission in the nucleus accumbens core. Our data suggest there are sex differences at multiple levels in this region that should be considered in the development of pharmacotherapies to treat psychiatric illnesses such as depression and substance use disorder.
Collapse
Affiliation(s)
- Melissa C Knouse
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Andre U Deutschmann
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Miroslav N Nenov
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Mathieu E Wimmer
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Lisa A Briand
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA.
- Neuroscience Program, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA.
| |
Collapse
|
7
|
Catalfio AM, Fetterly TL, Nieto AM, Robinson TE, Ferrario CR. Cocaine-induced sensitization and glutamate plasticity in the nucleus accumbens core: effects of sex. Biol Sex Differ 2023; 14:41. [PMID: 37355656 PMCID: PMC10290362 DOI: 10.1186/s13293-023-00525-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND The development and persistence of addiction is mediated in part by drug-induced alterations in nucleus accumbens (NAc) function. AMPA-type glutamate receptors (AMPARs) provide the main source of excitatory drive to the NAc and enhancements in transmission of calcium-permeable AMPARs (CP-AMPARs) mediate increased cue-triggered drug-seeking following prolonged withdrawal. Cocaine treatment regimens that result in psychomotor sensitization enhance subsequent drug-seeking and drug-taking behaviors. Furthermore, cocaine-induced locomotor sensitization followed by 14 days of withdrawal results in an increase in glutamatergic synaptic transmission. However, very few studies have examined cocaine-induced alterations in synaptic transmission of females or potential effects of experimenter-administered cocaine on NAc CP-AMPAR-mediated transmission in either sex. METHODS Male and female rats were given repeated systemic cocaine injections to induce psychomotor sensitization (15 mg/kg, i.p. 1 injection/day, 8 days). Controls received repeated saline (1 mL/kg, i.p). After 14-16 days of withdrawal brain slices were prepared and whole-cell patch-clamp approaches in the NAc core were used to measure spontaneous excitatory post-synaptic currents (sEPSC), paired pulse ratio, and CP-AMPAR transmission. Additional female rats from this same cohort were also given a challenge injection of cocaine at withdrawal day 14 to assess the expression of sensitization. RESULTS Repeated cocaine produced psychomotor sensitization in both sexes. In males this was accompanied by an increase in sEPSC frequency, but not amplitude, and there was no effect on the paired pulse ratio. Males treated with cocaine and saline had similar sensitivity to Naspm. In contrast, in females there were no significant differences between cocaine and saline groups on any measure, despite females showing robust psychomotor sensitization both during the induction and expression phase. CONCLUSIONS Overall, these data reveal striking sex differences in cocaine-induced NAc glutamate plasticity that accompany the induction of psychomotor sensitization. This suggests that the neural adaptations that contribute to sensitization vary by sex.
Collapse
Affiliation(s)
| | | | - Allison M. Nieto
- Pharmacology Department, University of Michigan, Ann Arbor, MI USA
- Neuroscience Graduate Program, University of California, Berkeley, CA USA
| | - Terry E. Robinson
- Psychology Department (Biopsychology Area), University of Michigan, Ann Arbor, MI USA
| | - Carrie R. Ferrario
- Pharmacology Department, University of Michigan, Ann Arbor, MI USA
- Psychology Department (Biopsychology Area), University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
8
|
Beeson ALS, Meitzen J. Estrous cycle impacts on dendritic spine plasticity in rat nucleus accumbens core and shell and caudate-putamen. J Comp Neurol 2023; 531:759-774. [PMID: 36756791 PMCID: PMC10994586 DOI: 10.1002/cne.25460] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/22/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
An important factor that can modulate neuron properties is sex-specific hormone fluctuations, including the human menstrual cycle and rat estrous cycle in adult females. Considering the striatal brain regions, the nucleus accumbens (NAc) core, NAc shell, and caudate-putamen (CPu), the estrous cycle has previously been shown to impact relevant behaviors and disorders, neuromodulator action, and medium spiny neuron (MSN) electrophysiology. Whether the estrous cycle impacts MSN dendritic spine attributes has not yet been examined, even though MSN spines and glutamatergic synapse properties are sensitive to exogenously applied estradiol. Thus, we hypothesized that MSN dendritic spine attributes would differ by estrous cycle phase. To test this hypothesis, brains from adult male rats and female rats in diestrus, proestrus AM, proestrus PM, and estrus were processed for Rapid Golgi-Cox staining. MSN dendritic spine density, size, and type were analyzed in the NAc core, NAc shell, and CPu. Overall spine size differed across estrous cycle phases in female NAc core and NAc shell, and spine length differed across estrous cycle phase in NAc shell and CPu. Consistent with previous work, dendritic spine density was increased in the NAc core compared to the NAc shell and CPu, independent of sex and estrous cycle. Spine attributes in all striatal regions did not differ by sex when estrous cycle was disregarded. These results indicate, for the first time, that estrous cycle phase impacts dendritic spine plasticity in striatal regions, providing a neuroanatomical avenue by which sex-specific hormone fluctuations can impact striatal function and disorders.
Collapse
Affiliation(s)
- Anna LS Beeson
- Department of Biological Sciences, NC State University, Raleigh, USA
- Graduate Program in Biology, NC State University, Raleigh, USA
| | - John Meitzen
- Department of Biological Sciences, NC State University, Raleigh, USA
- Comparative Medicine Institute, NC State University, Raleigh, USA
- Center for Human Health and the Environment, NC State University, Raleigh, USA
| |
Collapse
|
9
|
Maher EE, Strzelecki AM, Weafer JJ, Gipson CD. The importance of translationally evaluating steroid hormone contributions to substance use. Front Neuroendocrinol 2023; 69:101059. [PMID: 36758769 PMCID: PMC10182261 DOI: 10.1016/j.yfrne.2023.101059] [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: 08/24/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Clinically, women appear to be more susceptible to certain aspects of substance use disorders (SUDs). The steroid hormones 17β-estradiol (E2) and progesterone (Pg) have been linked to women-specific drug behaviors. Here, we review clinical and preclinical studies investigating how cycling ovarian hormones affect nicotine-, cocaine-, and opioid-related behaviors. We also highlight gaps in the literature regarding how synthetic steroid hormone use may influence drug-related behaviors. In addition, we explore how E2 and Pg are known to interact in brain reward pathways and provide evidence of how these interactions may influence drug-related behaviors. The synthesis of this review demonstrates the critical need to study women-specific factors that may influence aspects of SUDs, which may play important roles in addiction processes in a sex-specific fashion. It is important to understand factors that impact women's health and may be key to moving the field forward toward more efficacious and individualized treatment strategies.
Collapse
Affiliation(s)
- Erin E Maher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Ashley M Strzelecki
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Jessica J Weafer
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
10
|
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
|
11
|
Johnson CS, Mermelstein PG. The interaction of membrane estradiol receptors and metabotropic glutamate receptors in adaptive and maladaptive estradiol-mediated motivated behaviors in females. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:33-91. [PMID: 36868633 DOI: 10.1016/bs.irn.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Estrogen receptors were initially identified as intracellular, ligand-regulated transcription factors that result in genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor α and estrogen receptor β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) can rapidly alter cellular excitability and gene expression, particularly through the phosphorylation of CREB. A principal mechanism of neuronal mER action has been shown to occur through glutamate-independent transactivation of metabotropic glutamate receptors (mGlu), which elicits multiple signaling outcomes. The interaction of mERs with mGlu has been shown to be important in many diverse functions in females, including driving motivated behaviors. Experimental evidence suggests that a large part of estradiol-induced neuroplasticity and motivated behaviors, both adaptive and maladaptive, occurs through estradiol-dependent mER activation of mGlu. Herein we will review signaling through estrogen receptors, both "classical" nuclear receptors and membrane-bound receptors, as well as estradiol signaling through mGlu. We will focus on how the interactions of these receptors and their downstream signaling cascades are involved in driving motivated behaviors in females, discussing a representative adaptive motivated behavior (reproduction) and maladaptive motivated behavior (addiction).
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
12
|
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
|
13
|
Maher EE, Kipp ZA, Leyrer-Jackson JM, Khatri S, Bondy E, Martinez GJ, Beckmann JS, Hinds TD, Bimonte-Nelson HA, Gipson CD. Ovarian Hormones Regulate Nicotine Consumption and Accumbens Glutamatergic Plasticity in Female Rats. eNeuro 2022; 9:ENEURO.0286-21.2022. [PMID: 35697512 PMCID: PMC9239849 DOI: 10.1523/eneuro.0286-21.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 05/26/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022] Open
Abstract
Women report greater cigarette cravings during the menstrual cycle phase with higher circulating levels of 17β-estradiol (E2), which is metabolized to estrone (E1). Both E2 and E1 bind to estrogen receptors (ERs), which have been highly studied in the breast, uterus, and ovary. Recent studies have found that ERs are also located on GABAergic medium spiny neurons (MSNs) within the nucleus accumbens core (NAcore). Glutamatergic plasticity in NAcore MSNs is altered following nicotine use; however, it is unknown whether estrogens impact this neurobiological consequence. To test the effect of estrogen on nicotine use, we ovariectomized (OVX) female rats that then underwent nicotine self-administration acquisition and compared them to ovary-intact (sham) rats. The OVX animals then received either sesame oil (vehicle), E2, or E1+E2 supplementation for 4 or 20 d before nicotine sessions. While both ovary-intact and OVX females readily discriminated levers, OVX females consumed less nicotine than sham females. Further, neither E2 nor E1+E2 increased nicotine consumption back to sham levels following OVX, regardless of the duration of the treatment. OVX also rendered NAcore MSNs in a potentiated state following nicotine self-administration, which was reversed by 4 d of systemic E2 treatment. Finally, we found that E2 and E1+E2 increased ERα mRNA in the NAcore, but nicotine suppressed this regardless of hormone treatment. Together, these results show that estrogens regulate nicotine neurobiology, but additional factors may be required to restore nicotine consumption to ovary-intact levels.
Collapse
Affiliation(s)
- Erin E Maher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536
| | - Zachary A Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536
| | | | - Shailesh Khatri
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536
| | - Emma Bondy
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536
| | - Genesee J Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536
| | - Joshua S Beckmann
- Department of Psychology, University of Kentucky, Lexington, KY, 40506
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, KY, 40536
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, 85287
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536
| |
Collapse
|
14
|
Johnson CS, Micevych PE, Mermelstein PG. Membrane estrogen signaling in female reproduction and motivation. Front Endocrinol (Lausanne) 2022; 13:1009379. [PMID: 36246891 PMCID: PMC9557733 DOI: 10.3389/fendo.2022.1009379] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/13/2022] [Indexed: 01/13/2023] Open
Abstract
Estrogen receptors were initially identified in the uterus, and later throughout the brain and body as intracellular, ligand-regulated transcription factors that affect genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor-α and estrogen receptor-β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) not only rapidly effect cellular excitability, but can and do ultimately affect gene expression, as seen through the phosphorylation of CREB. A principal mechanism of neuronal mER action is through glutamate-independent transactivation of metabotropic glutamate receptors (mGluRs), which elicits multiple signaling outcomes. The interaction of mERs with mGluRs has been shown to be important in many diverse functions in females, including, but not limited to, reproduction and motivation. Here we review membrane-initiated estrogen receptor signaling in females, with a focus on the interactions between these mERs and mGluRs.
Collapse
Affiliation(s)
- Caroline S. Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Caroline S. Johnson,
| | - Paul E Micevych
- Laboratory of Neuroendocrinology, Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Paul G. Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
15
|
Chronic administration of ketamine induces cognitive deterioration by restraining synaptic signaling. Mol Psychiatry 2021; 26:4702-4718. [PMID: 32488127 DOI: 10.1038/s41380-020-0793-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 05/06/2020] [Accepted: 05/18/2020] [Indexed: 12/20/2022]
Abstract
The discovery of the rapid antidepressant effects of ketamine has arguably been the most important advance in depression treatment. Recently, it was reported that repeated long-term ketamine administration is effective in preventing relapse of depression, which may broaden the clinical use of ketamine. However, long-term treatment with ketamine produces cognitive impairments, and the underlying molecular mechanisms for these impairments are largely unknown. Here, we found that chronic in vivo exposure to ketamine for 28 days led to decreased expression of the glutamate receptor subunits GluA1, GluA2, GluN2A, and GluN2B; decreased expression of the synaptic proteins Syn and PSD-95; decreased dendrite spine density; impairments in long-term potentiation (LTP) and synaptic transmission in the hippocampal CA1 area; and deterioration of learning and memory in mice. Furthermore, the reduced glutamate receptor subunit and synaptic protein expression and the LTP deficits were still observed on day 28 after the last injection of ketamine. We found that the expression and phosphorylation of CaMKIIβ, ERK1/2, CREB, and NF-κB were inhibited by ketamine. The reductions in glutamate receptor subunit expression and dendritic spine density and the deficits in LTP, synaptic transmission, and cognition were alleviated by overexpression of CaMKIIβ. Our study indicates that inhibition of CaMKIIβ-ERK1/2-CREB/NF-κB signaling may mediate chronic ketamine use-associated cognitive impairments by restraining synaptic signaling. Hypofunction of the glutamatergic system might be the underlying mechanism accounting for chronic ketamine use-associated cognitive impairments. Our findings may suggest possible strategies to alleviate ketamine use-associated cognitive deficits and broaden the clinical use of ketamine in depression treatment.
Collapse
|
16
|
Knouse MC, Briand LA. Behavioral sex differences in cocaine and opioid use disorders: The role of gonadal hormones. Neurosci Biobehav Rev 2021; 128:358-366. [PMID: 34214512 DOI: 10.1016/j.neubiorev.2021.06.038] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/20/2021] [Accepted: 06/27/2021] [Indexed: 11/20/2022]
Abstract
Females are more vulnerable than males to many aspects of cocaine use disorder. This vulnerability also translates to opioid use disorder, with females exhibiting stronger behavioral responses than males to drugs such as heroin and morphine. While there is evidence for many overlapping neural mechanisms underlying cocaine and opioid abuse, there is also a breadth of evidence indicating divergent effects of the drugs on synaptic plasticity. This makes it unclear whether the behavioral sex differences seen in substance use disorder across different drugs of abuse rely on the same mechanisms. Ovarian hormones have consistently been implicated as drivers of the behavioral sex differences in cocaine taking and seeking. While there are far fewer studies on the role of ovarian hormones in opioid use disorder, the existing data suggest that ovarian hormones may not drive these behavioral effects in the same manner as in cocaine use disorder. This review highlights evidence that behavioral sex differences in substance use disorder might be driven by different mechanisms depending on drug class.
Collapse
Affiliation(s)
| | - Lisa A Briand
- Department of Psychology, Temple University, United States; Neuroscience Program, Temple University, United States.
| |
Collapse
|
17
|
S-Equol mitigates motivational deficits and dysregulation associated with HIV-1. Sci Rep 2021; 11:11870. [PMID: 34088932 PMCID: PMC8178385 DOI: 10.1038/s41598-021-91240-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/17/2021] [Indexed: 02/04/2023] Open
Abstract
Motivational deficits (e.g., apathy) and dysregulation (e.g., addiction) in HIV-1 seropositive individuals, despite treatment with combination antiretroviral therapy, necessitates the development of innovative adjunctive therapeutics. S-Equol (SE), a selective estrogen receptor β agonist, has been implicated as a neuroprotective and/or neurorestorative therapeutic for HIV-1 associated neurocognitive disorders (HAND); its therapeutic utility for motivational alterations, however, has yet to be systematically evaluated. Thus, HIV-1 transgenic (Tg) and control animals were treated with either a daily oral dose of SE (0.2 mg) or vehicle and assessed in a series of tasks to evaluate goal-directed and drug-seeking behavior. First, at the genotypic level, motivational deficits in HIV-1 Tg rats treated with vehicle were characterized by a diminished reinforcing efficacy of, and sensitivity to, sucrose. Motivational dysregulation was evidenced by enhanced drug-seeking for cocaine relative to control animals treated with vehicle. Second, treatment with SE ameliorated both motivational deficits and dysregulation in HIV-1 Tg rats. Following a history of cocaine self-administration, HIV-1 Tg animals treated with vehicle exhibited lower levels of dendritic branching and a shift towards longer dendritic spines with decreased head diameter. Treatment with SE, however, led to long-term enhancements in dendritic spine morphology in HIV-1 Tg animals supporting a potential underlying basis by which SE exerts its therapeutic effects. Taken together, SE restored motivated behavior in the HIV-1 Tg rat, expanding the potential clinical utility of SE to include both neurocognitive and affective alterations.
Collapse
|
18
|
Cao J, Meitzen J. Perinatal activation of ER α and ER β but not GPER-1 masculinizes female rat caudate-putamen medium spiny neuron electrophysiological properties. J Neurophysiol 2021; 125:2322-2338. [PMID: 33978486 DOI: 10.1152/jn.00063.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Exposure to steroid sex hormones such as 17β-estradiol (estradiol) during early life potentially permanently masculinize neuron electrophysiological phenotype. In rodents, one crucial component of this developmental process occurs in males, with estradiol aromatized in the brain from testes-sourced testosterone. However, it is unknown whether most neuron electrophysiological phenotypes are altered by this early masculinization process, including medium spiny neurons (MSNs) of the rat caudate-putamen. MSNs are the predominant and primary output neurons of the caudate-putamen and exhibit increased intrinsic excitability in females compared to males. Here, we hypothesize that since perinatal estradiol exposure occurs in males, then a comparable exposure in females to estradiol or its receptor agonists would be sufficient to induce masculinization. To test this hypothesis, we injected perinatal female rats with estradiol or its receptor agonists and then later assessed MSN electrophysiology. Female and male rats on postnatal day 0 and 1 were systemically injected with either vehicle, estradiol, the estrogen receptor (ER)α agonist PPT, the ERβ agonist DPN, or the G-protein-coupled receptor 1 (GPER-1) agonist G1. On postnatal days 19 ± 2, MSN electrophysiological properties were assessed using whole cell patch clamp recordings. Estradiol exposure abolished increased intrinsic excitability in female compared to male MSNs. Exposure to either an ERα or ERβ agonist masculinized female MSN evoked action potential firing rate properties, whereas exposure to an ERβ agonist masculinized female MSN inward rectification properties. Exposure to ER agonists minimally impacted male MSN electrophysiological properties. These findings indicate that perinatal estradiol exposure masculinizes MSN electrophysiological phenotype via activation of ERα and ERβ.NEW & NOTEWORTHY This study is the first to demonstrate that estradiol and estrogen receptor α and β stimulation during early development sexually differentiates the electrophysiological properties of caudate-putamen medium spiny neurons, the primary output neuron of the striatal regions. Overall, this evidence provides new insight into the neuroendocrine mechanism by which caudate-putamen neuron electrophysiology is sexually differentiated and demonstrates the powerful action of early hormone exposure upon individual neuron electrophysiology.
Collapse
Affiliation(s)
- Jinyan Cao
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina.,Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
19
|
Ma R, Mikhail ME, Culbert KM, Johnson AW, Sisk CL, Klump KL. Ovarian Hormones and Reward Processes in Palatable Food Intake and Binge Eating. Physiology (Bethesda) 2021; 35:69-78. [PMID: 31799907 DOI: 10.1152/physiol.00013.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Ovarian hormones are associated with risk for binge eating in women. Recent animal and human studies suggest that food-related reward processing may be one set of neurobiological factors that contribute to these relationships, but additional studies are needed to confirm and extend findings.
Collapse
Affiliation(s)
- Ruofan Ma
- Department of Psychology, Michigan State University, East Lansing, Michigan
| | - Megan E Mikhail
- Department of Psychology, Michigan State University, East Lansing, Michigan
| | - Kristen M Culbert
- Department of Psychology, University of Nevada-Las Vegas, Las Vegas, Nevada
| | - Alex W Johnson
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Cheryl L Sisk
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Kelly L Klump
- Department of Psychology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
20
|
Huijgens PT, Snoeren EMS, Meisel RL, Mermelstein PG. Effects of gonadectomy and dihydrotestosterone on neuronal plasticity in motivation and reward related brain regions in the male rat. J Neuroendocrinol 2021; 33:e12918. [PMID: 33340384 DOI: 10.1111/jne.12918] [Citation(s) in RCA: 4] [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: 07/02/2020] [Revised: 09/25/2020] [Accepted: 11/02/2020] [Indexed: 12/22/2022]
Abstract
Gonadal hormones affect neuronal morphology to ultimately regulate behaviour. In female rats, oestradiol mediates spine plasticity in hypothalamic and limbic brain structures, contributing to long-lasting effects on motivated behaviour. Parallel effects of androgens in male rats have not been extensively studied. Here, we investigated the effect of both castration and androgen replacement on spine plasticity in the nucleus accumbens shell and core (NAcSh and NAcC), caudate putamen (CPu), medial amygdala (MeA) and medial preoptic nucleus (MPN). Intact and castrated (gonadectomy [GDX]) male rats were treated with dihydrotestosterone (DHT, 1.5 mg) or vehicle (oil) in three experimental groups: intact-oil, GDX-oil and GDX-DHT. Spine density and morphology, measured 24 hours after injection, were determined through three-dimensional reconstruction of confocal z-stacks of DiI-labelled dendritic segments. We found that GDX decreased spine density in the MPN, which was rescued by DHT treatment. DHT also increased spine density in the MeA in GDX animals compared to intact oil-treated animals. By contrast, DHT decreased spine density in the NAcSh compared to GDX males. No effect on spine density was observed in the NAcC or CPu. Spine length and spine head diameter were unaffected by GDX and DHT in the investigated brain regions. In addition, immunohistochemistry revealed that DHT treatment of GDX animals rapidly increased the number of cell bodies in the NAcSh positive for phosphorylated cAMP response-element binding protein, a downstream messenger of the androgen receptor. These findings indicate that androgen signalling plays a role in the regulation of spine plasticity within neurocircuits involved in motivated behaviours.
Collapse
Affiliation(s)
- Patty T Huijgens
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Psychology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Eelke M S Snoeren
- Department of Psychology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Robert L Meisel
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
21
|
Abstract
Gonadal hormones contribute to the sexual differentiation of brain and behavior throughout the lifespan, from initial neural patterning to "activation" of adult circuits. Sexual behavior is an ideal system in which to investigate the mechanisms underlying hormonal activation of neural circuits. Sexual behavior is a hormonally regulated, innate social behavior found across species. Although both sexes seek out and engage in sexual behavior, the specific actions involved in mating are sexually dimorphic. Thus, the neural circuits mediating sexual motivation and behavior in males and females are overlapping yet distinct. Furthermore, sexual behavior is strongly dependent on circulating gonadal hormones in both sexes. There has been significant recent progress on elucidating how gonadal hormones modulate physiological properties within sexual behavior circuits with consequences for behavior. Therefore, in this mini-review we review the neural circuits of male and female sexual motivation and behavior, from initial sensory detection of pheromones to the extended amygdala and on to medial hypothalamic nuclei and reward systems. We also discuss how gonadal hormones impact the physiology and functioning of each node within these circuits. By better understanding the myriad of ways in which gonadal hormones impact sexual behavior circuits, we can gain a richer and more complete appreciation for the neural substrates of complex behavior.
Collapse
Affiliation(s)
- Kimberly J Jennings
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| |
Collapse
|
22
|
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
|
23
|
Gross KS, Mermelstein PG. Estrogen receptor signaling through metabotropic glutamate receptors. VITAMINS AND HORMONES 2020; 114:211-232. [PMID: 32723544 DOI: 10.1016/bs.vh.2020.06.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As the non-nuclear initiated effects of steroid hormone signaling have become more widely accepted, there has been a need to define the novel mechanisms of hormone receptor action that account for these outcomes. One mechanism that has emerged is the coupling of classical estrogen receptors (ERα and ERβ) with metabotropic glutamate receptors (mGluRs) to initiate G protein signaling cascades that ultimately influence neuronal physiology, structure, and behavior. Since its initial discovery in hippocampal neurons, evidence of ER/mGluR associations have been found throughout the nervous system, and the heterogeneity of possible receptor pairings afforded by multiple ER and mGluR subtypes appears to drive diverse molecular outcomes that can impact processes like cognition, motivation, movement, and pain. Recent evidence also suggests that the role of mGluRs in steroid hormone signaling may not be unique to ERs, but rather a conserved mechanism of membrane-initiated hormone receptor action.
Collapse
Affiliation(s)
- Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
24
|
Proaño SB, Meitzen J. Estradiol decreases medium spiny neuron excitability in female rat nucleus accumbens core. J Neurophysiol 2020; 123:2465-2475. [PMID: 32432511 DOI: 10.1152/jn.00210.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The menstrual cycle in humans and its analogous cycle in rodents, the estrous cycle, modulate brain function and behavior. Both cycles are characterized by the cyclical fluctuation of ovarian hormones including estrogens such as estradiol. Estradiol induces cycle- and sex-dependent differences in the phenotype and incidence of many behaviors, including those related to reward and motivation. The nucleus accumbens core (AcbC), a limbic and premotor system nexus region, directly regulates these behaviors. We previously showed that the estrous cycle modulates intrinsic excitability and excitatory synapse properties of medium spiny neurons (MSNs) in the AcbC. The identity of the underlying hormone mechanism is unknown, with estradiol being a prime candidate. The present study tests the hypothesis that estradiol induces estrous cycle-relevant differences in MSN electrophysiology. To accomplish this goal, a time- and dose-dependent estradiol replacement paradigm designed to simulate the rise of circulating estradiol levels across the estrous cycle was employed in ovariectomized adult female rats as well as a vehicle control group. Estradiol replacement decreased MSN excitability by modulating properties such as resting membrane potential, input resistance in both the linear and rectified ranges, and rheobase compared with vehicle-treated females. These differences in MSN excitability mimic those previously described regarding estrous cycle effects on MSN electrophysiology. Excitatory synapse properties were not modulated in response to this estradiol replacement paradigm. These data are the first to demonstrate that an estrous cycle-relevant estradiol exposure modulates MSN electrophysiology, providing evidence of the fundamental neuroendocrine mechanisms regulating the AcbC.NEW & NOTEWORTHY The present study shows, for the first time, that an estrous cycle-relevant estradiol exposure modulates nucleus accumbens neuron excitability. This evidence provides insight into the neuroendocrine mechanisms by which estradiol cyclically alters neuron properties during the estrous cycle. Overall, these data emphasize the significant influence of hormone action in the brain and especially individual neuron physiology.
Collapse
Affiliation(s)
- Stephanie B Proaño
- Graduate Program in Biology, North Carolina State University, Raleigh, North Carolina.,W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina.,Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
| | - John Meitzen
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina.,Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina.,Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
25
|
Giacometti LL, Barker JM. Sex differences in the glutamate system: Implications for addiction. Neurosci Biobehav Rev 2020; 113:157-168. [PMID: 32173404 DOI: 10.1016/j.neubiorev.2020.03.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 01/21/2020] [Accepted: 03/08/2020] [Indexed: 12/21/2022]
Abstract
Clinical and preclinical research have identified sex differences in substance use and addiction-related behaviors. Historically, substance use disorders are more prevalent in men than women, though this gap is closing. Despite this difference, women appear to be more susceptible to the effects of many drugs and progress to substance abuse treatment more quickly than men. While the glutamate system is a key regulator of addiction-related behaviors, much of the work implicating glutamate signaling and glutamatergic circuits has been conducted in men and male rodents. An increasing number of studies have identified sex differences in drug-induced glutamate alterations as well as sex and estrous cycle differences in drug seeking behaviors. This review will describe sex differences in the glutamate system with an emphasis on implications for substance use disorders, highlighting the gaps in our current understanding of how innate and drug-induced alterations in the glutamate system may contribute to sex differences in addiction-related behaviors.
Collapse
Affiliation(s)
- L L Giacometti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States.
| | - J M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States.
| |
Collapse
|
26
|
Miller CK, Krentzel AA, Patisaul HB, Meitzen J. Metabotropic glutamate receptor subtype 5 (mGlu 5) is necessary for estradiol mitigation of light-induced anxiety behavior in female rats. Physiol Behav 2019; 214:112770. [PMID: 31830486 DOI: 10.1016/j.physbeh.2019.112770] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/20/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023]
Abstract
Anxiety-related behaviors are influenced by steroid hormones such as 17β-estradiol and environmental stimuli such as acute stressors. For example, rats exhibit increased anxiety-related behaviors in the presence, but not the absence, of light. In females, estradiol potentially mitigates these effects. Experiments across behavioral paradigms and brain regions indicate that estradiol action can be mediated via activation of metabotropic glutamate receptors, including Group I subtype five (mGlu5). mGlu5 has been implicated in mediating estradiol's effects upon psychostimulant-induced behaviors, dopamine release and neuron phenotype in striatal regions. Whether estradiol activation of mGlu5 modulates anxiety or locomotor behavior in the absence of psychostimulants is unknown. Here we test if mGlu5 is necessary for estradiol mitigation of light-induced acute anxiety and locomotor behaviors. Ovariectomized adult female rats were pre-treated with either the mGlu5 antagonist MPEP or saline before estradiol or oil treatment. Anxiety and locomotor behaviors were assessed in the presence or absence of white light to induce high and low acute anxiety behavior phenotypes, respectively. In the presence of white light, estradiol treatment mitigated light-induced anxiety-related behaviors but not overall locomotor activity. MPEP treatment blocked estradiol effects upon light-induced anxiety-related behaviors but did not affect overall locomotor activity. In the absence of white light, estradiol or MPEP treatment did not influence anxiety-related behaviors or locomotor activity, consistent with a low anxiety phenotype. These novel findings indicate that mGlu5 activation is necessary for estradiol mitigation of anxiety-related behaviors induced by an acute stressor.
Collapse
Affiliation(s)
- Christiana K Miller
- Graduate Program in Biology, North Carolina State University, Raleigh, NC, USA; W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA.
| | - Amanda A Krentzel
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA; Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Heather B Patisaul
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA; Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - John Meitzen
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA; Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
27
|
Krentzel AA, Barrett LR, Meitzen J. Estradiol rapidly modulates excitatory synapse properties in a sex- and region-specific manner in rat nucleus accumbens core and caudate-putamen. J Neurophysiol 2019; 122:1213-1225. [PMID: 31314648 PMCID: PMC6766735 DOI: 10.1152/jn.00264.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/16/2019] [Accepted: 07/16/2019] [Indexed: 12/26/2022] Open
Abstract
Estradiol acutely facilitates sex differences in striatum-dependent behaviors. However, little is understood regarding the underlying mechanism. In striatal regions in adult rodents, estrogen receptors feature exclusively extranuclear expression, suggesting that estradiol rapidly modulates striatal neurons. We tested the hypothesis that estradiol rapidly modulates excitatory synapse properties onto medium spiny neurons (MSNs) of two striatal regions, the nucleus accumbens core and caudate-putamen in adult female and male rats. We predicted there would be sex-specific differences in pre- and postsynaptic locus and sensitivity. We further analyzed whether MSN intrinsic properties are predictive of estrogen sensitivity. Estradiol exhibited sex-specific acute effects in the nucleus accumbens core: miniature excitatory postsynaptic current (mEPSC) frequency robustly decreased in response to estradiol in female MSNs, and mEPSC amplitude moderately increased in response to estradiol in both male and female MSNs. This increase in mEPSC amplitude is associated with MSNs featuring increased intrinsic excitability. No MSN intrinsic electrical property associated with changes in mEPSC frequency. Estradiol did not acutely modulate mEPSC properties in the caudate-putamen of either sex. This is the first demonstration of acute estradiol action on MSN excitatory synapse function. This demonstration of sex and striatal region-specific acute estradiol neuromodulation revises our understanding of sex hormone action on striatal physiology and resulting behaviors.NEW & NOTEWORTHY This study is the first to demonstrate rapid estradiol neuromodulation of glutamatergic signaling on medium spiny neurons (MSNs), the major output neuron of the striatum. These findings emphasize that sex is a significant biological variable both in MSN sensitivity to estradiol and in pre- and postsynaptic mechanisms of glutamatergic signaling. MSNs in different regions exhibit diverse responses to estradiol. Sex- and region-specific estradiol-induced changes to excitatory signaling on MSNs explain sex differences partially underlying striatum-mediated behaviors and diseases.
Collapse
Affiliation(s)
- Amanda A Krentzel
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina
| | - Lily R Barrett
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina
- Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
28
|
Willett JA, Cao J, Johnson A, Patel OH, Dorris DM, Meitzen J. The estrous cycle modulates rat caudate-putamen medium spiny neuron physiology. Eur J Neurosci 2019; 52:2737-2755. [PMID: 31278786 DOI: 10.1111/ejn.14506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 06/25/2019] [Indexed: 12/27/2022]
Abstract
The neuroendocrine environment in which the brain operates is both dynamic and differs by sex. How differences in neuroendocrine state affect neuron properties has been significantly neglected in neuroscience research. Behavioral data across humans and rodents indicate that natural cyclical changes in steroid sex hormone production affect sensorimotor and cognitive behaviors in both normal and pathological contexts. These behaviors are critically mediated by the caudate-putamen. In the caudate-putamen, medium spiny neurons (MSNs) are the predominant and primary output neurons. MSNs express membrane-associated estrogen receptors and demonstrate estrogen sensitivity. However, how the cyclical hormone changes across the estrous cycle may modulate caudate-putamen MSN electrophysiological properties remains unknown. Here, we performed whole-cell patch-clamp recordings on male, diestrus female, proestrus female, and estrus female caudate-putamen MSNs. Action potential, passive membrane, and miniature excitatory post-synaptic current properties were assessed. Numerous MSN electrical properties robustly differed by cycle state, including resting membrane potential, rheobase, action potential threshold, maximum evoked action potential firing rate, and inward rectification. Strikingly, when considered independent of estrous cycle phase, all but one of these properties do not significantly differ from male MSNs. These data indicate that female caudate-putamen MSNs are sensitive to the estrous cycle, and more broadly, the importance of considering neuroendocrine state in studies of neuron physiology.
Collapse
Affiliation(s)
- Jaime A Willett
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA.,Graduate Program in Physiology, North Carolina State University, Raleigh, NC, USA.,Grass Laboratory, Marine Biological Laboratory, Woods Hole, MA, USA
| | - Jinyan Cao
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA
| | - Ashlyn Johnson
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Opal H Patel
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - David M Dorris
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
29
|
Krentzel AA, Meitzen J. Biological Sex, Estradiol and Striatal Medium Spiny Neuron Physiology: A Mini-Review. Front Cell Neurosci 2018; 12:492. [PMID: 30618639 PMCID: PMC6299026 DOI: 10.3389/fncel.2018.00492] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/30/2018] [Indexed: 12/21/2022] Open
Abstract
The caudate-putamen, nucleus accumbens core and shell are important striatal brain regions for premotor, limbic, habit formation, reward, and other critical cognitive functions. Striatal-relevant behaviors such as anxiety, motor coordination, locomotion, and sensitivity to reward, all change with fluctuations of the menstrual cycle in humans and the estrous cycle in rodents. These fluctuations implicate sex steroid hormones, such as 17β-estradiol, as potent neuromodulatory signals for striatal neuron activity. The medium spiny neuron (MSN), the primary neuron subtype of the striatal regions, expresses membrane estrogen receptors and exhibits sex differences both in intrinsic and synaptic electrophysiological properties. In this mini-review, we first describe sex differences in the electrophysiological properties of the MSNs in prepubertal rats. We then discuss specific examples of how the human menstrual and rat estrous cycles induce differences in striatal-relevant behaviors and neural substrate, including how female rat MSN electrophysiology is influenced by the estrous cycle. We then conclude the mini-review by discussing avenues for future investigation, including possible roles of striatal-localized membrane estrogen receptors and estradiol.
Collapse
Affiliation(s)
- Amanda A Krentzel
- 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
| | - 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
| |
Collapse
|
30
|
Gross KS, Moore KM, Meisel RL, Mermelstein PG. mGluR5 Mediates Dihydrotestosterone-Induced Nucleus Accumbens Structural Plasticity, but Not Conditioned Reward. Front Neurosci 2018; 12:855. [PMID: 30515075 PMCID: PMC6255826 DOI: 10.3389/fnins.2018.00855] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
Gonadal hormones play a vital role in driving motivated behavior. They not only modulate responses to naturally rewarding stimuli, but also influence responses to drugs of abuse. A commonality between gonadal hormones and drugs of abuse is that they both impact the neurocircuitry of reward, including the regulation of structural plasticity in the nucleus accumbens (NAc). Previous hormonal studies have focused on the mechanisms and behavioral correlates of estradiol-induced dendritic spine changes in the female NAc. Here we sought to determine the effects of androgens on medium spiny neuron (MSN) spine plasticity in the male NAc. Following treatment with the androgen receptor agonist dihydrotestosterone (DHT), MSNs in castrated male rats exhibited a significant decrease in dendritic spine density. This effect was isolated to the shell subregion of the NAc. The effect of DHT was dependent on mGluR5 activity, and local mGluR5 activation and subsequent endocannabinoid signaling produce an analogous NAc shell spine decrease. Somewhat surprisingly, DHT-induced conditioned place preference remained intact following systemic inhibition of mGluR5. These findings indicate that androgens can utilize mGluR signaling, similar to estrogens, to mediate changes in NAc dendritic structure. In addition, there are notable differences in the direction of spine changes, and site specificity of estrogen and androgen action, suggesting sex differences in the hormonal regulation of motivated behaviors.
Collapse
Affiliation(s)
- Kellie S Gross
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Kelsey M Moore
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Robert L Meisel
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
31
|
Cao J, Dorris DM, Meitzen J. Electrophysiological properties of medium spiny neurons in the nucleus accumbens core of prepubertal male and female Drd1a-tdTomato line 6 BAC transgenic mice. J Neurophysiol 2018; 120:1712-1727. [PMID: 29975170 PMCID: PMC6230806 DOI: 10.1152/jn.00257.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/27/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022] Open
Abstract
The nucleus accumbens core (AcbC) is a striatal brain region essential for integrating motivated behavior and reward processing with premotor function. In humans and rodents, research has identified sex differences and sex steroid hormone sensitivity in AcbC-mediated behaviors, in disorders, and in rats in the electrophysiological properties of the AcbC output neuron type, the medium spiny neuron (MSN). It is unknown whether the sex differences detected in MSN electrophysiological properties extend to mice. Furthermore, MSNs come in distinct subtypes with subtle differences in electrophysiological properties, and it is unknown whether MSN subtype-specific electrophysiology varies by sex. To address these questions, we used male and female Drd1a-tdTomato line 6 bacterial artificial chromosome transgenic mice. We made acute brain slices of the AcbC, and performed whole cell patch-clamp recordings across MSN subtypes to comprehensively assess AcbC MSN subtype electrophysiological properties. We found that ( 1 mice MSNs did not exhibit the sex differences detected in rat MSNs, and 2) electrophysiological properties differed between MSN subtypes in both sexes, including rheobase, resting membrane potential, action potential properties, intrinsic excitability, input resistance in both the linear and rectified ranges, and miniature excitatory postsynaptic current properties. These findings significantly extend previous studies of MSN subtypes performed in males or animals of undetermined sex and indicate that the influence of sex upon AcbC MSN properties varies between rodent species. NEW & NOTEWORTHY This research provides the most comprehensive assessment of medium spiny neuron subtype electrophysiological properties to date in a critical brain region, the nucleus accumbens core. It additionally represents the first evaluation of whether mouse medium spiny neuron subtype electrophysiological properties differ by sex.
Collapse
Affiliation(s)
- Jinyan Cao
- Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
- W. M. Keck Center for Behavioral Biology, North Carolina State University , Raleigh, North Carolina
| | - David M Dorris
- Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
- W. M. Keck Center for Behavioral Biology, North Carolina State University , Raleigh, North Carolina
- Center for Human Health and the Environment, North Carolina State University , Raleigh, North Carolina
- Comparative Medicine Institute, North Carolina State University , Raleigh, North Carolina
| |
Collapse
|
32
|
Tonn Eisinger KR, Gross KS, Head BP, Mermelstein PG. Interactions between estrogen receptors and metabotropic glutamate receptors and their impact on drug addiction in females. Horm Behav 2018; 104:130-137. [PMID: 29505763 PMCID: PMC6131090 DOI: 10.1016/j.yhbeh.2018.03.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 02/07/2023]
Abstract
Contribution to Special Issue on Fast effects of steroids. Estrogen receptors α and β (ERα and ERβ) have a unique relationship with metabotropic glutamate receptors (mGluRs) in the female rodent brain such that estradiol is able to recruit intracellular G-protein signaling cascades to influence neuronal physiology, structure, and ultimately behavior. While this association between ERs and mGluRs exists in many cell types and brain regions, its effects are perhaps most striking in the nucleus accumbens (NAc). This review will discuss the original characterization of ER/mGluR signaling and how estradiol activity in the NAc confers increased sensitivity to drugs of abuse in females through this mechanism.
Collapse
Affiliation(s)
- Katherine R Tonn Eisinger
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Brian P Head
- Department of Anesthesiology, University of California-San Diego, La Jolla, CA 92093, USA
| | - Paul G Mermelstein
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
33
|
Proaño SB, Morris HJ, Kunz LM, Dorris DM, Meitzen J. Estrous cycle-induced sex differences in medium spiny neuron excitatory synaptic transmission and intrinsic excitability in adult rat nucleus accumbens core. J Neurophysiol 2018; 120:1356-1373. [PMID: 29947588 DOI: 10.1152/jn.00263.2018] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Naturally occurring hormone cycles in adult female humans and rodents create a dynamic neuroendocrine environment. These cycles include the menstrual cycle in humans and its counterpart in rodents, the estrous cycle. These hormone fluctuations induce sex differences in the phenotypes of many behaviors, including those related to motivation, and associated disorders such as depression and addiction. This suggests that the neural substrate instrumental for these behaviors, including the nucleus accumbens core (AcbC), likewise differs between estrous cycle phases. It is unknown whether the electrophysiological properties of AcbC output neurons, medium spiny neurons (MSNs), change between estrous cycle phases. This is a critical knowledge gap given that MSN electrophysiological properties are instrumental for determining AcbC output to efferent targets. Here we test whether the intrinsic electrophysiological properties of adult rat AcbC MSNs differ across female estrous cycle phases and from males. We recorded MSNs with whole cell patch-clamp technique in two experiments, the first using gonad-intact adult males and females in differing phases of the estrous cycle and the second using gonadectomized males and females in which the estrous cycle was eliminated. MSN intrinsic electrophysiological and excitatory synaptic input properties robustly changed between female estrous cycle phases and males. Sex differences in MSN electrophysiology disappeared when the estrous cycle was eliminated. These novel findings indicate that AcbC MSN electrophysiological properties change across the estrous cycle, providing a new framework for understanding how biological sex and hormone cyclicity regulate motivated behaviors and other AcbC functions and disorders. NEW & NOTEWORTHY This research is the first demonstration that medium spiny neuron electrophysiological properties change across adult female hormone cycle phases in any striatal region. This influence of estrous cycle engenders sex differences in electrophysiological properties that are eliminated by gonadectomy. Broadly, these findings indicate that adult female hormone cycles are an important factor for neurophysiology.
Collapse
Affiliation(s)
- Stephanie B Proaño
- Graduate Program in Biology, North Carolina State University , Raleigh, North Carolina.,W. M. Keck Center for Behavioral Biology, North Carolina State University , Raleigh, North Carolina.,Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
| | - Hannah J Morris
- Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
| | - Lindsey M Kunz
- Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
| | - David M Dorris
- Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
| | - John Meitzen
- W. M. Keck Center for Behavioral Biology, North Carolina State University , Raleigh, North Carolina.,Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina.,Center for Human Health and the Environment, North Carolina State University , Raleigh, North Carolina.,Comparative Medicine Institute, North Carolina State University , Raleigh, North Carolina
| |
Collapse
|
34
|
Tonn Eisinger KR, Larson EB, Boulware MI, Thomas MJ, Mermelstein PG. Membrane estrogen receptor signaling impacts the reward circuitry of the female brain to influence motivated behaviors. Steroids 2018; 133:53-59. [PMID: 29195840 PMCID: PMC5864533 DOI: 10.1016/j.steroids.2017.11.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 11/23/2017] [Indexed: 12/12/2022]
Abstract
Within the adult female, estrogen signaling is well-described as an integral component of the physiologically significant hypothalamic-pituitary-gonadal axis. In rodents, the timing of ovulation is intrinsically entwined with the display of sexual receptivity. For decades, the importance of estradiol activating intracellular estrogen receptors within the hypothalamus and midbrain/spinal cord lordosis circuits has been appreciated. These signaling pathways primarily account for the ability of the female to reproduce. Yet, often overlooked is that the desire to reproduce is also tightly regulated by estrogen receptor signaling. This lack of emphasis can be attributed to an absence of nuclear estrogen receptors in brain regions associated with reward, such as the nucleus accumbens, which are associated with motivated behaviors. This review outlines how membrane-localized estrogen receptors affect metabotropic glutamate receptor signaling within the rodent nucleus accumbens. In addition, we discuss how, as estrogens drive increased motivation for reproduction, they also produce the untoward side effect of heightening female vulnerability to drug addiction.
Collapse
Affiliation(s)
- Katherine R Tonn Eisinger
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Erin B Larson
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Marissa I Boulware
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mark J Thomas
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Paul G Mermelstein
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
35
|
Meitzen J, Meisel RL, Mermelstein PG. Sex Differences and the Effects of Estradiol on Striatal Function. Curr Opin Behav Sci 2018; 23:42-48. [PMID: 30221186 DOI: 10.1016/j.cobeha.2018.03.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The striatal brain regions, including the caudate-putamen, nucleus accumbens core, and nucleus accumbens shell, mediate critical behavioral functions. These functions include but are not limited to motivated behavior, reward, learning, and sensorimotor function in both pathological and normal contexts. The phenotype and/or incidence of all of these behaviors either differ by sex or are sensitive to the presence of gonadal hormones such as 17β-estradiol and testosterone. All three striatal brain regions express membrane-associated estrogen receptors. Here we present a brief review of the recent literature reporting on sex differences and effects of the estrogenic hormone 17β-estradiol on behavioral and neural function across all three striatal regions, focusing upon the most prominent striatal neuron type, the medium spiny neuron. We emphasize recent findings in three broad domains: (1) select striatal-relevant behaviors and disorders, (2) striatal medium spiny neuron dendritic spine density, and (3), striatal medium spiny neuron electrophysiological properties including excitatory synaptic input and intrinsic cellular excitability. These recent advances in behavior, neuroanatomy, and electrophysiology collectively offer insight into the effects of sex and estrogen on striatal function, especially at the level of individual neurons.
Collapse
Affiliation(s)
- John Meitzen
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC.,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC.,Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Robert L Meisel
- Dept. of Neuroscience, University of Minnesota, Minneapolis, MN
| | | |
Collapse
|
36
|
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
|
37
|
Abstract
The hypothalamus is most often associated with innate behaviors such as is hunger, thirst and sex. While the expression of these behaviors important for survival of the individual or the species is nested within the hypothalamus, the desire (i.e., motivation) for them is centered within the mesolimbic reward circuitry. In this review, we will use female sexual behavior as a model to examine the interaction of these circuits. We will examine the evidence for a hypothalamic circuit that regulates consummatory aspects of reproductive behavior, i.e., lordosis behavior, a measure of sexual receptivity that involves estradiol membrane-initiated signaling in the arcuate nucleus (ARH), activating β-endorphin projections to the medial preoptic nucleus (MPN), which in turn modulate ventromedial hypothalamic nucleus (VMH) activity-the common output from the hypothalamus. Estradiol modulates not only a series of neuropeptides, transmitters and receptors but induces dendritic spines that are for estrogenic induction of lordosis behavior. Simultaneously, in the nucleus accumbens of the mesolimbic system, the mating experience produces long term changes in dopamine signaling and structure. Sexual experience sensitizes the response of nucleus accumbens neurons to dopamine signaling through the induction of a long lasting early immediate gene. While estrogen alone increases spines in the ARH, sexual experience increases dendritic spine density in the nucleus accumbens. These two circuits appear to converge onto the medial preoptic area where there is a reciprocal influence of motivational circuits on consummatory behavior and vice versa. While it has not been formally demonstrated in the human, such circuitry is generally highly conserved and thus, understanding the anatomy, neurochemistry and physiology can provide useful insight into the motivation for sexual behavior and other innate behaviors in humans.
Collapse
Affiliation(s)
- Paul E Micevych
- Laboratory of Neuroendocrinology, Department of Neurobiology, David Geffen School of Medicine at University of California, Los AngelesLos Angeles, CA, United States.,Brain Research Institute, University of California, Los AngelesLos Angeles, CA, United States
| | - Robert L Meisel
- Department of Neuroscience, University of MinnesotaMinneapolis, MN, United States
| |
Collapse
|
38
|
Peterson BM, Martinez LA, Meisel RL, Mermelstein PG. Estradiol impacts the endocannabinoid system in female rats to influence behavioral and structural responses to cocaine. Neuropharmacology 2016; 110:118-124. [PMID: 27266915 PMCID: PMC5028287 DOI: 10.1016/j.neuropharm.2016.06.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/31/2016] [Accepted: 06/02/2016] [Indexed: 12/31/2022]
Abstract
Compared with men, women show enhanced responses to drugs of abuse, and consequently are thought to be more vulnerable to addiction. The ovarian hormone estradiol has emerged as a key facilitator in the heightened development of addiction in females. These actions of estradiol appear mediated by estrogen receptor (ER) activation of metabotropic glutamate receptor type 5 (mGluR5). However, the downstream effectors of this ER/mGluR5 signaling pathway are unknown. Here we investigate whether cannabinoid 1 receptor (CB1R) activation is a part of the mechanism whereby estradiol influences behavioral and synaptic correlates of addiction. Following repeated cocaine administration, estradiol-treated ovariectomized rats exhibited both sensitized locomotor responses and decreases in the dendritic spine density of nucleus accumbens core medium-spiny neurons in comparison to oil-treated controls. Both effects of estradiol were blocked by AM251, a CB1R inverse agonist. These results indicate that part of the signaling mechanism through which estradiol impacts behavioral and synaptic correlates of addiction in female rats requires activation of CB1Rs.
Collapse
Affiliation(s)
- Brittni M Peterson
- Graduate Program in Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA; Department of Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Luis A Martinez
- Department of Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Robert L Meisel
- Graduate Program in Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA; Department of Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Paul G Mermelstein
- Graduate Program in Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA; Department of Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
39
|
Estradiol Facilitation of Cocaine Self-Administration in Female Rats Requires Activation of mGluR5. eNeuro 2016; 3:eN-NWR-0140-16. [PMID: 27822496 PMCID: PMC5079229 DOI: 10.1523/eneuro.0140-16.2016] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 10/07/2016] [Accepted: 10/07/2016] [Indexed: 02/06/2023] Open
Abstract
In comparison to men, women initiate drug use at earlier ages and progress from initial use to addiction more rapidly. This heightened intake and vulnerability to drugs of abuse is regulated in part by estradiol, although the signaling mechanisms by which this occurs are not well understood. Recent findings indicate that within the nucleus accumbens core, estradiol induces structural plasticity via membrane-localized estrogen receptor α, functionally coupled to metabotropic glutamate receptor subtype 5 (mGluR5). Hence, we sought to determine whether mGluR5 activation was essential for estradiol-mediated enhancement of cocaine self-administration. Ovariectomized (OVX) female rats were allowed to freely self-administer cocaine under extended access conditions (6 h/d) for 10 consecutive days. The mGluR5 antagonist 2-methyl-6-(phenylethynyl)pyridine hydrochloride (MPEP) or vehicle was administered before estradiol (or oil), on a 2 d on/2 d off schedule throughout the extended access period. MPEP treatment prevented the estradiol-dependent enhancement of cocaine self-administration in OVX females. In a separate experiment, potentiation of mGluR5 function with the positive allosteric modulator 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (in the absence of estradiol treatment) failed to increase cocaine self-administration. These data suggest that mGluR5 activation is necessary for estradiol-mediated enhancement of responses to cocaine, but that direct mGluR5 activation is insufficient to mimic the female response to estradiol. Building on previous studies in male animals, these findings further highlight the therapeutic potential of mGluR5 antagonism in the treatment of addiction and suggest that there may be added therapeutic benefit in females.
Collapse
|
40
|
Cao J, Dorris DM, Meitzen J. Neonatal Masculinization Blocks Increased Excitatory Synaptic Input in Female Rat Nucleus Accumbens Core. Endocrinology 2016; 157:3181-96. [PMID: 27285859 PMCID: PMC4967116 DOI: 10.1210/en.2016-1160] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/04/2016] [Indexed: 01/18/2023]
Abstract
Steroid sex hormones and genetic sex regulate the phenotypes of motivated behaviors and relevant disorders. Most studies seeking to elucidate the underlying neuroendocrine mechanisms have focused on how 17β-estradiol modulates the role of dopamine in striatal brain regions, which express membrane-associated estrogen receptors. Dopamine action is an important component of striatal function, but excitatory synaptic neurotransmission has also emerged as a key striatal substrate and target of estradiol action. Here, we focus on excitatory synaptic input onto medium spiny neurons (MSNs) in the striatal region nucleus accumbens core (AcbC). In adult AcbC, miniature excitatory postsynaptic current (mEPSC) frequency is increased in female compared with male MSNs. We tested whether increased mEPSC frequency in female MSNs exists before puberty, whether this increased excitability is due to the absence of estradiol or testosterone during the early developmental critical period, and whether it is accompanied by stable neuron intrinsic membrane properties. We found that mEPSC frequency is increased in female compared with male MSNs before puberty. Increased mEPSC frequency in female MSNs is abolished after neonatal estradiol or testosterone exposure. MSN intrinsic membrane properties did not differ by sex. These data indicate that neonatal masculinization via estradiol and/or testosterone action is sufficient for down-regulating excitatory synaptic input onto MSNs. We conclude that excitatory synaptic input onto AcbC MSNs is organized long before adulthood via steroid sex hormone action, providing new insight into a mechanism by which sex differences in motivated behavior and other AbcC functions may be generated or compromised.
Collapse
Affiliation(s)
- Jinyan Cao
- Department of Biological Sciences (J.C., D.M.D., J.M.), North Carolina State University, Raleigh, North Carolina 27695; W.M. Keck Center for Behavioral Biology (J.C., J.M.), North Carolina State University, Raleigh, North Carolina 27695; Center for Human Health and the Environment (J.M.), North Carolina State University, Raleigh, North Carolina 27695; and Comparative Medicine Institute (J.M.), North Carolina State University, Raleigh, North Carolina 27695
| | - David M Dorris
- Department of Biological Sciences (J.C., D.M.D., J.M.), North Carolina State University, Raleigh, North Carolina 27695; W.M. Keck Center for Behavioral Biology (J.C., J.M.), North Carolina State University, Raleigh, North Carolina 27695; Center for Human Health and the Environment (J.M.), North Carolina State University, Raleigh, North Carolina 27695; and Comparative Medicine Institute (J.M.), North Carolina State University, Raleigh, North Carolina 27695
| | - John Meitzen
- Department of Biological Sciences (J.C., D.M.D., J.M.), North Carolina State University, Raleigh, North Carolina 27695; W.M. Keck Center for Behavioral Biology (J.C., J.M.), North Carolina State University, Raleigh, North Carolina 27695; Center for Human Health and the Environment (J.M.), North Carolina State University, Raleigh, North Carolina 27695; and Comparative Medicine Institute (J.M.), North Carolina State University, Raleigh, North Carolina 27695
| |
Collapse
|
41
|
No Evidence for Sex Differences in the Electrophysiological Properties and Excitatory Synaptic Input onto Nucleus Accumbens Shell Medium Spiny Neurons. eNeuro 2016; 3:eN-NRS-0147-15. [PMID: 27022621 PMCID: PMC4757778 DOI: 10.1523/eneuro.0147-15.2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/19/2016] [Accepted: 01/25/2016] [Indexed: 02/01/2023] Open
Abstract
Sex differences exist in how the brain regulates motivated behavior and reward, both in normal and pathological contexts. Investigations into the underlying neural mechanisms have targeted the striatal brain regions, including the dorsal striatum and nucleus accumbens core and shell. Sex differences exist in how the brain regulates motivated behavior and reward, both in normal and pathological contexts. Investigations into the underlying neural mechanisms have targeted the striatal brain regions, including the dorsal striatum and nucleus accumbens core and shell. These investigations yield accumulating evidence of sexually different electrophysiological properties, excitatory synaptic input, and sensitivity to neuromodulator/hormone action in select striatal regions both before and after puberty. It is unknown whether the electrical properties of neurons in the nucleus accumbens shell differ by sex, and whether sex differences in excitatory synaptic input are present before puberty. To test the hypothesis that these properties differ by sex, we performed whole-cell patch-clamp recordings on male and female medium spiny neurons (MSNs) in acute brain slices obtained from prepubertal rat nucleus accumbens shell. We analyzed passive and active electrophysiological properties, and miniature EPSCs (mEPSCs). No sex differences were detected; this includes those properties, such as intrinsic excitability, action potential afterhyperpolarization, threshold, and mEPSC frequency, that have been found to differ by sex in other striatal regions and/or developmental periods. These findings indicate that, unlike other striatal brain regions, the electrophysiological properties of nucleus accumbens shell MSNs do not differ by sex. Overall, it appears that sex differences in striatal function, including motivated behavior and reward, are likely mediated by other factors and striatal regions.
Collapse
|
42
|
Wong JE, Cao J, Dorris DM, Meitzen J. Genetic sex and the volumes of the caudate-putamen, nucleus accumbens core and shell: original data and a review. Brain Struct Funct 2015; 221:4257-4267. [PMID: 26666530 DOI: 10.1007/s00429-015-1158-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 11/24/2015] [Indexed: 11/24/2022]
Abstract
Sex differences are widespread across vertebrate nervous systems. Such differences are sometimes reflected in the neural substrate via neuroanatomical differences in brain region volume. One brain region that displays sex differences in its associated functions and pathologies is the striatum, including the caudate-putamen (dorsal striatum), nucleus accumbens core and shell (ventral striatum). The extent to which these differences can be attributed to alterations in volume is unclear. We thus tested whether the volumes of the caudate-putamen, nucleus accumbens core, and nucleus accumbens shell differed by region, sex, and hemisphere in adult Sprague-Dawley rats. As a positive control for detecting sex differences in brain region volume, we measured the sexually dimorphic nucleus of the medial preoptic area (SDN-POA). As expected, SDN-POA volume was larger in males than in females. No sex differences were detected in the volumes of the caudate-putamen, nucleus accumbens core or shell. Nucleus accumbens core volume was larger in the right than left hemisphere across males and females. These findings complement previous reports of lateralized nucleus accumbens volume in humans, and suggest that this may possibly be driven via hemispheric differences in nucleus accumbens core volume. In contrast, striatal sex differences seem to be mediated by factors other than striatal region volume. This conclusion is presented within the context of a detailed review of studies addressing sex differences and similarities in striatal neuroanatomy.
Collapse
Affiliation(s)
- Jordan E Wong
- Department of Biological Sciences, North Carolina State University, Campus Box 7617, Raleigh, NC, 27695-7617, USA
| | - Jinyan Cao
- Department of Biological Sciences, North Carolina State University, Campus Box 7617, Raleigh, NC, 27695-7617, USA.,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA
| | - David M Dorris
- Department of Biological Sciences, North Carolina State University, Campus Box 7617, Raleigh, NC, 27695-7617, USA
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Campus Box 7617, Raleigh, NC, 27695-7617, USA. .,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA. .,Center for Human Health and the Environment, Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
43
|
Dorris DM, Cao J, Willett JA, Hauser CA, Meitzen J. Intrinsic excitability varies by sex in prepubertal striatal medium spiny neurons. J Neurophysiol 2014; 113:720-9. [PMID: 25376786 DOI: 10.1152/jn.00687.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sex differences in neuron electrophysiological properties were traditionally associated with brain regions directly involved in reproduction in adult, postpubertal animals. There is growing acknowledgement that sex differences can exist in other developmental periods and brain regions as well. This includes the dorsal striatum (caudate/putamen), which shows robust sex differences in gene expression, neuromodulator action (including dopamine and 17β-estradiol), and relevant sensorimotor behaviors and pathologies such as the responsiveness to drugs of abuse. Here we examine whether these sex differences extend to striatal neuron electrophysiology. We test the hypothesis that passive and active medium spiny neuron (MSN) electrophysiological properties in prepubertal rat dorsal striatum differ by sex. We made whole cell recordings from male and females MSNs from acute brain slices. The slope of the evoked firing rate to current injection curve was increased in MSNs recorded from females compared with males. The initial action potential firing rate was increased in MSNs recorded from females compared with males. Action potential after-hyperpolarization peak was decreased, and threshold was hyperpolarized in MSNs recorded from females compared with males. No sex differences in passive electrophysiological properties or miniature excitatory synaptic currents were detected. These findings indicate that MSN excitability is increased in prepubertal females compared with males, providing a new mechanism that potentially contributes to generating sex differences in striatal-mediated processes. Broadly, these findings demonstrate that sex differences in neuron electrophysiological properties can exist prepuberty in brain regions not directly related to reproduction.
Collapse
Affiliation(s)
- David M Dorris
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
| | - Jinyan Cao
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina; W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina
| | - Jaime A Willett
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina; W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina; Graduate Program in Physiology, North Carolina State University, Raleigh, North Carolina
| | - Caitlin A Hauser
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina; W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina; Center for Human Health and the Environment, Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina; and Grass Laboratory, Marine Biological Laboratory, Woods Hole, Massachusetts
| |
Collapse
|
44
|
Martinez LA, Peterson BM, Meisel RL, Mermelstein PG. Estradiol facilitation of cocaine-induced locomotor sensitization in female rats requires activation of mGluR5. Behav Brain Res 2014; 271:39-42. [PMID: 24893316 DOI: 10.1016/j.bbr.2014.05.052] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/20/2014] [Accepted: 05/24/2014] [Indexed: 12/24/2022]
Abstract
In comparison to men, women exhibit enhanced responsiveness to the stimulating and addictive properties of cocaine. A growing body of evidence implicates the steroid hormone estradiol in mediating this sex difference, yet the mechanisms underlying estradiol enhancement of behavioral responses to cocaine in females are not known. Recently, we have found that estrogen receptor alpha (ERα) functionally couples with the metabotropic glutamate receptor 5 (mGluR5) to mediate the effects of estradiol on both cellular activation as well as dendritic spine plasticity in brain regions involved in cocaine-induced behavioral sensitization. Thus, we sought to determine whether mGluR5 activation is required for the facilitative effects of estradiol on locomotor responses to cocaine. To test this hypothesis, ovariectomized (OVX) female rats were tested for locomotor activity on the first and fifth days of daily systemic injections of cocaine. For the 2 days prior to each locomotor test, animals were injected with the mGluR5 antagonist MPEP (or vehicle) and estradiol (or oil). MPEP treatment blocked the facilitative effects of estradiol on cocaine-induced locomotor sensitization, without affecting acute responses to cocaine or the inhibitory actions of estradiol on weight gain. Considered together, these data indicate that mGluR5 activation is critical for the actions of estradiol on cocaine-induced behavioral sensitization.
Collapse
Affiliation(s)
- Luis A Martinez
- Department of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church St SE, Minneapolis, MN 55455, USA.
| | - Brittni M Peterson
- Department of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church St SE, Minneapolis, MN 55455, USA; Graduate Program of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Robert L Meisel
- Department of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church St SE, Minneapolis, MN 55455, USA; Graduate Program of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church St SE, Minneapolis, MN 55455, USA; Graduate Program of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church St SE, Minneapolis, MN 55455, USA
| |
Collapse
|
45
|
Peterson BM, Mermelstein PG, Meisel RL. Estradiol mediates dendritic spine plasticity in the nucleus accumbens core through activation of mGluR5. Brain Struct Funct 2014; 220:2415-22. [PMID: 24878822 DOI: 10.1007/s00429-014-0794-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/11/2014] [Indexed: 11/26/2022]
Abstract
Accumulating evidence from human and rodent studies suggests that females are more sensitive to the motivating and rewarding properties of drugs of abuse. Numerous reports implicate estradiol in enhancing drug-related responses in females, yet the neurobiological mechanisms underlying this effect of estradiol are unknown. Because dendritic spine plasticity in the nucleus accumbens (NAc) is linked to the addictive effects of drugs, we examined the influence of estradiol on dendritic spines in this region. Previously our laboratory demonstrated that in female medium spiny neurons, estradiol activates metabotropic glutamate receptor subtype five (mGluR5), a G protein-coupled receptor already implicated in the etiology of drug addiction. Thus, we sought to determine whether mGluR5 is a part of the mechanism by which estradiol affects dendritic spine density in the NAc. To test this hypothesis, ovariectomized female rats were treated with the mGluR5 antagonist, MPEP, or vehicle prior to estradiol (or oil) treatment and 24 h later dendritic spine density was evaluated by DiI labeling and confocal microscopy. We found that estradiol decreased dendritic spine density in the NAc core and that pretreatment with MPEP blocked this effect. In contrast, MPEP had no effect on dendritic spine density in the NAc shell or CA1 region of the hippocampus, two regions in which estradiol increased the density of dendritic spines. As dendritic spine plasticity in the NAc core has behavioral consequences for drug addiction, these data provide a clue as to how estradiol acts in females to enhance behavioral responses to drugs of abuse.
Collapse
Affiliation(s)
- Brittni M Peterson
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA,
| | | | | |
Collapse
|
46
|
Urošević S, Collins P, Muetzel R, Lim KO, Luciana M. Pubertal status associations with reward and threat sensitivities and subcortical brain volumes during adolescence. Brain Cogn 2014; 89:15-26. [PMID: 24512818 DOI: 10.1016/j.bandc.2014.01.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 01/06/2014] [Accepted: 01/08/2014] [Indexed: 12/19/2022]
Abstract
Adolescence is characterized by complex developmental processes that impact behavior, biology, and social functioning. Two such adolescence-specific processes are puberty and increases in reward sensitivity. Relations between these processes are poorly understood. The present study focused on examining unique effects of puberty, age, and sex on reward and threat sensitivities and volumes of subcortical brain structures relevant for reward/threat processing in a healthy sample of 9-18year-olds. Unlike age, pubertal status had a significant unique positive relationship with reward sensitivity. In addition, there was a trend for adolescent females to exhibit higher threat sensitivity with more advanced pubertal development and higher reward and threat sensitivity with older age. Similarly, there were significant puberty by sex interaction effects on striatal volumes, i.e., left nucleus accumbens and right pallidum. The present pattern of results suggests that pubertal development, independent of chronological age, is uniquely associated with reward hypersensitivity and with structural differences in striatal regions implicated in reward processing.
Collapse
Affiliation(s)
- Snežana Urošević
- Department of Psychology, University of Minnesota-Twin Cities, N218 Elliott Hall, 75 East River Parkway, Minneapolis, MN 55408, United States.
| | - Paul Collins
- Department of Psychology, University of Minnesota-Twin Cities, N218 Elliott Hall, 75 East River Parkway, Minneapolis, MN 55408, United States
| | - Ryan Muetzel
- Department of Psychiatry, University of Minnesota-Twin Cities, F282/2A West, 2450 Riverside Avenue South, Minneapolis, MN 55454, United States
| | - Kelvin O Lim
- Department of Psychiatry, University of Minnesota-Twin Cities, F282/2A West, 2450 Riverside Avenue South, Minneapolis, MN 55454, United States
| | - Monica Luciana
- Department of Psychology, University of Minnesota-Twin Cities, N218 Elliott Hall, 75 East River Parkway, Minneapolis, MN 55408, United States
| |
Collapse
|
47
|
Micevych P, Sinchak K. Temporal and concentration-dependent effects of oestradiol on neural pathways mediating sexual receptivity. J Neuroendocrinol 2013; 25:1012-23. [PMID: 24028299 PMCID: PMC3943611 DOI: 10.1111/jne.12103] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 08/13/2013] [Accepted: 09/05/2013] [Indexed: 11/27/2022]
Abstract
The acceptance of oestradiol signalling through receptors found in the cell membrane, as well as, the nucleus, has provided for a re-examination of the timing and location of the actions of oestradiol on neural circuits mediating sexual receptivity (lordosis). Oestradiol membrane signalling involves the transactivation of metabotrophic glutamate receptors (mGluRs) that transduce steroid information through protein kinase C signalling cascades producing rapid activation of lordosis-regulating circuits. It has been known for some time that oestradiol initially produces an inhibition of the medial preoptic nucleus. We have demonstrated that underlying this inhibition is oestradiol acting in the arcuate nucleus to induce β-endorphin release, which inhibits the medial preoptic nucleus through a μ-opioid receptor mechanism. This transient inhibition is relieved by either subsequent progesterone treatment or longer exposure to higher doses of oestradiol to facilitate lordosis behaviour. We review recent findings about oestradiol membrane signalling inducing dendritic spine formation in the arcuate nucleus that is critical for oestradiol induction of sexual receptivity. Moreover, we discuss the evidence that, in addition to oestrogen receptor α, several other putative membrane oestrogen receptors facilitate lordosis behaviour through regulation of the arcuate nucleus. These include the GRP30 and the STX activated Gq-mER. Finally, we report on the importance of GABA acting at GABAB receptors for oestradiol membrane signalling that regulates lordosis circuit activation and sexual receptivity.
Collapse
Affiliation(s)
- Paul Micevych
- Department of Neurobiology, David Geffen School of Medicine at UCLA, the Laboratory of Neuroendocrinology, Brain Research Institute, University of California, Los Angeles, CA 90095
| | - Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, CA 90840
| |
Collapse
|
48
|
Abstract
Since this journal's inception, the field of adolescent brain development has flourished, as researchers have investigated the underpinnings of adolescent risk-taking behaviors. Explanations based on translational models initially attributed such behaviors to executive control deficiencies and poor frontal lobe function. This conclusion was bolstered by evidence that the prefrontal cortex and its interconnections are among the last brain regions to structurally and functionally mature. As substantial heterogeneity of prefrontal function was revealed, applications of neuroeconomic theory to adolescent development led to dual systems models of behavior. Current epidemiological trends, behavioral observations, and functional magnetic resonance imaging based brain activity patterns suggest a quadratic increase in limbically mediated incentive motivation from childhood to adolescence and a decline thereafter. This elevation occurs in the context of immature prefrontal function, so motivational strivings may be difficult to regulate. Theoretical models explain this patterning through brain-based accounts of subcortical-cortical integration, puberty-based models of adolescent sensation seeking, and neurochemical dynamics. Empirically sound tests of these mechanisms, as well as investigations of biology-context interactions, represent the field's most challenging future goals, so that applications to psychopathology can be refined and so that developmental cascades that incorporate neurobiological variables can be modeled.
Collapse
|
49
|
Bayer J, Bandurski P, Sommer T. Differential modulation of activity related to the anticipation of monetary gains and losses across the menstrual cycle. Eur J Neurosci 2013; 38:3519-26. [DOI: 10.1111/ejn.12347] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 07/16/2013] [Accepted: 07/26/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Janine Bayer
- Department of Systems Neuroscience; University Medical Center Hamburg-Eppendorf; Martinistr. 52 Hamburg 20246 Germany
| | - Pia Bandurski
- Department of Systems Neuroscience; University Medical Center Hamburg-Eppendorf; Martinistr. 52 Hamburg 20246 Germany
| | - Tobias Sommer
- Department of Systems Neuroscience; University Medical Center Hamburg-Eppendorf; Martinistr. 52 Hamburg 20246 Germany
| |
Collapse
|
50
|
Cell-type specific increases in female hamster nucleus accumbens spine density following female sexual experience. Brain Struct Funct 2013; 219:2071-81. [PMID: 23934655 DOI: 10.1007/s00429-013-0624-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/02/2013] [Indexed: 01/01/2023]
Abstract
Female sexual behavior is an established model of a naturally motivated behavior which is regulated by activity within the mesolimbic dopamine system. Repeated activation of the mesolimbic circuit by female sexual behavior elevates dopamine release and produces persistent postsynaptic alterations to dopamine D1 receptor signaling within the nucleus accumbens. Here we demonstrate that sexual experience in female Syrian hamsters significantly increases spine density and alters morphology selectively in D1 receptor-expressing medium spiny neurons within the nucleus accumbens core, with no corresponding change in dopamine receptor binding or protein expression. Our findings demonstrate that previous life experience with a naturally motivated behavior has the capacity to induce persistent structural alterations to the mesolimbic circuit that can increase reproductive success and are analogous to the persistent structural changes following repeated exposure to many drugs of abuse.
Collapse
|