101
|
A sex-dependent role for the prelimbic cortex in impulsive action both before and following early cocaine abstinence. Neuropsychopharmacology 2021; 46:1565-1573. [PMID: 33972695 PMCID: PMC8280154 DOI: 10.1038/s41386-021-01024-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/29/2021] [Accepted: 04/16/2021] [Indexed: 11/08/2022]
Abstract
Although impulsive action is strongly associated with addiction, the neural underpinnings of this relationship and how they are influenced by sex have not been well characterized. Here, we used a titrating reaction time task to assess differences in impulsive action in male and female Long Evans rats both before and after short (4-6 days) or long (25-27 days) abstinence from 2 weeks of cocaine or water/saline self-administration (6 h daily access). Neural activity in the prelimbic cortex (PrL) and nucleus accumbens (NAc) core was assessed at each time point. We found that a history of cocaine self-administration increased impulsivity in all rats following short, but not long, abstinence. Furthermore, male rats with an increased ratio of excited to inhibited neurons in the PrL at the start of each trial in the task exhibited higher impulsivity in the naïve state (before self-administration). Following short abstinence from cocaine, PrL activity in males became more inhibited, and this change in activity predicted the shift in impulsivity. However, PrL activity did not track impulsivity in female rats. Additionally, although the NAc core tracked several aspects of behavior in the task, it did not track impulsivity in either sex. Together, these findings demonstrate a sex-dependent role for the PrL in impulsivity both before and after a history of cocaine.
Collapse
|
102
|
Altidor LKP, Bruner MM, Deslauriers JF, Garman TS, Ramirez S, Dirr EW, Olczak KP, Maurer AP, Lamb DG, Otto KJ, Burke SN, Bumanglag AV, Setlow B, Bizon JL. Acute vagus nerve stimulation enhances reversal learning in rats. Neurobiol Learn Mem 2021; 184:107498. [PMID: 34332068 DOI: 10.1016/j.nlm.2021.107498] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/01/2021] [Accepted: 07/24/2021] [Indexed: 01/19/2023]
Abstract
Cognitive flexibility is a prefrontal cortex-dependent neurocognitive process that enables behavioral adaptation in response to changes in environmental contingencies. Electrical vagus nerve stimulation (VNS) enhances several forms of learning and neuroplasticity, but its effects on cognitive flexibility have not been evaluated. In the current study, a within-subjects design was used to assess the effects of VNS on performance in a novel visual discrimination reversal learning task conducted in touchscreen operant chambers. The task design enabled simultaneous assessment of acute VNS both on reversal learning and on recall of a well-learned discrimination problem. Acute VNS delivered in conjunction with stimuli presentation during reversal learning reliably enhanced learning of new reward contingencies. Enhancement was not observed, however, if VNS was delivered during the session but was not coincident with presentation of to-be-learned stimuli. In addition, whereas VNS delivered at 30 HZ enhanced performance, the same enhancement was not observed using 10 or 50 Hz. Together, these data show that acute VNS facilitates reversal learning and indicate that the timing and frequency of the VNS are critical for these enhancing effects. In separate rats, administration of the norepinephrine reuptake inhibitor atomoxetine also enhanced reversal learning in the same task, consistent with a noradrenergic mechanism through which VNS enhances cognitive flexibility.
Collapse
Affiliation(s)
| | - Matthew M Bruner
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | | | - Tyler S Garman
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Saúl Ramirez
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Elliott W Dirr
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Kaitlynn P Olczak
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Andrew P Maurer
- Department of Neuroscience, University of Florida, Gainesville, FL, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Evelyn F. & William L. McKnight Brain Institute, University of Florida, USA; Engineering School of Sustainable Infrastructure and Environment, University of Florida, Gainesville, FL, USA
| | - Damon G Lamb
- Department of Neuroscience, University of Florida, Gainesville, FL, USA; Department of Psychiatry, University of Florida, Gainesville, FL, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Evelyn F. & William L. McKnight Brain Institute, University of Florida, USA; Brain Rehabilitation Research Center, Malcom Randall VAMC, Gainesville, FL, USA
| | - Kevin J Otto
- Department of Neuroscience, University of Florida, Gainesville, FL, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Evelyn F. & William L. McKnight Brain Institute, University of Florida, USA
| | - Sara N Burke
- Department of Neuroscience, University of Florida, Gainesville, FL, USA; Evelyn F. & William L. McKnight Brain Institute, University of Florida, USA
| | - Argyle V Bumanglag
- Department of Neuroscience, University of Florida, Gainesville, FL, USA; Evelyn F. & William L. McKnight Brain Institute, University of Florida, USA
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL, USA; Evelyn F. & William L. McKnight Brain Institute, University of Florida, USA
| | - Jennifer L Bizon
- Department of Neuroscience, University of Florida, Gainesville, FL, USA; Evelyn F. & William L. McKnight Brain Institute, University of Florida, USA.
| |
Collapse
|
103
|
Peters J, Olson DE. Engineering Safer Psychedelics for Treating Addiction. Neurosci Insights 2021; 16:26331055211033847. [PMID: 34350400 PMCID: PMC8295933 DOI: 10.1177/26331055211033847] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 06/30/2021] [Indexed: 12/03/2022] Open
Abstract
Addiction is best described as a disorder of maladaptive neuroplasticity involving the simultaneous strengthening of reward circuitry that drives compulsive drug seeking and weakening of circuits involved in executive control over harmful behaviors. Psychedelics have shown great promise for treating addiction, with many people attributing their therapeutic effects to insights gained while under the influence of the drug. However, psychedelics are also potent psychoplastogens—molecules capable of rapidly re-wiring the adult brain. The advent of non-hallucinogenic psychoplastogens with anti-addictive properties raises the intriguing possibility that hallucinations might not be necessary for all therapeutic effects of psychedelic-based medicines, so long as the underlying pathological neural circuitry can be remedied. One of these non-hallucinogenic psychoplastogens, tabernanthalog (TBG), appears to have long-lasting therapeutic effects in preclinical models relevant to alcohol and opioid addiction. Here, we discuss the implications of these results for the development of addiction treatments, as well as the next steps for advancing TBG and related non-hallucinogenic psychoplastogens as addiction therapeutics.
Collapse
Affiliation(s)
- Jamie Peters
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - David E Olson
- Department of Chemistry, University of California, Davis, Davis, CA, USA.,Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA.,Center for Neuroscience, University of California, Davis, Davis, CA, USA
| |
Collapse
|
104
|
Dissociable roles of the nucleus accumbens core and shell subregions in the expression and extinction of conditioned fear. Neurobiol Stress 2021; 15:100365. [PMID: 34355048 PMCID: PMC8319794 DOI: 10.1016/j.ynstr.2021.100365] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/23/2021] [Accepted: 07/09/2021] [Indexed: 12/25/2022] Open
Abstract
The nucleus accumbens (NAc), consisting of core (NAcC) and shell (NAcS) sub-regions, has primarily been studied as a locus mediating the effects of drug reward and addiction. However, there is ample evidence that this region is also involved in regulating aversive responses, but the exact role of the NAc and its subregions in regulating associative fear processing remains unclear. Here, we investigated the specific contribution of the NAcC and NAcS in regulating both fear expression and fear extinction in C57BL/6J mice. Using Arc expression as an indicator of neuronal activity, we first show that the NAcC is specifically active only in response to an associative fear cue during an expression test. In contrast, the NAcS is specifically active during fear extinction. We next inactivated each subregion using lidocaine and demonstrated that the NAcC is necessary for fear expression, but not for extinction learning or consolidation of extinction. In contrast, we demonstrate that the NAcS is necessary for the consolidation of extinction, but not fear expression or extinction learning. Further, inactivation of mGluR1 or ERK signaling specifically in the NAcS disrupted the consolidation of extinction but had no effect on fear expression or extinction learning itself. Our data provide the first evidence for the importance of the ERK/MAPK pathway as the underlying neural mechanism facilitating extinction consolidation within the NAcS. These findings suggest that the NAc subregions play dissociable roles in regulating fear recall and the consolidation of fear extinction, and potentially implicate them as critical regions within the canonical fear circuit.
Collapse
|
105
|
Prelimbic cortical projections to rostromedial tegmental nucleus play a suppressive role in cue-induced reinstatement of cocaine seeking. Neuropsychopharmacology 2021; 46:1399-1406. [PMID: 33230269 PMCID: PMC8209220 DOI: 10.1038/s41386-020-00909-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022]
Abstract
The prelimbic (PL) region of prefrontal cortex has been implicated in both driving and suppressing cocaine seeking in animal models of addiction. We hypothesized that these opposing roles for PL may be supported by distinct efferent projections. While PL projections to nucleus accumbens core have been shown to be involved in driving reinstatement of cocaine seeking, PL projections to the rostromedial tegmental nucleus (RMTg) may instead suppress reinstatement of cocaine seeking, due to the role of RMTg in behavioral inhibition. Here, we used a functional disconnection approach to temporarily disrupt the PL-RMTg pathway during cue- or cocaine-induced reinstatement. Male Sprague Dawley rats self-administered cocaine during daily 2-h sessions for ≥10 days and then underwent extinction training. Reinstatement of extinguished cocaine seeking was elicited by cocaine-associated cues or cocaine prime. Prior to reinstatement, rats received microinjections of the GABA agonists baclofen/muscimol (1/0.1 mM) into unilateral PL and the AMPA receptor antagonist NBQX (1 mM) into contralateral or ipsilateral RMTg. Functional disconnection of PL-RMTg via contralateral inactivation markedly increased cue-induced reinstatement, but did not increase cocaine-induced reinstatement or drive reinstatement of extinguished cocaine seeking in the absence of cues or cocaine. Enhanced cue-induced reinstatement was also observed with ipsilateral inactivation of PL and RMTg, but not with unilateral inactivation of PL or RMTg alone, indicating that both ipsilateral and contralateral projections from PL to RMTg have an inhibitory influence on behavior. These data further support a suppressive role for PL in cocaine seeking by implicating PL efferent projections to RMTg in inhibiting cue-induced reinstatement.
Collapse
|
106
|
Contó MB, Dos Santos NB, Munhoz CD, Marcourakis T, D'Almeida V, Camarini R. Exposure to Running Wheels Prevents Ethanol Rewarding Effects: The Role of CREB and Deacetylases SIRT-1 and SIRT-2 in the Nucleus Accumbens and Prefrontal Cortex. Neuroscience 2021; 469:125-137. [PMID: 34175423 DOI: 10.1016/j.neuroscience.2021.06.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 10/21/2022]
Abstract
Alcohol use disorder is one of the most prevalent addictions, strongly influenced by environmental factors. Voluntary physical activity (VPA) has proven to be intrinsically reinforcing and we hypothesized that, as a non-drug reinforcer, VPA could mitigate ethanol-induced rewarding effects. The transcriptional factor cAMP response element binding protein (CREB), and deacetylases isozymes sirtuins 1 and 2 (SIRT-1 and SIRT-2) have a complex interplay and both play a role in the rewarding effects of ethanol. To test whether the exposure of mice to running wheels inhibits the development of ethanol-induced conditioned place preference (CPP), mice were assigned into four groups: housed in home cages with locked ("Sedentary") or unlocked running wheels (VPA), and treated with saline or 1.8 g/kg ethanol during the conditioning phase. The groups were referred as Saline-Sedentary, Saline-VPA, Ethanol-Sedentary and Ethanol-VPA. The expression of CREB, SIRT-1 and SIRT-2 were evaluated in the prefrontal cortex (PFC) and nucleus accumbens (NAc). VPA prevented the development of ethanol-induced CPP. VPA, ethanol and the combination of both inhibited pCREB and pCREB/CREB ratio in the NAc, suggesting that both reward stimuli can share similar patterns of CREB activation. However, we have found that ethanol-induced increased CREB levels were prevented by VPA. Both VPA groups presented lower SIRT-1 levels in the NAc compared to the Sedentary groups. Thus, exposure to running wheels prevented ethanol-rewarding effects and ethanol-induced increases in CREB in the NAc. The molecular alterations underlying CPP prevention may be related to a lower expression of CREB in the NAc of Ethanol-VPA compared to the respective Sedentary group, given the positive correlation between CPP and CREB levels in the Ethanol-Sedentary group.
Collapse
Affiliation(s)
- Marcos Brandão Contó
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de Sao Paulo, Sao Paulo, Brazil.
| | - Nilton Barreto Dos Santos
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Carolina Demarchi Munhoz
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Vânia D'Almeida
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina (UNIFESP/EPM), Sao Paulo, Brazil
| | - Rosana Camarini
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
107
|
Michaels TI, Stone E, Singal S, Novakovic V, Barkin RL, Barkin S. Brain reward circuitry: The overlapping neurobiology of trauma and substance use disorders. World J Psychiatry 2021; 11:222-231. [PMID: 34168969 PMCID: PMC8209534 DOI: 10.5498/wjp.v11.i6.222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/14/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Mental health symptoms secondary to trauma exposure and substance use disorders (SUDs) co-occur frequently in both clinical and community samples. The possibility of a shared aetiology remains an important question in translational neuroscience. Advancements in genetics, basic science, and neuroimaging have led to an improved understanding of the neural basis of these disorders, their frequent comorbidity and high rates of relapse remain a clinical challenge. This project aimed to conduct a review of the field's current understanding regarding the neural circuitry underlying posttraumatic stress disorder and SUD. A comprehensive review was conducted of available published literature regarding the shared neurobiology of these disorders, and is summarized in detail, including evidence from both animal and clinical studies. Upon summarizing the relevant literature, this review puts forth a hypothesis related to their shared neurobiology within the context of fear processing and reward cues. It provides an overview of brain reward circuitry and its relation to the neurobiology, symptomology, and phenomenology of trauma and substance use. This review provides clinical insights and implications of the proposed theory, including the potential development of novel pharmacological and therapeutic treatments to address this shared neurobiology. Limitations and extensions of this theory are discussed to provide future directions and insights for this shared phenomena.
Collapse
Affiliation(s)
- Timothy I Michaels
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, United States
- Department of Psychiatry, The Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, NY 11004, United States
| | - Emily Stone
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, United States
| | - Sonali Singal
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, United States
| | - Vladan Novakovic
- Department of Psychiatry, The Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, NY 11004, United States
| | - Robert L Barkin
- Department of Anesthesiology, Rush University Medical College, Chicago, IL 60612, United States
| | - Stacy Barkin
- Private Practice, Scottsdale, AZ 85250, United States
| |
Collapse
|
108
|
Wang YC, Chiu WC, Cheng CN, Lee C, Chih Wei Huang A. Examination of neuroinflammatory cytokine interleukin-1 beta expression in the medial prefrontal cortex, amygdala, and hippocampus for the paradoxical effects of reward and aversion induced by morphine. Neurosci Lett 2021; 760:136076. [PMID: 34153368 DOI: 10.1016/j.neulet.2021.136076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 10/21/2022]
Abstract
A growing body of evidence has shown that abused drugs could simultaneously induce the paradoxical effect-reward and aversion. Moreover, the medial prefrontal cortex (mPFC), amygdala, and hippocampus were involved in this paradoxical effect by abused drugs. However, no research examined whether neuroinflammatory changes in the mPFC [including cingulate cortex area 1 (Cg1); prelimbic cortex (PrL); infralimbic cortex (IL)], basolateral amygdala, and hippocampus [e.g., CA1, CA2, CA3, and dentate gyrus (DG)] after morphine-induced reward in conditioned place preference (CPP) and aversion in conditioned taste aversion (CTA). The results showed that after morphine administration, the consumption of a 0.1% saccharin solution decreased; the mean time spent in the morphine-paired side compartment of the CPP box increased, indicating that morphine simultaneously induced the paradoxical effects of reward and aversion. The PrL and IL of the mPFC, the BLA of the amygdala, the CA1, CA2, CA3, and DG of the hippocampus but not the Cg1 presented hyperactive IL-1β expression in response to morphine's aversion and reward. The mPFC, amygdala, and hippocampus may appear neuroinflammation activity following morphine-induced paradoxical effect-reward in CPP and aversion in CTA. The present data may provide a better understanding of the relationship between neuroinflammation and morphine addiction.
Collapse
Affiliation(s)
- Ying-Chou Wang
- Department of Clinical Psychology, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Wei-Che Chiu
- Department of Psychiatry, Cathay General Hospital, Taipei, Taiwan; School of Medicine, Fu Jen Catholic University, Taipei, Taiwan
| | - Cai-N Cheng
- Department of Psychology, Fo Guang University, Yilan County 26247, Taiwan; Department of Life Sciences, National Central University, Jhong-Li District, Taoyuan City 32001, Taiwan
| | - Chiang Lee
- Department of Psychology, Fo Guang University, Yilan County 26247, Taiwan
| | | |
Collapse
|
109
|
Nall RW, Heinsbroek JA, Nentwig TB, Kalivas PW, Bobadilla AC. Circuit selectivity in drug versus natural reward seeking behaviors. J Neurochem 2021; 157:1450-1472. [PMID: 33420731 PMCID: PMC8178159 DOI: 10.1111/jnc.15297] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/16/2020] [Accepted: 01/03/2021] [Indexed: 12/23/2022]
Abstract
Substance use disorder (SUD) is characterized, in part by behavior biased toward drug use and away from natural sources of reward (e.g., social interaction, food, sex). The neurobiological underpinnings of SUDs reveal distinct brain regions where neuronal activity is necessary for the manifestation of SUD-characteristic behaviors. Studies that specifically examine how these regions are involved in behaviors motivated by drug versus natural reward allow determinations of which regions are necessary for regulating seeking of both reward types, and appraisals of novel SUD therapies for off-target effects on behaviors motivated by natural reward. Here, we evaluate studies directly comparing regulatory roles for specific brain regions in drug versus natural reward. While it is clear that many regions drive behaviors motivated by all reward types, based on the literature reviewed we propose a set of interconnected regions that become necessary for behaviors motivated by drug, but not natural rewards. The circuitry is selectively necessary for drug seeking includes an Action/Reward subcircuit, comprising nucleus accumbens, ventral pallidum, and ventral tegmental area, a Prefrontal subcircuit comprising prelimbic, infralimbic, and insular cortices, a Stress subcircuit comprising the central nucleus of the amygdala and the bed nucleus of the stria terminalis, and a Diencephalon circuit including lateral hypothalamus. Evidence was mixed for nucleus accumbens shell, insular cortex, and ventral pallidum. Studies for all other brain nuclei reviewed supported a necessary role in regulating both drug and natural reward seeking. Finally, we discuss emerging strategies to further disambiguate the necessity of brain regions in drug- versus natural reward-associated behaviors.
Collapse
Affiliation(s)
- Rusty W. Nall
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jasper A. Heinsbroek
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Todd B. Nentwig
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- These authors share senior authorship
| | - Ana-Clara Bobadilla
- School of Pharmacy, University of Wyoming, Laramie, WY, USA
- These authors share senior authorship
| |
Collapse
|
110
|
Kane L, Venniro M, Quintana‐Feliciano R, Madangopal R, Rubio FJ, Bossert JM, Caprioli D, Shaham Y, Hope BT, Warren BL. Fos-expressing neuronal ensemble in rat ventromedial prefrontal cortex encodes cocaine seeking but not food seeking in rats. Addict Biol 2021; 26:e12943. [PMID: 32683756 DOI: 10.1111/adb.12943] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/02/2020] [Accepted: 07/07/2020] [Indexed: 12/30/2022]
Abstract
Neuronal ensembles in ventromedial prefrontal cortex (vmPFC) play a role in both cocaine and palatable food seeking. However, it is unknown whether similar or different vmPFC neuronal ensembles mediate food and cocaine seeking. Here, we used the Daun02 inactivation procedure to assess whether the neuronal ensembles mediating food and cocaine seeking can be functionally distinguished. We trained male and female Fos-LacZ rats to self-administer palatable food pellets and cocaine on alternating days for 18 days. We then exposed the rats to a brief nonreinforced food- or cocaine-seeking test to induce Fos and β-gal in neuronal ensembles associated with food or cocaine seeking, respectively and infused Daun02 into vmPFC to ablate the β-gal-expressing ensembles. Two days later, we tested the rats for food or cocaine seeking under extinction conditions. Although inactivation of the food-seeking ensemble did not influence food or cocaine seeking, inactivation of the cocaine-seeking ensemble reduced cocaine seeking but not food seeking. Results indicate that the neuronal ensemble activated by cocaine seeking in vmPFC is functionally separate from the ensemble activated by food seeking.
Collapse
Affiliation(s)
- Louisa Kane
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | - Marco Venniro
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | - Richard Quintana‐Feliciano
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
- Department of Pharmacodynamics University of Florida Gainesville Florida USA
| | | | - F. Javier Rubio
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | | | - Daniele Caprioli
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia) Rome Italy
- Department of Physiology and Pharmacology Sapienza University of Rome Rome Italy
| | - Yavin Shaham
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | - Bruce T. Hope
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | - Brandon L. Warren
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
- Department of Pharmacodynamics University of Florida Gainesville Florida USA
| |
Collapse
|
111
|
Le Merre P, Ährlund-Richter S, Carlén M. The mouse prefrontal cortex: Unity in diversity. Neuron 2021; 109:1925-1944. [PMID: 33894133 DOI: 10.1016/j.neuron.2021.03.035] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/28/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022]
Abstract
The prefrontal cortex (PFC) is considered to constitute the highest stage of neural integration and to be devoted to representation and production of actions. Studies in primates have laid the foundation for theories regarding the principles of prefrontal function and provided mechanistic insights. The recent surge of studies of the PFC in mice holds promise for evolvement of present theories and development of novel concepts, particularly regarding principles shared across mammals. Here we review recent empirical work on the mouse PFC capitalizing on the experimental toolbox currently privileged to studies in this species. We conclude that this line of research has revealed cellular and structural distinctions of the PFC and neuronal activity with direct relevance to theories regarding the functions of the PFC. We foresee that data-rich mouse studies will be key to shed light on the general prefrontal architecture and mechanisms underlying cognitive aspects of organized actions.
Collapse
Affiliation(s)
- Pierre Le Merre
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Marie Carlén
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden.
| |
Collapse
|
112
|
Lothmann K, Amunts K, Herold C. The Neurotransmitter Receptor Architecture of the Mouse Olfactory System. Front Neuroanat 2021; 15:632549. [PMID: 33967704 PMCID: PMC8102831 DOI: 10.3389/fnana.2021.632549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/03/2021] [Indexed: 11/13/2022] Open
Abstract
The uptake, transmission and processing of sensory olfactory information is modulated by inhibitory and excitatory receptors in the olfactory system. Previous studies have focused on the function of individual receptors in distinct brain areas, but the receptor architecture of the whole system remains unclear. Here, we analyzed the receptor profiles of the whole olfactory system of adult male mice. We examined the distribution patterns of glutamatergic (AMPA, kainate, mGlu2/3, and NMDA), GABAergic (GABAA, GABAA(BZ), and GABAB), dopaminergic (D1/5) and noradrenergic (α1 and α2) neurotransmitter receptors by quantitative in vitro receptor autoradiography combined with an analysis of the cyto- and myelo-architecture. We observed that each subarea of the olfactory system is characterized by individual densities of distinct neurotransmitter receptor types, leading to a region- and layer-specific receptor profile. Thereby, the investigated receptors in the respective areas and strata showed a heterogeneous expression. Generally, we detected high densities of mGlu2/3Rs, GABAA(BZ)Rs and GABABRs. Noradrenergic receptors revealed a highly heterogenic distribution, while the dopaminergic receptor D1/5 displayed low concentrations, except in the olfactory tubercle and the dorsal endopiriform nucleus. The similarities and dissimilarities of the area-specific multireceptor profiles were analyzed by a hierarchical cluster analysis. A three-cluster solution was found that divided the areas into the (1) olfactory relay stations (main and accessory olfactory bulb), (2) the olfactory cortex (anterior olfactory cortex, dorsal peduncular cortex, taenia tecta, piriform cortex, endopiriform nucleus, entorhinal cortex, orbitofrontal cortex) and the (3) olfactory tubercle, constituting its own cluster. The multimodal receptor-architectonic analysis of each component of the olfactory system provides new insights into its neurochemical organization and future possibilities for pharmaceutic targeting.
Collapse
Affiliation(s)
- Kimberley Lothmann
- C. & O. Vogt-Institute of Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Katrin Amunts
- C. & O. Vogt-Institute of Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.,Institute of Neuroscience and Medicine INM-1, Research Centre Jülich, Jülich, Germany
| | - Christina Herold
- C. & O. Vogt-Institute of Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
113
|
Oswald LM, Dunn KE, Seminowicz DA, Storr CL. Early Life Stress and Risks for Opioid Misuse: Review of Data Supporting Neurobiological Underpinnings. J Pers Med 2021; 11:315. [PMID: 33921642 PMCID: PMC8072718 DOI: 10.3390/jpm11040315] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 01/02/2023] Open
Abstract
A robust body of research has shown that traumatic experiences occurring during critical developmental periods of childhood when neuronal plasticity is high increase risks for a spectrum of physical and mental health problems in adulthood, including substance use disorders. However, until recently, relatively few studies had specifically examined the relationships between early life stress (ELS) and opioid use disorder (OUD). Associations with opioid use initiation, injection drug use, overdose, and poor treatment outcome have now been demonstrated. In rodents, ELS has also been shown to increase the euphoric and decrease antinociceptive effects of opioids, but little is known about these processes in humans or about the neurobiological mechanisms that may underlie these relationships. This review aims to establish a theoretical model that highlights the mechanisms by which ELS may alter opioid sensitivity, thereby contributing to future risks for OUD. Alterations induced by ELS in mesocorticolimbic brain circuits, and endogenous opioid and dopamine neurotransmitter systems are described. The limited but provocative evidence linking these alterations with opioid sensitivity and risks for OUD is presented. Overall, the findings suggest that better understanding of these mechanisms holds promise for reducing vulnerability, improving prevention strategies, and prescribing guidelines for high-risk individuals.
Collapse
Affiliation(s)
- Lynn M. Oswald
- Department of Family and Community Health, University of Maryland School of Nursing, Baltimore, MD 21201, USA;
| | - Kelly E. Dunn
- Behavioral Pharmacology Research Unit, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21230, USA;
| | - David A. Seminowicz
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA;
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA
| | - Carla L. Storr
- Department of Family and Community Health, University of Maryland School of Nursing, Baltimore, MD 21201, USA;
| |
Collapse
|
114
|
Bouton ME, Maren S, McNally GP. BEHAVIORAL AND NEUROBIOLOGICAL MECHANISMS OF PAVLOVIAN AND INSTRUMENTAL EXTINCTION LEARNING. Physiol Rev 2021; 101:611-681. [PMID: 32970967 PMCID: PMC8428921 DOI: 10.1152/physrev.00016.2020] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This article reviews the behavioral neuroscience of extinction, the phenomenon in which a behavior that has been acquired through Pavlovian or instrumental (operant) learning decreases in strength when the outcome that reinforced it is removed. Behavioral research indicates that neither Pavlovian nor operant extinction depends substantially on erasure of the original learning but instead depends on new inhibitory learning that is primarily expressed in the context in which it is learned, as exemplified by the renewal effect. Although the nature of the inhibition may differ in Pavlovian and operant extinction, in either case the decline in responding may depend on both generalization decrement and the correction of prediction error. At the neural level, Pavlovian extinction requires a tripartite neural circuit involving the amygdala, prefrontal cortex, and hippocampus. Synaptic plasticity in the amygdala is essential for extinction learning, and prefrontal cortical inhibition of amygdala neurons encoding fear memories is involved in extinction retrieval. Hippocampal-prefrontal circuits mediate fear relapse phenomena, including renewal. Instrumental extinction involves distinct ensembles in corticostriatal, striatopallidal, and striatohypothalamic circuits as well as their thalamic returns for inhibitory (extinction) and excitatory (renewal and other relapse phenomena) control over operant responding. The field has made significant progress in recent decades, although a fully integrated biobehavioral understanding still awaits.
Collapse
Affiliation(s)
- Mark E Bouton
- Department of Psychological Science, University of Vermont, Burlington, Vermont
| | - Stephen Maren
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, Texas
| | - Gavan P McNally
- School of Psychology, University of New South Wales, Sydney, Australia
| |
Collapse
|
115
|
Rao-Ruiz P, Visser E, Mitrić M, Smit AB, van den Oever MC. A Synaptic Framework for the Persistence of Memory Engrams. Front Synaptic Neurosci 2021; 13:661476. [PMID: 33841124 PMCID: PMC8024575 DOI: 10.3389/fnsyn.2021.661476] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/26/2021] [Indexed: 12/31/2022] Open
Abstract
The ability to store and retrieve learned information over prolonged periods of time is an essential and intriguing property of the brain. Insight into the neurobiological mechanisms that underlie memory consolidation is of utmost importance for our understanding of memory persistence and how this is affected in memory disorders. Recent evidence indicates that a given memory is encoded by sparsely distributed neurons that become highly activated during learning, so-called engram cells. Research by us and others confirms the persistent nature of cortical engram cells by showing that these neurons are required for memory expression up to at least 1 month after they were activated during learning. Strengthened synaptic connectivity between engram cells is thought to ensure reactivation of the engram cell network during retrieval. However, given the continuous integration of new information into existing neuronal circuits and the relatively rapid turnover rate of synaptic proteins, it is unclear whether a lasting learning-induced increase in synaptic connectivity is mediated by stable synapses or by continuous dynamic turnover of synapses of the engram cell network. Here, we first discuss evidence for the persistence of engram cells and memory-relevant adaptations in synaptic plasticity, and then propose models of synaptic adaptations and molecular mechanisms that may support memory persistence through the maintenance of enhanced synaptic connectivity within an engram cell network.
Collapse
Affiliation(s)
- Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther Visser
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Miodrag Mitrić
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
116
|
Pastor V, Medina JH. Medial prefrontal cortical control of reward- and aversion-based behavioral output: Bottom-up modulation. Eur J Neurosci 2021; 53:3039-3062. [PMID: 33660363 DOI: 10.1111/ejn.15168] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 12/22/2022]
Abstract
How does the brain guide our actions? This is a complex issue, where the medial prefrontal cortex (mPFC) plays a crucial role. The mPFC is essential for cognitive flexibility and decision making. These functions are related to reward- and aversion-based learning, which ultimately drive behavior. Though, cortical projections and modulatory systems that may regulate those processes in the mPFC are less understood. How does the mPFC regulate approach-avoidance behavior in the case of conflicting aversive and appetitive stimuli? This is likely dependent on the bottom-up neuromodulation of the mPFC projection neurons. In this review, we integrate behavioral-, pharmacological-, and viral-based circuit manipulation data showing the involvement of mPFC dopaminergic, noradrenergic, cholinergic, and serotoninergic inputs in reward and aversion processing. Given that an incorrect balance of reward and aversion value could be a key problem in mental diseases such as substance use disorders, we discuss outstanding questions for future research on the role of mPFC modulation in reward and aversion.
Collapse
Affiliation(s)
- Verónica Pastor
- CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis" (IBCN), Buenos Aires, Argentina.,Universidad de Buenos Aires, Facultad de Medicina, Departamento de Ciencias Fisiológicas, Buenos Aires, Argentina
| | - Jorge Horacio Medina
- CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis" (IBCN), Buenos Aires, Argentina.,Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
| |
Collapse
|
117
|
Opposing Regulation of Cocaine Seeking by Glutamate and GABA Neurons in the Ventral Pallidum. Cell Rep 2021; 30:2018-2027.e3. [PMID: 32049028 PMCID: PMC7045305 DOI: 10.1016/j.celrep.2020.01.023] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/11/2019] [Accepted: 01/07/2020] [Indexed: 11/21/2022] Open
Abstract
Projections from the nucleus accumbens to the ventral pallidum (VP) regulate relapse in animal models of addiction. The VP contains GABAergic (VPGABA) and glutamatergic (VPGlu) neurons, and a subpopulation of GABAergic neurons co-express enkephalin (VPPenk). Rabies tracing reveals that VPGlu and VPPenk neurons receive preferential innervation from upstream D1- relative to D2-expressing accumbens neurons. Chemogenetic stimulation of VPGlu neurons inhibits, whereas stimulation of VPGABA and VPPenk neurons potentiates cocaine seeking in mice withdrawn from intravenous cocaine self-administration. Calcium imaging reveals cell type-specific activity patterns when animals learn to suppress drug seeking during extinction training versus engaging in cue-induced cocaine seeking. During cued seeking, VPGABA neurons increase their overall activity, and VPPenk neurons are selectively activated around nose pokes for cocaine. In contrast, VPGlu neurons increase their spike rate following extinction training. These data show that VP subpopulations differentially encode and regulate cocaine seeking, with VPPenk and VPGABA neurons facilitating and VPGlu neurons inhibiting cocaine seeking. Heinsbroek et al. show that glutamate and GABA neurons in ventral pallidum differentially regulate cued cocaine seeking. Calcium activity in glutamate neurons increases when mice refrain from cocaine seeking. Activating glutamate neurons inhibits cocaine seeking. Calcium activity increases in GABA neurons during cocaine seeking, and activating GABA or enkephalin neurons induces cocaine seeking.
Collapse
|
118
|
BDNF Protein and BDNF mRNA Expression of the Medial Prefrontal Cortex, Amygdala, and Hippocampus during Situational Reminder in the PTSD Animal Model. Behav Neurol 2021; 2021:6657716. [PMID: 33763156 PMCID: PMC7964114 DOI: 10.1155/2021/6657716] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 11/17/2022] Open
Abstract
Whether BDNF protein and BDNF mRNA expression of the medial prefrontal cortex (mPFC; cingulated cortex area 1 (Cg1), prelimbic cortex (PrL), and infralimbic cortex (IL)), amygdala, and hippocampus (CA1, CA2, CA3, and dentate gyrus (DG)) was involved in fear of posttraumatic stress disorder (PTSD) during the situational reminder of traumatic memory remains uncertain. Footshock rats experienced an inescapable footshock (3 mA, 10 s), and later we have measured fear behavior for 2 min in the footshock environment on the situational reminder phase. In the final retrieval of situational reminder, BDNF protein and mRNA levels were measured. The results showed that higher BDNF expression occurred in the Cg1, PrL, and amygdala. Lower BDNF expression occurred in the IL, CA1, CA2, CA3, and DG. BDNF mRNA levels were higher in the mPFC and amygdala but lower in the hippocampus. The neural connection analysis showed that BDNF protein and BDNF mRNA exhibited weak connections among the mPFC, amygdala, and hippocampus during situational reminders. The present data did not support the previous viewpoint in neuroimaging research that the mPFC and hippocampus revealed hypoactivity and the amygdala exhibited hyperactivity for PTSD symptoms. These findings should be discussed with the previous evidence and provide clinical implications for PTSD.
Collapse
|
119
|
Caffino L, Mottarlini F, Van Reijmersdal B, Telese F, Verheij MM, Fumagalli F, Homberg JR. The role of the serotonin transporter in prefrontal cortex glutamatergic signaling following short- and long-access cocaine self-administration. Addict Biol 2021; 26:e12896. [PMID: 32187792 PMCID: PMC7988536 DOI: 10.1111/adb.12896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/28/2020] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
Vulnerability to drug addiction relies on substantial individual differences. We previously demonstrated that serotonin transporter knockout (SERT−/−) rats show increased cocaine intake and develop signs of compulsivity. However, the underlying neural mechanisms are not fully understood. Given the pivotal role of glutamate and prefrontal cortex in cocaine‐seeking behavior, we sought to investigate the expression of proteins implicated in glutamate neurotransmission in the prefrontal cortex of naïve and cocaine‐exposed rats lacking SERT. We focused on the infralimbic (ILc) and prelimbic (PLc) cortices, which are theorized to exert opposing effects on the control over subcortical brain areas. SERT−/− rats, which compared to wild‐type (SERT+/+) rats show increased ShA and LgA intake short‐access (ShA) and long‐access (LgA) cocaine intake, were sacrificed 24 h into withdrawal for ex vivo molecular analyses. In the ILc homogenate of SERT−/− rats, we observed a sharp increase in glial glutamate transporter 1 (GLT‐1) after ShA, but not LgA, cocaine intake. This was paralleled by ShA‐induced increases in GluN1, GluN2A, and GluN2B NMDA receptor subunits and their scaffolding protein SAP102 in the ILc homogenate, but not postsynaptic density, of these knockout animals. In the PLc, we found no major changes in the homogenate; conversely, the expression of GluN1 and GluN2A NMDA receptor subunits was increased in the postsynaptic density under ShA conditions and reduced under LgA conditions. These results point to SERT as a critical regulator of glutamate homeostasis in a way that differs between the subregions investigated, the duration of cocaine exposure as well as the cellular compartment analyzed.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Boyd Van Reijmersdal
- Department of Cognitive Neuroscience, Division of Molecular Neurogenetics, Donders Institute for Brain, Cognition and Behaviour Radboud University Nijmegen Medical Centre Nijmegen The Netherlands
| | - Francesca Telese
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Michel M.M. Verheij
- Department of Cognitive Neuroscience, Division of Molecular Neurogenetics, Donders Institute for Brain, Cognition and Behaviour Radboud University Nijmegen Medical Centre Nijmegen The Netherlands
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Division of Molecular Neurogenetics, Donders Institute for Brain, Cognition and Behaviour Radboud University Nijmegen Medical Centre Nijmegen The Netherlands
| |
Collapse
|
120
|
Abstract
Abstract
Purpose of Review
Current theories of alcohol use disorders (AUD) highlight the importance of Pavlovian and instrumental learning processes mainly based on preclinical animal studies. Here, we summarize available evidence for alterations of those processes in human participants with AUD with a focus on habitual versus goal-directed instrumental learning, Pavlovian conditioning, and Pavlovian-to-instrumental transfer (PIT) paradigms.
Recent Findings
The balance between habitual and goal-directed control in AUD participants has been studied using outcome devaluation or sequential decision-making procedures, which have found some evidence of reduced goal-directed/model-based control, but little evidence for stronger habitual responding. The employed Pavlovian learning and PIT paradigms have shown considerable differences regarding experimental procedures, e.g., alcohol-related or conventional reinforcers or stimuli.
Summary
While studies of basic learning processes in human participants with AUD support a role of Pavlovian and instrumental learning mechanisms in the development and maintenance of drug addiction, current studies are characterized by large variability regarding methodology, sample characteristics, and results, and translation from animal paradigms to human research remains challenging. Longitudinal approaches with reliable and ecologically valid paradigms of Pavlovian and instrumental processes, including alcohol-related cues and outcomes, are warranted and should be combined with state-of-the-art imaging techniques, computational approaches, and ecological momentary assessment methods.
Collapse
|
121
|
Prefrontal cortex nicotinic receptor inhibition by methyllycaconitine impaired cocaine-associated memory acquisition and retrieval. Behav Brain Res 2021; 406:113212. [PMID: 33657437 DOI: 10.1016/j.bbr.2021.113212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/15/2022]
Abstract
Cocaine administration has been shown to induce plastic changes in the medial prefrontal cortex (mPFC), which could represent a mechanism by which cocaine facilitates the association between cocaine rewarding effects with contextual cues. Nicotinic acetylcholine receptors (nAChRs) in the mPFC have critical roles in cognitive function including attention and memory and are key players in plasticity processes. However, whether nAChRs in the mPFC are required for the acquisition and maintenance of cocaine-associated memories is still unknown. To assess this question, we used the conditioning place preference (CPP) model to study the effect of intra-mPFC infusion of methyllycaconitine, a selective antagonist of α7 nAChRs, on the acquisition, consolidation and expression of cocaine-associated memory in adult rats. Our findings reveal that mPFC α7 nAChRs activation is necessary for the acquisition and retrieval, but not consolidation, of cocaine induced CPP. Moreover, cocaine-induced sensitization during CPP conditioning sessions was abolished by methyllycaconitine infusion in the mPFC. Together, these results identify mPFC α7 nAChRs as critical players involved in both acquiring and retrieving cocaine-associated memories. Considering that drug seeking often depends on the association between drug-paired cues and the rewarding effects of the drug, α7 nAChRs in the mPFC could be considered as potential targets for the prevention or treatment of cocaine use disorder.
Collapse
|
122
|
Barbier E, Barchiesi R, Domi A, Chanthongdee K, Domi E, Augier G, Augier E, Xu L, Adermark L, Heilig M. Downregulation of Synaptotagmin 1 in the Prelimbic Cortex Drives Alcohol-Associated Behaviors in Rats. Biol Psychiatry 2021; 89:398-406. [PMID: 33160605 DOI: 10.1016/j.biopsych.2020.08.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/17/2020] [Accepted: 08/25/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Alcohol addiction is characterized by persistent neuroadaptations in brain structures involved in motivation, emotion, and decision making, including the medial prefrontal cortex, the nucleus accumbens, and the amygdala. We previously reported that induction of alcohol dependence was associated with long-term changes in the expression of genes involved in neurotransmitter release. Specifically, Syt1, which plays a key role in neurotransmitter release and neuronal functions, was downregulated. Here, we therefore examined the role of Syt1 in alcohol-associated behaviors in rats. METHODS We evaluated the effect of Syt1 downregulation using an adeno-associated virus (AAV) containing a short hairpin RNA against Syt1. Cre-dependent Syt1 was also used in combination with an rAAV2 retro-Cre virus to assess circuit-specific effects of Syt1 knockdown (KD). RESULTS Alcohol-induced downregulation of Syt1 is specific to the prelimbic cortex (PL), and KD of Syt1 in the PL resulted in escalated alcohol consumption, increased motivation to consume alcohol, and increased alcohol drinking despite negative consequences ("compulsivity"). Syt1 KD in the PL altered the excitation/inhibition balance in the basolateral amygdala, while the nucleus accumbens core was unaffected. Accordingly, a projection-specific Syt1 KD in the PL-basolateral amygdala projection was sufficient to increase compulsive alcohol drinking, while a KD of Syt1 restricted to PL-nucleus accumbens core projecting neurons had no effect on tested alcohol-related behaviors. CONCLUSIONS Together, these data suggest that dysregulation of Syt1 is an important mechanism in long-term neuroadaptations observed after a history of alcohol dependence, and that Syt1 regulates alcohol-related behaviors in part by affecting a PL-basolateral amygdala brain circuit.
Collapse
Affiliation(s)
- Estelle Barbier
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Riccardo Barchiesi
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ana Domi
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kanat Chanthongdee
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Physiology, Faculty of Medicine Siraj Hospital, Mahidol University, Bangkok, Thailand
| | - Esi Domi
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Gaelle Augier
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Eric Augier
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Li Xu
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Psychosomatic Medicine Center, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Markus Heilig
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
123
|
Coley AA, Padilla-Coreano N, Patel R, Tye KM. Valence processing in the PFC: Reconciling circuit-level and systems-level views. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 158:171-212. [PMID: 33785145 DOI: 10.1016/bs.irn.2020.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An essential component in animal behavior is the ability to process emotion and dissociate among positive and negative valence in response to a rewarding or aversive stimulus. The medial prefrontal cortex (mPFC)-responsible for higher order executive functions that include cognition, learning, and working memory; and is also involved in sociability-plays a major role in emotional processing and control. Although the amygdala is widely regarded as the "emotional hub," the mPFC encodes for context-specific salience and elicits top-down control over limbic circuitry. The mPFC can then conduct behavioral responses, via cortico-striatal and cortico-brainstem pathways, that correspond to emotional stimuli. Evidence shows that abnormalities within the mPFC lead to sociability deficits, working memory impairments, and drug-seeking behavior that include addiction and compulsive disorders; as well as conditions such as anhedonia. Recent studies investigate the effects of aberrant salience processing on cortical circuitry and neuronal populations associated with these behaviors. In this chapter, we discuss mPFC valence processing, neuroanatomical connections, and physiological substrates involved in mPFC-associated behavior. We review neurocomputational and theoretical models such as "mixed selectivity," that describe cognitive control, attentiveness, and motivational drives. Using this knowledge, we describe the effects of valence imbalances and its influence on mPFC neural pathways that contribute to deficits in social cognition, while understanding the effects in addiction/compulsive behaviors and anhedonia.
Collapse
Affiliation(s)
- Austin A Coley
- Salk Institute for Biological Studies, La Jolla, CA, United States
| | | | - Reesha Patel
- Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Kay M Tye
- Salk Institute for Biological Studies, La Jolla, CA, United States.
| |
Collapse
|
124
|
Impact of Acute and Persistent Excitation of Prelimbic Pyramidal Neurons on Motor Activity and Trace Fear Learning. J Neurosci 2021; 41:960-971. [PMID: 33402420 DOI: 10.1523/jneurosci.2606-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 11/21/2022] Open
Abstract
Drug-induced neuroadaptations in the mPFC have been implicated in addictive behaviors. Repeated cocaine exposure has been shown to increase pyramidal neuron excitability in the prelimbic (PL) region of the mouse mPFC, an adaptation attributable to a suppression of G protein-gated inwardly rectifying K+ (GIRK) channel activity. After establishing that this neuroadaptation is not seen in adjacent GABA neurons, we used viral GIRK channel ablation and complementary chemogenetic approaches to selectively enhance PL pyramidal neuron excitability in adult mice, to evaluate the impact of this form of plasticity on PL-dependent behaviors. GIRK channel ablation decreased somatodendritic GABAB receptor-dependent signaling and rheobase in PL pyramidal neurons. This manipulation also enhanced the motor-stimulatory effect of cocaine but did not impact baseline activity or trace fear learning. In contrast, selective chemogenetic excitation of PL pyramidal neurons, or chemogenetic inhibition of PL GABA neurons, increased baseline and cocaine-induced activity and disrupted trace fear learning. These effects were mirrored in male mice by selective excitation of PL pyramidal neurons projecting to the VTA, but not NAc or BLA. Collectively, these data show that manipulations enhancing the excitability of PL pyramidal neurons, and specifically those projecting to the VTA, recapitulate behavioral hallmarks of repeated cocaine exposure in mice.SIGNIFICANCE STATEMENT Prolonged exposure to drugs of abuse triggers neuroadaptations that promote core features of addiction. Understanding these neuroadaptations and their implications may suggest interventions capable of preventing or treating addiction. While previous work showed that repeated cocaine exposure increased the excitability of pyramidal neurons in the prelimbic cortex (PL), the behavioral implications of this neuroadaptation remained unclear. Here, we used neuron-specific manipulations to evaluate the impact of increased PL pyramidal neuron excitability on PL-dependent behaviors. Acute or persistent excitation of PL pyramidal neurons potentiated cocaine-induced motor activity and disrupted trace fear conditioning, effects replicated by selective excitation of the PL projection to the VTA. Our work suggests that hyperexcitability of this projection drives key behavioral hallmarks of addiction.
Collapse
|
125
|
Thompson BL, Oscar-Berman M, Kaplan GB. Opioid-induced structural and functional plasticity of medium-spiny neurons in the nucleus accumbens. Neurosci Biobehav Rev 2021; 120:417-430. [PMID: 33152423 PMCID: PMC7855607 DOI: 10.1016/j.neubiorev.2020.10.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022]
Abstract
Opioid Use Disorder (OUD) is a chronic relapsing clinical condition with tremendous morbidity and mortality that frequently persists, despite treatment, due to an individual's underlying psychological, neurobiological, and genetic vulnerabilities. Evidence suggests that these vulnerabilities may have neurochemical, cellular, and molecular bases. Key neuroplastic events within the mesocorticolimbic system that emerge through chronic exposure to opioids may have a determinative influence on behavioral symptoms associated with OUD. In particular, structural and functional alterations in the dendritic spines of medium spiny neurons (MSNs) within the nucleus accumbens (NAc) and its dopaminergic projections from the ventral tegmental area (VTA) are believed to facilitate these behavioral sequelae. Additionally, glutamatergic neurons from the prefrontal cortex, the basolateral amygdala, the hippocampus, and the thalamus project to these same MSNs, providing an enriched target for synaptic plasticity. Here, we review literature related to neuroadaptations in NAc MSNs from dopaminergic and glutamatergic pathways in OUD. We also describe new findings related to transcriptional, epigenetic, and molecular mechanisms in MSN plasticity in the different stages of OUD.
Collapse
Affiliation(s)
- Benjamin L Thompson
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA; Research Service, VA Boston Healthcare System, 150 South Huntington Avenue, Boston, MA 02130, USA.
| | - Marlene Oscar-Berman
- Research Service, VA Boston Healthcare System, 150 South Huntington Avenue, Boston, MA 02130, USA; Department of Anatomy & Neurobiology, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA; Department of Psychiatry, Boston University School of Medicine, 720 Harrison Avenue, Boston, MA, 02118, USA; Department of Neurology, Boston University School of Medicine, Boston University Medical Center, 80 East Concord Street, Boston, MA 02118, USA.
| | - Gary B Kaplan
- Department of Psychiatry, Boston University School of Medicine, 720 Harrison Avenue, Boston, MA, 02118, USA; Mental Health Service, VA Boston Healthcare System, 940 Belmont Street, Brockton, MA, 02301, USA; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA.
| |
Collapse
|
126
|
Muench C, Charlet K, Balderston NL, Grillon C, Heilig M, Cortes CR, Momenan R, Lohoff FW. Fear conditioning and extinction in alcohol dependence: Evidence for abnormal amygdala reactivity. Addict Biol 2021; 26:e12835. [PMID: 31702089 DOI: 10.1111/adb.12835] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/28/2019] [Accepted: 09/08/2019] [Indexed: 11/29/2022]
Abstract
Fear conditioning and extinction (FCE) are vital processes in adaptive emotion regulation and disrupted in anxiety disorders. Despite substantial comorbidity between alcohol dependence (ALC) and anxiety disorders and reports of altered negative emotion processing in ALC, neural correlates of FCE in this clinical population remain unknown. Here, we used a 2-day fear learning paradigm in 43 healthy participants and 43 individuals with ALC at the National Institutes of Health. Main outcomes of this multimodal study included structural and functional brain magnetic resonance imaging, clinical measures, as well as skin conductance responses (SCRs) to confirm differential conditioning. Successful FCE was demonstrated across participants by differential SCRs in the conditioning phase and no difference in SCRs to the conditioned stimuli in the extinction phase. The ALC group showed significantly reduced blood oxygenation level-dependent responses in the right amygdala during conditioning (Cohen's d = .89, P(FWE) = .037) and in the left amygdala during fear renewal (Cohen's d = .68, P(FWE) = .039). Right amygdala activation during conditioning was significantly correlated with ALC severity (r = .39, P(Bonferroni) = .009), depressive symptoms (r = .37, P(Bonferroni) = .015), trait anxiety (r = .41, P(Bonferroni) = .006), and perceived stress (r = .45, P(Bonferroni) = .002). Our data suggest that individuals with ALC have dysregulated fear learning, in particular, dysregulated neural activation patterns, in the amygdala. Furthermore, amygdala activation during fear conditioning was associated with ALC-related clinical measures. The FCE paradigm may be a promising tool to investigate structures involved in negative affect regulation, which might inform the development of novel treatment approaches for ALC.
Collapse
Affiliation(s)
- Christine Muench
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| | - Katrin Charlet
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| | - Nicholas L. Balderston
- Section on Neurobiology of Fear and Anxiety, National Institute of Mental Health National Institutes of Health Bethesda MD USA
| | - Christian Grillon
- Section on Neurobiology of Fear and Anxiety, National Institute of Mental Health National Institutes of Health Bethesda MD USA
| | - Markus Heilig
- Department of Clinical and Experimental Medicine Linköping University Linköping Sweden
| | - Carlos R. Cortes
- Clinical NeuroImaging Research Core, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| | - Reza Momenan
- Clinical NeuroImaging Research Core, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| | - Falk W. Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| |
Collapse
|
127
|
Granulocyte-Colony Stimulating Factor Reduces Cocaine-Seeking and Downregulates Glutamatergic Synaptic Proteins in Medial Prefrontal Cortex. J Neurosci 2020; 41:1553-1565. [PMID: 33361463 DOI: 10.1523/jneurosci.1452-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Psychostimulant use disorder is a major public health issue, and despite the scope of the problem there are currently no Food and Drug Administration (FDA)-approved treatments. There would be tremendous utility in development of a treatment that could help patients both achieve and maintain abstinence. Previous work from our group has identified granulocyte-colony stimulating factor (G-CSF) as a neuroactive cytokine that alters behavioral response to cocaine, increases synaptic dopamine release, and enhances cognitive flexibility. Here, we investigate the role of G-CSF in affecting extinction and reinstatement of cocaine-seeking and perform detailed characterization of its proteomic effects in multiple limbic substructures. Male Sprague Dawley rats were injected with PBS or G-CSF during (1) extinction or (2) abstinence from cocaine self-administration, and drug seeking behavior was measured. Quantitative assessment of changes in the proteomic landscape in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC) were performed via data-independent acquisition (DIA) mass spectrometry analysis. Administration of G-CSF during extinction accelerated the rate of extinction, and administration during abstinence attenuated cue-induced cocaine-seeking. Analysis of global protein expression demonstrated that G-CSF regulated proteins primarily in mPFC that are critical to glutamate signaling and synapse maintenance. Taken together, these findings support G-CSF as a viable translational research target with the potential to reduce drug craving or seeking behaviors. Importantly, recombinant G-CSF exists as an FDA-approved medication which may facilitate rapid clinical translation. Additionally, using cutting-edge multiregion discovery proteomics analyses, these studies identify a novel mechanism underlying G-CSF effects on behavioral plasticity.SIGNIFICANCE STATEMENT Pharmacological treatments for psychostimulant use disorder are desperately needed, especially given the disease's chronic, relapsing nature. However, there are currently no Food and Drug Administration (FDA)-approved pharmacotherapies. Emerging evidence suggests that targeting the immune system may be a viable translational research strategy; preclinical studies have found that the neuroactive cytokine granulocyte-colony stimulating factor (G-CSF) alters cocaine reward and reinforcement and can enhance cognitive flexibility. Given this basis of evidence we studied the effects of G-CSF treatment on extinction and reinstatement of cocaine seeking. We find that administration of G-CSF accelerates extinction and reduces cue-induced drug seeking after cocaine self-administration. In addition, G-CSF leads to downregulation of synaptic glutamatergic proteins in medial prefrontal cortex (mPFC), suggesting that G-CSF influences drug seeking via glutamatergic mechanisms.
Collapse
|
128
|
Kaminska B, Caballero JP, Moorman DE. Integration of value and action in medial prefrontal neural systems. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 158:57-82. [PMID: 33785156 DOI: 10.1016/bs.irn.2020.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The rodent medial prefrontal cortex (mPFC) plays a key role in regulating cognition, emotion, and behavior. mPFC neurons are activated in diverse experimental paradigms, raising the questions of whether there are specific task elements or dimensions encoded by mPFC neurons, and whether these encoded parameters are selective to neurons in particular mPFC subregions or networks. Here, we consider the role of mPFC neurons in processing appetitive and aversive cues, outcomes, and related behaviors. mPFC neurons are strongly activated in tasks probing value and outcome-associated actions, but these responses vary across experimental paradigms. Can we identify specific categories of responses (e.g., positive or negative value), or do mPFC neurons exhibit response properties that are too heterogeneous/complex to cluster into distinct conceptual groups? Based on a review of relevant studies, we consider what has been done and what needs to be further explored in order to address these questions.
Collapse
Affiliation(s)
- Beata Kaminska
- Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, United States
| | - Jessica P Caballero
- Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, United States
| | - David E Moorman
- Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, United States; Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, United States.
| |
Collapse
|
129
|
Basolateral Amygdala but Not Medial Prefrontal Cortex Contributes to Chronic Fluoxetine Treatments for PTSD Symptoms in Mice. Behav Neurol 2020; 2020:8875087. [PMID: 33299494 PMCID: PMC7710423 DOI: 10.1155/2020/8875087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/16/2020] [Accepted: 11/02/2020] [Indexed: 11/17/2022] Open
Abstract
Do chronic fluoxetine treatments reduced footshock-induced posttraumatic stress disorder (PTSD) symptoms, including fear and comorbid depression, in the situational reminder phase? Moreover, are the subareas of the medial prefrontal cortex (mPFC), including the cingulate cortex 1 (Cg1), prelimbic cortex (PrL), infralimbic cortex (IL), and basolateral amygdala (BLA), involved in the fluoxetine amelioration of PTSD symptoms? These two crucial issues were addressed in the present study. All mice were injected with chronic fluoxetine or normal saline treatments for the adaptation (14 days), footshock fear conditioning (1 day), and situational reminder (3 days) phases. After adaptation, the mice were subjected to footshock (2 mA, 10 seconds) or nonfootshock and stayed 2 min in a footshock box for 2 min for fear conditioning. Later, they were placed in the footshock box for 2 min in the situational reminder phase. In the final session of the situational reminder phase, a forced swimming test (FST) and immunohistochemical staining were conducted. The results indicated that footshock induced fear and comorbid depression. Meanwhile, chronic fluoxetine treatments reduced fear and depression behaviors. The Cg1, PrL, IL, and BLA were seemingly to increase c-Fos expression after footshock-induced PTSD symptoms in the situational reminder phase. The fluoxetine treatments reduced only the BLA's c-Fos expression. The findings suggest that BLA contributes to the fluoxetine amelioration of PTSD symptoms; however, the mPFC, including the Cg1, PrL, and IL, did not mediate PTSD symptoms' amelioration stemming from fluoxetine. The present data might help us to further understand the neural mechanism of fluoxetine treatments in PTSD symptoms.
Collapse
|
130
|
Vallerini GP, Cheng YH, Chase KA, Sharma RP, Kusumo H, Khakhkhar S, Feinstein DL, Guizzetti M, Gavin DP. Modulation of Poly ADP Ribose Polymerase (PARP) Levels and Activity by Alcohol Binge-Like Drinking in Male Mice. Neuroscience 2020; 448:1-13. [PMID: 32920042 DOI: 10.1016/j.neuroscience.2020.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 08/14/2020] [Accepted: 09/02/2020] [Indexed: 01/09/2023]
Abstract
Binge drinking is a frequent pattern of ethanol consumption within Alcohol Use Disorders (AUDs). Binge-like ethanol exposure increases Poly(ADP-ribose) polymerase (PARP) expression and activity. PARP enzymes have been implicated in addiction and serve multiple roles in the cell, including gene expression regulation. In this study, we examined the effects of binge-like alcohol consumption in the prefrontal cortex (PFC) of adult C57BL/6J male mice via a 4-day Drinking-in-the-Dark (DID) paradigm. The role of PARP in associated gene expression and behavioral changes was assessed by administering the PARP inhibitor ABT-888 on the last DID day. We then conducted an RNA-seq analysis of the PFC gene expression changes associated with DID-consumed ethanol or ABT-888 treatment. A separate cohort of mice was inoculated with an HSV-PARP1 vector in the PFC and subject to a DID experiment to verify whether overexpressed PARP1 increased ethanol drinking. We confirmed that alcohol increases Parp1 gene expression and PARP activity in the PFC. RNA-seq showed significantly altered expression of 41 genes by DID-consumed ethanol, and of 48 genes by ABT-888. These results were confirmed by qPCR in 7 of the 10 genes validated, 4 of which have been previously associated with addiction. ABT-888 reduced, and overexpression of PFC PARP1 increased DID ethanol consumption. In our model, alcohol binge drinking induced specific alterations in the PFC expression of genes potentially involved in addiction. Pharmacological PARP inhibition proved effective in reversing these changes and preventing further alcohol consumption. Our results suggest an involvement of ethanol-induced PARP1 in reinforcing binge-like addictive behavior.
Collapse
Affiliation(s)
- Gian Paolo Vallerini
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, United States; Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - You-Hong Cheng
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, United States; Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Kayla A Chase
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, United States; Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Rajiv P Sharma
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, United States; Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Handojo Kusumo
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, United States; Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Shivani Khakhkhar
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Douglas L Feinstein
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, United States; Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, 60612, United States
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States; VA Portland Health Care System, Portland, OR 97239, United States.
| | - David P Gavin
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, United States; Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States.
| |
Collapse
|
131
|
Carlson HN, Weiner JL. The neural, behavioral, and epidemiological underpinnings of comorbid alcohol use disorder and post-traumatic stress disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:69-142. [PMID: 33648676 DOI: 10.1016/bs.irn.2020.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alcohol use disorder (AUD) and (PTSD) frequently co-occur and individuals suffering from this dual diagnosis often exhibit increased symptom severity and poorer treatment outcomes than those with only one of these diseases. Although there have been significant advances in our understanding of the neurobiological mechanisms underlying each of these disorders, the neural underpinnings of the comorbid condition remain poorly understood. This chapter summarizes recent epidemiological findings on comorbid AUD and PTSD, with a focus on vulnerable populations, the temporal relationship between these disorders, and the clinical consequences associated with the dual diagnosis. We then review animal models of the comorbid condition and emerging human and non-human animal research that is beginning to identify maladaptive neural changes common to both disorders, primarily involving functional changes in brain reward and stress networks. We end by proposing a neural framework, based on the emerging field of affective valence encoding, that may better explain the epidemiological and neural findings on AUD and PTSD.
Collapse
Affiliation(s)
- Hannah N Carlson
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jeff L Weiner
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
132
|
Mattera A, Pagani M, Baldassarre G. A Computational Model Integrating Multiple Phenomena on Cued Fear Conditioning, Extinction, and Reinstatement. Front Syst Neurosci 2020; 14:569108. [PMID: 33132856 PMCID: PMC7550679 DOI: 10.3389/fnsys.2020.569108] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/13/2020] [Indexed: 11/23/2022] Open
Abstract
Conditioning, extinction, and reinstatement are fundamental learning processes of animal adaptation, also strongly involved in human pathologies such as post-traumatic stress disorder, anxiety, depression, and dependencies. Cued fear conditioning, extinction, restatement, and systematic manipulations of the underlying brain amygdala and medial prefrontal cortex, represent key experimental paradigms to study such processes. Numerous empirical studies have revealed several aspects and the neural systems and plasticity underlying them, but at the moment we lack a comprehensive view. Here we propose a computational model based on firing rate leaky units that contributes to such integration by accounting for 25 different experiments on fear conditioning, extinction, and restatement, on the basis of a single neural architecture having a structure and plasticity grounded in known brain biology. This allows the model to furnish three novel contributions to understand these open issues: (a) the functioning of the central and lateral amygdala system supporting conditioning; (b) the role played by the endocannabinoids system in within- and between-session extinction; (c) the formation of three important types of neurons underlying fear processing, namely fear, extinction, and persistent neurons. The model integration of the results on fear conditioning goes substantially beyond what was done in previous models.
Collapse
Affiliation(s)
- Andrea Mattera
- Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Marco Pagani
- Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Gianluca Baldassarre
- Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| |
Collapse
|
133
|
Zhang S, Zhornitsky S, Wang W, Dhingra I, Le TM, Li CSR. Cue-elicited functional connectivity of the periaqueductal gray and tonic cocaine craving. Drug Alcohol Depend 2020; 216:108240. [PMID: 32853997 PMCID: PMC7606798 DOI: 10.1016/j.drugalcdep.2020.108240] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Withdrawal from chronic cocaine use leads to anxiety and dysphoria that may perpetuate habitual drug use. The pain circuit is widely implicated in the processing and manifestations of negative emotions. Numerous studies have focused on characterizing reward circuit dysfunction but relatively little is known about the pain circuit response during cocaine withdrawal. METHODS Here we examined the activity and connectivity of the periaqueductal gray (PAG), a hub of the pain circuit, during cocaine cue exposure in 52 recently abstinent cocaine dependent participants (CD, 42 men). Imaging data were processed with published routines, and the results were evaluated at a corrected threshold. RESULTS CD showed higher activation of the PAG and connectivity of the PAG with the ventromedial prefrontal cortex (vmPFC) during cocaine as compared to neutral cue exposure. PAG-vmPFC connectivity was positively and negatively correlated with tonic cocaine craving, as assessed by the Cocaine Craving Questionnaire, in male and female CD, respectively, and the sex difference was confirmed by a slope test. Granger causality analyses showed that the PAG Granger caused vmPFC time series in men and the reverse was true in women, substantiating sex differences in the directional interactions of the PAG and vmPFC. CONCLUSION The findings provide the first evidence in humans implicating the PAG circuit in cocaine withdrawal and cocaine craving and advance our understanding of the role of the pain circuit and negative reinforcement in sustaining habitual drug use in cocaine addiction.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States.
| | - Simon Zhornitsky
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Wuyi Wang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Isha Dhingra
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Thang M. Le
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Chiang-shan R. Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT,Department of Neuroscience, Yale University School of Medicine, New Haven, CT,Interdepartmental Neuroscience Program, Yale University, New Haven, CT
| |
Collapse
|
134
|
The infralimbic cortex and mGlu5 mediate the effects of chronic intermittent ethanol exposure on fear learning and memory. Psychopharmacology (Berl) 2020; 237:3417-3433. [PMID: 32767063 PMCID: PMC7572878 DOI: 10.1007/s00213-020-05622-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/27/2020] [Indexed: 01/19/2023]
Abstract
RATIONALE AND OBJECTIVES Alcohol use disorder (AUD) and post-traumatic stress disorder (PTSD) often occur comorbidly. While the incidence of these disorders is increasing, there is little investigation into the interacting neural mechanisms between these disorders. These studies aim to identify cognitive deficits that occur as a consequence of fear and ethanol exposure, implement a novel pharmaceutical intervention, and determine relevant underlying neurocircuitry. Additionally, due to clinical sex differences in PTSD prevalence and alcohol abuse, these studies examine the nature of this relationship in rodent models. METHODS Animals were exposed to a model of PTSD+AUD using auditory fear conditioning followed by chronic intermittent ethanol exposure (CIE). Then, rats received extinction training consisting of multiple conditioned stimulus presentations in absence of the shock. Extinction recall and context-induced freezing were measured in subsequent tests. CDPPB, a metabotropic glutamate receptor 5 (mGlu5) positive allosteric modulator, was used to treat these deficits, and region-specific effects were determined using microinjections. RESULTS These studies determined that CIE exposure led to deficits in fear extinction learning and heightened context-induced freezing while sex differences emerged in fear conditioning and extinction cue recall tests. Furthermore, using CDPPB, these studies found that enhancement of infralimbic (IfL) mGlu5 activity was able to recover CIE-induced deficits in both males and females. CONCLUSIONS These studies show that CIE induces deficits in fear-related behaviors and that enhancement of IfL glutamatergic activity can facilitate learning during extinction. Additionally, we identify novel pharmacological targets for the treatment of individuals who suffer from PTSD and AUD.
Collapse
|
135
|
Improving translation of animal models of addiction and relapse by reverse translation. Nat Rev Neurosci 2020; 21:625-643. [PMID: 33024318 DOI: 10.1038/s41583-020-0378-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2020] [Indexed: 12/13/2022]
Abstract
Critical features of human addiction are increasingly being incorporated into complementary animal models, including escalation of drug intake, punished drug seeking and taking, intermittent drug access, choice between drug and non-drug rewards, and assessment of individual differences based on criteria in the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV). Combined with new technologies, these models advanced our understanding of brain mechanisms of drug self-administration and relapse, but these mechanistic gains have not led to improvements in addiction treatment. This problem is not unique to addiction neuroscience, but it is an increasing source of disappointment and calls to regroup. Here we first summarize behavioural and neurobiological results from the animal models mentioned above. We then propose a reverse translational approach, whose goal is to develop models that mimic successful treatments: opioid agonist maintenance, contingency management and the community-reinforcement approach. These reverse-translated 'treatments' may provide an ecologically relevant platform from which to discover new circuits, test new medications and improve translation.
Collapse
|
136
|
Neuroimaging reveals functionally distinct neuronal networks associated with high-level alcohol consumption in two genetic rat models. Behav Pharmacol 2020; 32:229-238. [PMID: 32925226 DOI: 10.1097/fbp.0000000000000582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human imaging data suggest that the motivational processes associated with alcohol reward are reflected in the patterns of neural activation after alcohol or alcohol-related cues. In animal models of alcohol drinking, however, the changes in brain activation during voluntary alcohol ingestion are poorly known. In order to improve the translational utility of animal models, we examined alcohol-induced functional brain activation in Alko Alcohol (AA) and Marchigian-Sardinian alcohol-preferring (msP) rats that drink voluntarily high levels of alcohol, but exhibit widely different neurochemical and behavioral traits cosegregated with alcohol preference. Brain imaging was performed using manganese-enhanced MRI (MEMRI), which is based on accumulation of Mn2+ ions in activated neurons, allowing the identification of functional neuronal networks recruited during specific behaviors in awake animals during a subsequent imaging session under anesthesia. MEMRI was performed following 4 weeks of voluntary alcohol drinking, using water drinking as the control. Despite similar levels of alcohol drinking, strikingly different alcohol-induced neuronal activity patterns were observed in AA and msP rats. Overall, functional activation in the AA rats was more widespread, involving large cortical areas and subcortical structures, such as the bed nucleus of the stria terminalis, preoptic area, hypothalamus, periaqueductal grey, and substantia nigra. In the msP rats, however, alcohol-related activation was largely confined to prefrontal cortical regions and insular cortex, and olfactory areas. Overlapping areas of activation found in both rat lines included the nucleus accumbens, prelimbic, orbital, and insular cortex. In conclusion, our data reveal strikingly different brain circuits associated with alcohol drinking in two genetically different rat lines and suggest innately different motivational and behavioral processes driving alcohol drinking. These findings have important implications for the use of these lines in translational alcohol research.
Collapse
|
137
|
Integrative analysis of shared genetic pathogenesis by autism spectrum disorder and obsessive-compulsive disorder. Biosci Rep 2020; 39:221433. [PMID: 31808517 PMCID: PMC6928520 DOI: 10.1042/bsr20191942] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/13/2019] [Accepted: 11/22/2019] [Indexed: 12/15/2022] Open
Abstract
Many common pathological features have been observed for both autism spectrum disorders (ASDs) and obsessive-compulsive disorder (OCD). However, no systematic analysis of the common gene markers associated with both ASD and OCD has been conducted so far. Here, two batches of large-scale literature-based disease–gene relation data (updated in 2017 and 2019, respectively) and gene expression data were integrated to study the possible association between OCD and ASD at the genetic level. Genes linked to OCD and ASD present significant overlap (P-value <2.64e-39). A genetic network of over 20 genes was constructed, through which OCD and ASD may exert influence on each other. The 2017-based analysis suggested six potential common risk genes for OCD and ASD (CDH2, ADCY8, APOE, TSPO, TOR1A, and OLIG2), and the 2019-based study identified two more genes (DISP1 and SETD1A). Notably, the gene APOE identified by the 2017-based analysis has been implicated to have an association with ASD in a recent study (2018) with DNA methylation analysis. Our results support the possible complex genetic associations between OCD and ASD. Genes linked to one disease are worth further investigation as potential risk factors for the other.
Collapse
|
138
|
Piantadosi PT, Yeates DCM, Floresco SB. Prefrontal cortical and nucleus accumbens contributions to discriminative conditioned suppression of reward-seeking. ACTA ACUST UNITED AC 2020; 27:429-440. [PMID: 32934096 PMCID: PMC7497111 DOI: 10.1101/lm.051912.120] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
Fear can potently inhibit ongoing behavior, including reward-seeking, yet the neural circuits that underlie such suppression remain to be clarified. Prior studies have demonstrated that distinct subregions of the rodent medial prefrontal cortex (mPFC) differentially affect fear behavior, whereby fear expression is promoted by the more dorsal prelimbic cortex (PL) and inhibited by the more ventral infralimbic cortex (IL). These mPFC regions project to subregions of the nucleus accumbens, the core (NAcC) and shell (NAcS), that differentially contribute to reward-seeking as well as affective processes that may be relevant to fear expression. Here, we investigated how these mPFC and NAc subregions contribute to discriminative fear conditioning, assessed by conditioned suppression of reward-seeking. Bilateral inactivation of the NAcS or PL reduced the expression of conditioned suppression to a shock-associated CS+, whereas NAcC inactivation reduced reward-seeking without affecting suppression. IL inactivation caused a general reduction in conditioned suppression following discriminative conditioning, but not when using a single-stimulus design. Pharmacological disconnection of the PL → NAcS pathway revealed that this projection mediates conditioned suppression. These data add to a growing literature implicating discrete cortico-striatal pathways in the suppression of reward-seeking in response to aversive stimuli. Dysfunction within related structures may contribute to aberrant patterns of behavior in psychiatric illnesses including substance use disorders.
Collapse
Affiliation(s)
- Patrick T Piantadosi
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Dylan C M Yeates
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
139
|
Engleman EA, Ingraham CM, Rodd ZA, Murphy JM, McBride WJ, Ding ZM. The reinforcing effects of ethanol within the prelimbic cortex and ethanol drinking: Involvement of local dopamine D 2 receptor-mediated neurotransmission. Drug Alcohol Depend 2020; 214:108165. [PMID: 32688071 PMCID: PMC7431019 DOI: 10.1016/j.drugalcdep.2020.108165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/21/2020] [Accepted: 06/20/2020] [Indexed: 11/17/2022]
Abstract
Previous studies have identified important mesolimbic regions in supporting the reinforcing effects of ethanol. However, the involvement of the medial prefrontal cortex (mPFC), another key region within the mesocorticolimbic system, in ethanol reinforcement has been understudied. The objective of the current study was to examine the role of the prelimbic (PL) cortex sub-region of the mPFC in ethanol reinforcement and drinking. Intracranial self-administration was used to examine the reinforcing effects of ethanol within the PL cortex. Quantitative microdialysis was used to measure basal extracellular DA concentrations and clearance in the PL cortex following chronic ethanol drinking. In addition, the involvement of dopamine (DA) D2 receptors within the PL cortex on the reinforcing effects of ethanol and ethanol drinking was determined. Ethanol was dose-dependent self-administered into the PL cortex, with significantly more infusions elicited by 100-200 mg% ethanol than vehicle. Co-infusion of the D2 receptor antagonist sulpiride significantly reduced ethanol self-administration. Chronic ethanol drinking significantly elevated basal extracellular DA concentrations without altering DA clearance. Microinjection of sulpiride into the PL cortex selectively reduced ethanol, but not saccharine, drinking. These results indicate that the PL cortex supported the reinforcing effects of ethanol, and that ethanol drinking enhanced basal DA neurotransmission within the PL cortex. In addition, D2 receptor antagonism within the PL cortex reduced ethanol self-administration and drinking. Collectively, these findings revealed important DA mechanisms within the PL cortex in mediating ethanol reinforcement and drinking.
Collapse
Affiliation(s)
- Eric A Engleman
- Department of Psychiatry, Indiana University School of Medicine, 355 West 16th Street, Indianapolis, IN 46202, United States
| | - Cynthia M Ingraham
- Department of Psychiatry, Indiana University School of Medicine, 355 West 16th Street, Indianapolis, IN 46202, United States
| | - Zachary A Rodd
- Department of Psychiatry, Indiana University School of Medicine, 355 West 16th Street, Indianapolis, IN 46202, United States
| | - James M Murphy
- Department of Psychiatry, Indiana University School of Medicine, 355 West 16th Street, Indianapolis, IN 46202, United States
| | - William J McBride
- Department of Psychiatry, Indiana University School of Medicine, 355 West 16th Street, Indianapolis, IN 46202, United States
| | - Zheng-Ming Ding
- Department of Psychiatry, Indiana University School of Medicine, 355 West 16th Street, Indianapolis, IN 46202, United States; Department of Anesthesiology and Perioperative Medicine, Department of Pharmacology, Pennsylvania State University College of Medicine, 700 HMC Crescent Road, Hershey, PA 17033, United States.
| |
Collapse
|
140
|
Kyriazi P, Headley DB, Paré D. Different Multidimensional Representations across the Amygdalo-Prefrontal Network during an Approach-Avoidance Task. Neuron 2020; 107:717-730.e5. [PMID: 32562662 PMCID: PMC7442738 DOI: 10.1016/j.neuron.2020.05.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/21/2020] [Accepted: 05/28/2020] [Indexed: 01/07/2023]
Abstract
The prelimbic (PL) area and basolateral amygdala (lateral [LA] and basolateral [BL] nuclei) have closely related functions and similar extrinsic connectivity. Reasoning that the computational advantage of such redundancy should be reflected in differences in how these structures represent information, we compared the coding properties of PL and amygdala neurons during a task that requires rats to produce different conditioned defensive or appetitive behaviors. Rather than unambiguous regional differences in the identities of the variables encoded, we found gradients in how the same variables are represented. Whereas PL and BL neurons represented many different parameters through minor variations in firing rates, LA cells coded fewer task features with stronger changes in activity. At the population level, whereas valence could be easily distinguished from amygdala activity, PL neurons could distinguish both valence and trial identity as well as or better than amygdala neurons. Thus, PL has greater representational capacity.
Collapse
Affiliation(s)
- Pinelopi Kyriazi
- Center for Molecular and Behavioral Neuroscience, Rutgers State University, Newark, NJ 07102, USA; Behavioral and Neural Sciences Graduate Program, Rutgers State University, Newark, NJ 07102, USA
| | - Drew B Headley
- Center for Molecular and Behavioral Neuroscience, Rutgers State University, Newark, NJ 07102, USA.
| | - Denis Paré
- Center for Molecular and Behavioral Neuroscience, Rutgers State University, Newark, NJ 07102, USA.
| |
Collapse
|
141
|
Joshi DD, Puaud M, Fouyssac M, Belin‐Rauscent A, Everitt B, Belin D. The anterior insular cortex in the rat exerts an inhibitory influence over the loss of control of heroin intake and subsequent propensity to relapse. Eur J Neurosci 2020; 52:4115-4126. [DOI: 10.1111/ejn.14889] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Dhaval D. Joshi
- Department of Psychology University of Cambridge Cambridge UK
| | - Mickaël Puaud
- Department of Psychology University of Cambridge Cambridge UK
| | - Maxime Fouyssac
- Department of Psychology University of Cambridge Cambridge UK
| | | | - Barry Everitt
- Department of Psychology University of Cambridge Cambridge UK
| | - David Belin
- Department of Psychology University of Cambridge Cambridge UK
| |
Collapse
|
142
|
Guerin AA, Zbukvic IC, Luikinga SJ, Drummond KD, Lawrence AJ, Madsen HB, Kim JH. Extinction and drug-induced reinstatement of cocaine seeking following self-administration or conditioned place preference in adolescent and adult rats. Dev Psychobiol 2020; 63:125-137. [PMID: 32666555 DOI: 10.1002/dev.22017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022]
Abstract
Adolescence marks a particularly vulnerable period to developing substance use disorders, and people who start using drugs in adolescence are more likely to relapse. A limited number of studies have investigated age difference in relapse following re-exposure to the drug after a period of abstinence. Using a cocaine self-administration paradigm, we showed no age difference in acquisition or extinction of self-administration. Interestingly, adolescent rats displayed impaired cocaine-primed reinstatement of cocaine seeking. Using the same dose as that self-administered in the first experiment, we then investigated age differences in acquisition and extinction of conditioned place preference, as well as locomotor sensitization. While there were no differences in locomotor activity or acquisition of preference, adolescents failed to extinguish their preference, even when the number of extinction sessions was doubled from what adults received. Taken together, these results suggest that while cocaine has similar rewarding and reinforcing effects regardless of age, adolescents may attribute stronger salience to the drug-associated context. In addition, re-exposure to cocaine itself may not be a strong relapse trigger in adolescence. Overall, these findings suggest that we should focus more on alleviating drug-context salience compared to re-exposure to substance in order to reduce relapse of drug seeking in adolescents.
Collapse
Affiliation(s)
- Alexandre A Guerin
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Isabel C Zbukvic
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Sophia J Luikinga
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Katherine D Drummond
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Andrew J Lawrence
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Heather B Madsen
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Jee Hyun Kim
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
143
|
Pharmacological Inactivation of Medial Prefrontal Cortex Does Not Support Dichotomous "Go/Stop" Roles for Dorsal and Ventral Subdivisions in Natural Reward Seeking in Rats. eNeuro 2020; 7:7/4/ENEURO.0246-20.2020. [PMID: 32646921 PMCID: PMC8114868 DOI: 10.1523/eneuro.0246-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Highlighted Research Paper:Differential Effects of Dorsal and Ventral Medial Prefrontal Cortex Inactivation during Natural Reward Seeking, Extinction, and Cue-Induced Reinstatement. Jessica P. Caballero, Garrett B. Scarpa, Luke Remage-Healey, David E. Moorman.
Collapse
|
144
|
Randall PA, McElligott ZA, Besheer J. Role of mPFC and nucleus accumbens circuitry in modulation of a nicotine plus alcohol compound drug state. Addict Biol 2020; 25:e12782. [PMID: 31173443 DOI: 10.1111/adb.12782] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/17/2019] [Accepted: 04/30/2019] [Indexed: 12/23/2022]
Abstract
Combined use of nicotine and alcohol constitute a significant public health risk. An important aspect of drug use and dependence are the various cues, both external (contextual) and internal (interoceptive) that influence drug-seeking and drug-taking behavior. The present experiments employed the use of Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) and complementary Pavlovian drug discrimination procedures (feature-positive and feature-negative training conditions) in order to examine whether medial prefrontal cortex (prelimbic; mPFC-PL) projections to the nucleus accumbens core (AcbC) modulate sensitivity to a nicotine + alcohol (N + A) interoceptive cue. First, we show neuronal activation in mPFC-PL and AcbC following treatment with N + A. Next, we demonstrate that chemogenetic silencing of projections from mPFC-PL to nucleus accumbens core decrease sensitivity to the N + A interoceptive cue, while enhancing sensitivity to the individual components, suggesting an important role for this specific projection. Furthermore, we demonstrate that clozapine-N-oxide (CNO), the ligand used to activate the DREADDs, had no effect in parallel mCherry controls. These findings contribute important information regarding our understanding of the cortical-striatal circuitry that regulates sensitivity to the interoceptive effects of a compound N + A cue.
Collapse
Affiliation(s)
- Patrick A. Randall
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill NC USA
- Neuroscience Curriculum University of North Carolina at Chapel Hill Chapel Hill NC USA
- Department of Psychiatry University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Joyce Besheer
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill NC USA
- Neuroscience Curriculum University of North Carolina at Chapel Hill Chapel Hill NC USA
- Department of Psychiatry University of North Carolina at Chapel Hill Chapel Hill NC USA
| |
Collapse
|
145
|
Moro F, Giannotti G, Caffino L, Marzo CM, Di Clemente A, Fumagalli F, Cervo L. Lasting reduction of nicotine-seeking behavior by chronic N-acetylcysteine during experimental cue-exposure therapy. Addict Biol 2020; 25:e12771. [PMID: 31132808 DOI: 10.1111/adb.12771] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 02/10/2019] [Accepted: 04/09/2019] [Indexed: 10/26/2022]
Abstract
Nicotine-associated cues can trigger reinstatement in humans as well as in animal models of drug addiction. To date, no behavioral intervention or pharmacological treatment has been effective in preventing relapse in the long term. A large body of evidence indicates that N-acetylcysteine (N-AC) blunts the activation of glutamatergic (GLUergic) neurons in the nucleus accumbens (Nacc) associated with reinstatement. We evaluated the effect of an experimental cue exposure therapy (eCET) alone or in combination with N-AC to verify whether restoring GLU homeostasis enhances extinction of nicotine-associated cues. Rats were trained to associate discriminative stimuli with intravenous nicotine or saline self-administration. Reinforced response was followed by cue signals. After rats met the self-administration criteria, the lasting anti-relapse activity of i.p. N-AC or vehicle was assessed in three different experimental conditions after 14 days of treatment: treatment + eCET; treatment + lever-presses extinction (LP-EXT); and treatment + abstinence. N-AC 100 mg/kg, but not 60 mg/kg, induced anti-relapse activity that persisted 50 days after treatment only when paired with either LP-EXT or eCET with the greater activity found in the latter condition. To identify potential mechanisms for behavioral results, separate groups of rats that received either N-AC or vehicle + eCET were killed at different time points for Nacc Western-blot analysis. Seven days after treatment, chronic N-AC restored the expression of proteins crucial for GLU homeostasis, while at 50 days, it increased the expression of type II metabotropic GLU receptors. These results suggest that N-AC treatment in combination with eCET may offer a novel strategy to prevent relapse in nicotine addiction.
Collapse
Affiliation(s)
- Federico Moro
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research—IRCCS Milan Italy
| | | | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | | | - Angelo Di Clemente
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research—IRCCS Milan Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Luigi Cervo
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research—IRCCS Milan Italy
| |
Collapse
|
146
|
Coelho CM, Gonçalves-Bradley D, Zsido AN. Who worries about specific phobias? - A population-based study of risk factors. J Psychiatr Res 2020; 126:67-72. [PMID: 32417598 DOI: 10.1016/j.jpsychires.2020.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/28/2020] [Accepted: 05/01/2020] [Indexed: 10/24/2022]
Abstract
Although specific phobia is one of the most prevalent lifetime anxiety disorders, little is known about the particular risk factors related to its development. The underlying goal of this study was to analyse the risk factors associated with worrying about specific phobias (SP) in a representative sample of community dwelling adults. The sample was composed of 8461 participants (mean age 47.68 years, range 18-85, 54.60% female), from the Australian National Mental Health Survey. A total of 188 participants (2.22%) reported worrying about SPs. Multivariate logistic regression analysis indicated that female sex (odds ratio (OR) = 1.98, p < 0.0001) and a comorbid diagnosis of lifetime major depression disorder (OR = 2.80, p < 0.0001) were the factors most strongly associated with worrying about SPs. Having experienced traumatic experiences involving significant others (OR = 1.18, p = 0.02), the number of chronic diseases (OR = 1.21, p < 0.01), and a comorbid diagnosis of substance use (OR = 2.80, p = 0.02) were also associated. Our results are in line with previous studies focusing on other anxiety disorders. We provide further evidence that substance dependence appears to serve as a unique risk factor for the subsequent onset of SP. Further empirical and clinical implications are discussed.
Collapse
Affiliation(s)
- Carlos M Coelho
- School of Psychology, ISMAI University Institute of Maia, Portugal; School of Health of Porto Polytechnic, Psychosocial Rehabilitation Lab, Center for Rehabilitation Research, Porto, Portugal; Department of Psychology, Faculty of Psychology, Chulalongkorn University, Bangkok, Thailand
| | | | - Andras N Zsido
- Institute of Psychology, University of Pécs, Pécs, Hungary.
| |
Collapse
|
147
|
Interactions between prelimbic cortex and basolateral amygdala contribute to morphine-induced conditioned taste aversion in conditioning and extinction. Neurobiol Learn Mem 2020; 172:107248. [DOI: 10.1016/j.nlm.2020.107248] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/28/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022]
|
148
|
Cuesta S, Nouel D, Reynolds LM, Morgunova A, Torres-Berrío A, White A, Hernandez G, Cooper HM, Flores C. Dopamine Axon Targeting in the Nucleus Accumbens in Adolescence Requires Netrin-1. Front Cell Dev Biol 2020; 8:487. [PMID: 32714924 PMCID: PMC7344302 DOI: 10.3389/fcell.2020.00487] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
The fine arrangement of neuronal connectivity during development involves the coordinated action of guidance cues and their receptors. In adolescence, the dopamine circuitry is still developing, with mesolimbic dopamine axons undergoing target-recognition events in the nucleus accumbens (NAcc), while mesocortical projections continue to grow toward the prefrontal cortex (PFC) until adulthood. This segregation of mesolimbic versus mesocortical dopamine pathways is mediated by the guidance cue receptor DCC, which signals dopamine axons intended to innervate the NAcc to recognize this region as their final target. Whether DCC-dependent mesolimbic dopamine axon targeting in adolescence requires the action of its ligand, Netrin-1, is unknown. Here we combined shRNA strategies, quantitative analysis of pre- and post-synaptic markers of neuronal connectivity, and pharmacological manipulations to address this question. Similar to DCC levels in the ventral tegmental area, Netrin-1 expression in the NAcc is dynamic across postnatal life, transitioning from high to low expression across adolescence. Silencing Netrin-1 in the NAcc in adolescence results in an increase in the expanse of the dopamine input to the PFC in adulthood, with a corresponding increase in the number of presynaptic dopamine sites. This manipulation also results in altered dendritic spine density and morphology of medium spiny neurons in the NAcc in adulthood and in reduced sensitivity to the behavioral activating effects of the stimulant drug of abuse, amphetamine. These cellular and behavioral effects mirror those induced by Dcc haploinsufficiency within dopamine neurons in adolescence. Dopamine targeting in adolescence requires the complementary interaction between DCC receptors in mesolimbic dopamine axons and Netrin-1 in the NAcc. Factors regulating either DCC or Netrin-1 in adolescence can disrupt mesocorticolimbic dopamine development, rendering vulnerability or protection to phenotypes associated with psychiatric disorders.
Collapse
Affiliation(s)
- Santiago Cuesta
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Dominique Nouel
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Lauren M Reynolds
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Alice Morgunova
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Angélica Torres-Berrío
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Amanda White
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Giovanni Hernandez
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Cecilia Flores
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
149
|
Wang Y, Liu Z, Cai L, Guo R, Dong Y, Huang YH. A Critical Role of Basolateral Amygdala-to-Nucleus Accumbens Projection in Sleep Regulation of Reward Seeking. Biol Psychiatry 2020; 87:954-966. [PMID: 31924324 PMCID: PMC7210061 DOI: 10.1016/j.biopsych.2019.10.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/09/2019] [Accepted: 10/27/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Sleep impacts reward-motivated behaviors partly by retuning the brain reward circuits. The nucleus accumbens (NAc) is a reward processing hub sensitive to acute sleep deprivation. Glutamatergic transmission carrying reward-associated signals converges in the NAc and regulates various aspects of reward-motivated behaviors. The basolateral amygdala projection (BLAp) innervates broad regions of the NAc and critically regulates reward seeking. METHODS Using slice electrophysiology, we measured how acute sleep deprivation alters transmission at BLAp-NAc synapses in male C57BL/6 mice. Moreover, using SSFO (stabilized step function opsin) and DREADDs (designer receptors exclusively activated by designer drugs) (Gi) to amplify and reduce transmission, respectively, we tested behavioral consequences following bidirectional manipulations of BLAp-NAc transmission. RESULTS Acute sleep deprivation increased sucrose self-administration in mice and altered the BLAp-NAc transmission in a topographically specific manner. It selectively reduced glutamate release at the rostral BLAp (rBLAp) onto ventral and lateral NAc (vlNAc) synapses, but spared caudal BLAp onto medial NAc synapses. Furthermore, experimentally facilitating glutamate release at rBLAp-vlNAc synapses suppressed sucrose reward seeking. Conversely, mimicking sleep deprivation-induced reduction of rBLAp-vlNAc transmission increased sucrose reward seeking. Finally, facilitating rBLAp-vlNAc transmission per se did not promote either approach motivation or aversion. CONCLUSIONS Sleep acts on rBLAp-vINAc transmission gain control to regulate established reward seeking but does not convey approach motivation or aversion on its own.
Collapse
Affiliation(s)
- Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA,These authors contributed equally to this work
| | - Zheng Liu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,These authors contributed equally to this work
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Yan Dong
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA
| | - Yanhua H. Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
150
|
Halladay LR, Kocharian A, Piantadosi PT, Authement ME, Lieberman AG, Spitz NA, Coden K, Glover LR, Costa VD, Alvarez VA, Holmes A. Prefrontal Regulation of Punished Ethanol Self-administration. Biol Psychiatry 2020; 87:967-978. [PMID: 31937415 PMCID: PMC7217757 DOI: 10.1016/j.biopsych.2019.10.030] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND A clinical hallmark of alcohol use disorder is persistent drinking despite potential adverse consequences. The ventromedial prefrontal cortex (vmPFC) and dorsomedial prefrontal cortex (dmPFC) are positioned to exert top-down control over subcortical regions, such as the nucleus accumbens shell (NAcS) and basolateral amygdala, which encode positive and negative valence of ethanol (EtOH)-related stimuli. Prior rodent studies have implicated these regions in regulation of punished EtOH self-administration (EtOH-SA). METHODS We conducted in vivo electrophysiological recordings in mouse vmPFC and dmPFC to obtain neuronal correlates of footshock-punished EtOH-SA. Ex vivo recordings were performed in NAcS D1 receptor-expressing medium spiny neurons receiving vmPFC input to examine punishment-related plasticity in this pathway. Optogenetic photosilencing was employed to assess the functional contribution of the vmPFC, dmPFC, vmPFC projections to NAcS, or vmPFC projections to basolateral amygdala, to punished EtOH-SA. RESULTS Punishment reduced EtOH lever pressing and elicited aborted presses (lever approach followed by rapid retraction). Neurons in the vmPFC and dmPFC exhibited phasic firing to EtOH lever presses and aborts, but only in the vmPFC was there a population-level shift in coding from lever presses to aborts with punishment. Closed-loop vmPFC, but not dmPFC, photosilencing on a postpunishment probe test negated the reduction in EtOH lever presses but not in aborts. Punishment was associated with altered plasticity at vmPFC inputs to D1 receptor-expressing medium spiny neurons in the NAcS. Photosilencing vmPFC projections to the NAcS, but not to the basolateral amygdala, partially reversed suppression of EtOH lever presses on probe testing. CONCLUSIONS These findings demonstrate a key role for the vmPFC in regulating EtOH-SA after punishment, with implications for understanding the neural basis of compulsive drinking in alcohol use disorder.
Collapse
Affiliation(s)
- Lindsay R Halladay
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Department of Psychology, Santa Clara University, Santa Clara, California.
| | - Adrina Kocharian
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Patrick T Piantadosi
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Michael E Authement
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Abby G Lieberman
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Nathen A Spitz
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Kendall Coden
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Lucas R Glover
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Vincent D Costa
- Department of Behavioral Neuroscience, Oregon Health Sciences University, Portland, Oregon
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|