1
|
Ardinger CE, Lapish CC, Linsenbardt DN. Repeated Binge Alcohol Drinking Leads to Reductions in Corticostriatal Theta Coherence in Female but not Male Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.581791. [PMID: 38496601 PMCID: PMC10942409 DOI: 10.1101/2024.03.07.581791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Decreased functional connectivity between the striatum and frontal cortex is observed in individuals with alcohol use disorder (AUD), and predicts the probability of relapse in abstinent individuals with AUD. To further our understanding of how repeated alcohol (ethanol; EtOH) consumption impacts the corticostriatal circuit, extracellular electrophysiological recordings (local field potentials; LFPs) were gathered from the nucleus accumbens (NAc) and prefrontal cortex (PFC) of C57BL/6J mice voluntarily consuming EtOH or water using a 'drinking-in-the-dark' (DID) procedure. Following a three-day acclimation period wherein only water access was provided during DID, mice were given 15 consecutive days of access to EtOH. Each session consisted of a 30-minute baseline period where water was available and was followed immediately by a 2-hour period where sippers containing water were replaced with new sippers containing either unsweetened 20% (v/v) EtOH (days 4-18; DID) or water (days 1-3; acclimation). Our analyses focused primarily on theta coherence during bouts of drinking, as differences in this band are associated with several behavioral markers of AUD. Both sexes displayed decreases in theta coherence during the first day of binge EtOH consumption. However, only females displayed further decreases in theta coherence on the 14th day of EtOH access. No differences in theta coherence were observed between the first and final bout on any EtOH drinking days. These results provide additional support for decreases in the functional coupling of corticostriatal circuits as a consequence of alcohol consumption and suggests that female mice are uniquely vulnerable to these effects following repeated EtOH drinking.
Collapse
Affiliation(s)
- Cherish E Ardinger
- Addiction Neuroscience, Department of Psychology and Indiana Alcohol Research Center, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202
| | - Christopher C Lapish
- Addiction Neuroscience, Department of Psychology and Indiana Alcohol Research Center, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202
- Indiana University School of Medicine Stark Neuroscience Institute, Indianapolis, Indiana, 46202
| | - David N Linsenbardt
- Department of Neurosciences School of Medicine and Health Sciences Center, University of New Mexico, Albuquerque, New Mexico, 87131
| |
Collapse
|
2
|
Sallie SN, Sonkusare S, Mandali A, Casero V, Cui H, Guzman NV, Allison M, Voon V. Cortical paired associative stimulation shows impaired plasticity of inhibition networks as a function of chronic alcohol use. Psychol Med 2024; 54:698-709. [PMID: 37712403 DOI: 10.1017/s0033291723002374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
BACKGROUND Response inhibition - or the ability to withhold a suboptimal response - relies on the efficacy of fronto-striatal networks, and is impaired in neuropsychiatric disorders including addiction. Cortical paired associative stimulation (cPAS) is a form of transcranial magnetic stimulation (TMS) which can strengthen neuronal connections via spike-timing-dependent plasticity mechanisms. Here, we used cPAS targeting the fronto-striatal inhibitory network to modulate performance on a response inhibition measure in chronic alcohol use. METHODS Fifty-five participants (20 patients with a formal alcohol use disorder (AUD) diagnosis (26-74 years, 6[30%] females) and 20 matched healthy controls (HCs) (27-73 years, 6[30%] females) within a larger sample of 35 HCs (23-84 years, 11[31.4%] females) underwent two randomized sessions of cPAS 1-week apart: right inferior frontal cortex stimulation preceding right presupplementary motor area stimulation by either 4 ms (excitation condition) or 100 ms (control condition), and were subsequently administered the Stop Signal Task (SST) in both sessions. RESULTS HCs showed decreased stop signal reaction time in the excitation condition (t(19) = -3.01, p = 0.007, [CIs]:-35.6 to -6.42); this facilitatory effect was not observed for AUD (F(1,31) = 9.57, p = 0.004, CIs: -68.64 to -14.11). Individually, rates of SST improvement were substantially higher for healthy (72%) relative to AUD (13.6%) groups (OR: 2.33, p = 0.006, CIs:-3.34 to -0.55). CONCLUSION In line with previous findings, cPAS improved response inhibition in healthy adults by strengthening the fronto-striatal network through putative long-term potentiation-like plasticity mechanisms. Furthermore, we identified a possible marker of impaired cortical excitability, and, thus, diminished capacity for cPAS-induced neuroplasticity in AUD with direct implications to a disorder-relevant cognitive process.
Collapse
Affiliation(s)
- Samantha N Sallie
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Saurabh Sonkusare
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Alekhya Mandali
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- MRC Brain Network Dynamics Unit, University of Oxford, Oxford, OX13TH, UK
| | - Violeta Casero
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Hailun Cui
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Natalie V Guzman
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Michael Allison
- Liver Unit, Department of Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Valerie Voon
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0QQ, UK
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Pagni BA, Petridis PD, Podrebarac SK, Grinband J, Claus ED, Bogenschutz MP. Psilocybin-induced changes in neural reactivity to alcohol and emotional cues in patients with alcohol use disorder: an fMRI pilot study. Sci Rep 2024; 14:3159. [PMID: 38326432 PMCID: PMC10850478 DOI: 10.1038/s41598-024-52967-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/25/2024] [Indexed: 02/09/2024] Open
Abstract
This pilot study investigated psilocybin-induced changes in neural reactivity to alcohol and emotional cues in patients with alcohol use disorder (AUD). Participants were recruited from a phase II, randomized, double-blind, placebo-controlled clinical trial investigating psilocybin-assisted therapy (PAT) for the treatment of AUD (NCT02061293). Eleven adult patients completed task-based blood oxygen dependent functional magnetic resonance imaging (fMRI) approximately 3 days before and 2 days after receiving 25 mg of psilocybin (n = 5) or 50 mg of diphenhydramine (n = 6). Visual alcohol and emotionally valanced (positive, negative, or neutral) stimuli were presented in block design. Across both alcohol and emotional cues, psilocybin increased activity in the medial and lateral prefrontal cortex (PFC) and left caudate, and decreased activity in the insular, motor, temporal, parietal, and occipital cortices, and cerebellum. Unique to negative cues, psilocybin increased supramarginal gyrus activity; unique to positive cues, psilocybin increased right hippocampus activity and decreased left hippocampus activity. Greater PFC and caudate engagement and concomitant insula, motor, and cerebellar disengagement suggests enhanced goal-directed action, improved emotional regulation, and diminished craving. The robust changes in brain activity observed in this pilot study warrant larger neuroimaging studies to elucidate neural mechanisms of PAT.Trial registration: NCT02061293.
Collapse
Affiliation(s)
- B A Pagni
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - P D Petridis
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - S K Podrebarac
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - J Grinband
- Departments of Psychiatry and Radiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - E D Claus
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, USA
| | - M P Bogenschutz
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Zhang H, Sun H, Li J, Yang J, Fan Y, Jülich ST, Lei X. Response inhibition impairment related to altered frontal-striatal functional connectivity in insomnia disorder: A pilot and non-clinical study. J Psychiatr Res 2024; 170:138-146. [PMID: 38134723 DOI: 10.1016/j.jpsychires.2023.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND It is not clear whether and how insomnia disorder (ID) impairs response inhibition ability. Fronto-striatal functional connectivity (FC) plays a critical role in response inhibition and is found be abnormal in patients with ID. In this study, we examined whether insomnia symptoms impair response inhibition in a large non-clinical sample and whether impaired response inhibition is related to abnormal fronto-striatal FC. METHODS One hundred and fifteen young ID patients and 160 age and sex-matched healthy controls (HC) underwent resting-state functional magnetic response imaging scans and performed the stop-signal task (SST). Performance of SST, Gray Matter Volumes (GMVs), and connections of brain regions related to fronto-striatal circuits was compared between groups. Further examined the association between response inhibition impairment and fronto-striatal FC. RESULTS The behavioral results showed that patients with ID had significantly longer stop-signal reaction time (SSRT) compared with the HC, reflecting the impaired response inhibition among IDs. Brain imaging results showed IDs had decreased GMVs of the Right Superior Frontal (SFG) and left Supplementary Motor area (SMA). Seed-based FC results showed that compared to HC, the ID showed decreased FC between left SMA and left Paracentral lobule, left SMA and right SMA, and right SFG and right Orbital Middle Frontal gyrus, and increased FC between right SFG and right putamen. Meanwhile, the FC between right SFG and putamen was positively correlated with SSRT in IDs. CONCLUSIONS The current study found significantly impaired response inhibition among ID and this impairment may be related to abnormal fronto-striatal FC in ID.
Collapse
Affiliation(s)
- Haobo Zhang
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing, 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing, 400715, China
| | - Haonan Sun
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing, 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing, 400715, China
| | - Jiatao Li
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing, 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing, 400715, China
| | - Jingqi Yang
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing, 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing, 400715, China
| | - Yuhan Fan
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing, 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing, 400715, China
| | - Simon Theodor Jülich
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing, 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing, 400715, China
| | - Xu Lei
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing, 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing, 400715, China.
| |
Collapse
|
5
|
Bellmunt-Gil A, Majuri J, Arponen E, Kaasinen V, Joutsa J. Abnormal frontostriatal connectivity and serotonin function in gambling disorder: A preliminary exploratory study. J Behav Addict 2023; 12:670-681. [PMID: 37561637 PMCID: PMC10562820 DOI: 10.1556/2006.2023.00037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/04/2023] [Accepted: 06/26/2023] [Indexed: 08/12/2023] Open
Abstract
Background The neurobiological mechanisms of gambling disorder are not yet fully characterized, limiting the development of treatments. Defects in frontostriatal connections have been shown to play a major role in substance use disorders, but data on behavioral addictions, such as gambling disorder, are scarce. The aim of this study was to 1) investigate whether gambling disorder is associated with abnormal frontostriatal connectivity and 2) characterize the key neurotransmitter systems underlying the connectivity abnormalities. Methods Fifteen individuals with gambling disorder and 17 matched healthy controls were studied with resting-state functional connectivity MRI and three brain positron emission tomography scans, investigating dopamine (18F-FDOPA), opioid (11C-carfentanil) and serotonin (11C-MADAM) function. Frontostriatal connectivity was investigated using striatal seed-to-voxel connectivity and compared between the groups. Neurotransmitter systems underlying the identified connectivity differences were investigated using region-of-interest and voxelwise approaches. Results Individuals with gambling disorder showed loss of functional connectivity between the right nucleus accumbens (NAcc) and a region in the right dorsolateral prefrontal cortex (DLPFC) (PFWE <0.05). Similarly, there was a significant Group x right NAcc interaction in right DLPFC 11C-MADAM binding (p = 0.03) but not in 18F-FDOPA uptake or 11C-carfentanil binding. This was confirmed in voxelwise analyses showing a widespread Group x right NAcc interaction in the prefrontal cortex 11C-MADAM binding (PFWE <0.05). Right NAcc 11C-MADAM binding potential correlated with attentional impulsivity in individuals with gambling disorder (r = -0.73, p = 0.005). Discussion Gambling disorder is associated with right hemisphere abnormal frontostriatal connectivity and serotonergic function. These findings will contribute to understanding the neurobiological mechanism and may help identify potential treatment targets for gambling disorder.
Collapse
Affiliation(s)
- Albert Bellmunt-Gil
- Turku Brain and Mind Center, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Joonas Majuri
- Turku Brain and Mind Center, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | | | - Valtteri Kaasinen
- Turku Brain and Mind Center, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
| | - Juho Joutsa
- Turku Brain and Mind Center, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
| |
Collapse
|
6
|
Maggioni E, Rossetti MG, Allen NB, Batalla A, Bellani M, Chye Y, Cousijn J, Goudriaan AE, Hester R, Hutchison K, Li CR, Martin‐Santos R, Momenan R, Sinha R, Schmaal L, Solowij N, Suo C, van Holst RJ, Veltman DJ, Yücel M, Thompson PM, Conrod P, Mackey S, Garavan H, Brambilla P, Lorenzetti V. Brain volumes in alcohol use disorder: Do females and males differ? A whole-brain magnetic resonance imaging mega-analysis. Hum Brain Mapp 2023; 44:4652-4666. [PMID: 37436103 PMCID: PMC10400785 DOI: 10.1002/hbm.26404] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/03/2023] [Accepted: 06/09/2023] [Indexed: 07/13/2023] Open
Abstract
Emerging evidence suggests distinct neurobiological correlates of alcohol use disorder (AUD) between sexes, which however remain largely unexplored. This work from ENIGMA Addiction Working Group aimed to characterize the sex differences in gray matter (GM) and white matter (WM) correlates of AUD using a whole-brain, voxel-based, multi-tissue mega-analytic approach, thereby extending our recent surface-based region of interest findings on a nearly matching sample using a complementary methodological approach. T1-weighted magnetic resonance imaging (MRI) data from 653 people with AUD and 326 controls was analyzed using voxel-based morphometry. The effects of group, sex, group-by-sex, and substance use severity in AUD on brain volumes were assessed using General Linear Models. Individuals with AUD relative to controls had lower GM volume in striatal, thalamic, cerebellar, and widespread cortical clusters. Group-by-sex effects were found in cerebellar GM and WM volumes, which were more affected by AUD in females than males. Smaller group-by-sex effects were also found in frontotemporal WM tracts, which were more affected in AUD females, and in temporo-occipital and midcingulate GM volumes, which were more affected in AUD males. AUD females but not males showed a negative association between monthly drinks and precentral GM volume. Our results suggest that AUD is associated with both shared and distinct widespread effects on GM and WM volumes in females and males. This evidence advances our previous region of interest knowledge, supporting the usefulness of adopting an exploratory perspective and the need to include sex as a relevant moderator variable in AUD.
Collapse
Affiliation(s)
- Eleonora Maggioni
- Department of Electronics, Information and BioengineeringPolitecnico di MilanoMilanItaly
| | - Maria G. Rossetti
- Department of Neurosciences and Mental HealthFondazione IRCCS Ca'Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of PsychiatryUniversity of VeronaVeronaItaly
| | | | - Albert Batalla
- Department of PsychiatryUniversity Medical Center Utrecht Brain Center, Utrecht UniversityUtrechtthe Netherlands
| | - Marcella Bellani
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of PsychiatryUniversity of VeronaVeronaItaly
| | - Yann Chye
- BrainPark, Turner Institute for Brain and Mental HealthSchool of Psychological SciencesMelbourneAustralia
- Monash Biomedical ImagingMonash UniversityMelbourneAustralia
| | - Janna Cousijn
- Neuroscience of Addiction Lab, Department of Psychology, Education and Child StudiesErasmus UniversityRotterdamthe Netherlands
| | - Anna E. Goudriaan
- Department of Psychiatry, Amsterdam Institute for Addiction ResearchAmsterdam UMC, University of AmsterdamAmsterdamthe Netherlands
| | - Robert Hester
- School of Psychological SciencesUniversity of MelbourneMelbourneAustralia
| | - Kent Hutchison
- Department of Psychology and NeuroscienceUniversity of Colorado BoulderBoulderColoradoUSA
| | - Chiang‐Shan R. Li
- Department of Psychiatry and of NeuroscienceYale University School of MedicineNew HavenConnecticutUSA
| | - Rocio Martin‐Santos
- Department of Psychiatry and Psychology, Hospital Clinic, IDIBAPS, CIBERSAM and Institute of NeuroscienceUniversity of BarcelonaBarcelonaSpain
| | - Reza Momenan
- Clinical NeuroImaging Research Core, Office of the Clinical DirectorNational Institute on Alcohol Abuse and AlcoholismBethesdaMarylandUSA
| | - Rajita Sinha
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Lianne Schmaal
- OrygenParkvilleAustralia
- Centre for Youth Mental HealthThe University of MelbourneMelbourneAustralia
| | - Nadia Solowij
- School of Psychology and Illawarra Health and Medical Research InstituteUniversity of WollongongWollongongAustralia
| | - Chao Suo
- Monash Biomedical ImagingMonash UniversityMelbourneAustralia
- Australian Characterisation Commons at Scale (ACCS) ProjectMonash eResearch CentreMelbourneAustralia
| | - Ruth J. van Holst
- Department of Psychiatry, Amsterdam Institute for Addiction ResearchAmsterdam UMC, University of AmsterdamAmsterdamthe Netherlands
| | - Dick J. Veltman
- Department of PsychiatryVU University Medical CenterAmsterdamthe Netherlands
| | - Murat Yücel
- BrainPark, Turner Institute for Brain and Mental HealthSchool of Psychological SciencesMelbourneAustralia
- Monash Biomedical ImagingMonash UniversityMelbourneAustralia
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics InstituteKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Patricia Conrod
- Department of PsychiatryUniversite de Montreal, CHU Ste Justine HospitalMontrealCanada
| | - Scott Mackey
- Department of PsychiatryUniversity of VermontBurlingtonVermontUSA
| | - Hugh Garavan
- Department of PsychiatryUniversity of VermontBurlingtonVermontUSA
| | - Paolo Brambilla
- Department of Neurosciences and Mental HealthFondazione IRCCS Ca'Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Valentina Lorenzetti
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioral and Health SciencesFaculty of Health Sciences, Australian Catholic UniversityFitzroyVictoriaAustralia
| |
Collapse
|
7
|
Wang W, Kang Y, Niu X, Zhang Z, Li S, Gao X, Zhang M, Cheng J, Zhang Y. Connectome-based predictive modeling of smoking severity using individualized structural covariance network in smokers. Front Neurosci 2023; 17:1227422. [PMID: 37547147 PMCID: PMC10400777 DOI: 10.3389/fnins.2023.1227422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/04/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Abnormal interactions among distributed brain systems are implicated in the mechanisms of nicotine addiction. However, the relationship between the structural covariance network, a measure of brain connectivity, and smoking severity remains unclear. To fill this gap, this study aimed to investigate the relationship between structural covariance network and smoking severity in smokers. Methods A total of 101 male smokers and 51 male non-smokers were recruited, and they underwent a T1-weighted anatomical image scan. First, an individualized structural covariance network was derived via a jackknife-bias estimation procedure for each participant. Then, a data-driven machine learning method called connectome-based predictive modeling (CPM) was conducted to infer smoking severity measured with Fagerström Test for Nicotine Dependence (FTND) scores using an individualized structural covariance network. The performance of CPM was evaluated using the leave-one-out cross-validation and a permutation testing. Results As a result, CPM identified the smoking severity-related structural covariance network, as indicated by a significant correlation between predicted and actual FTND scores (r = 0.23, permutation p = 0.020). Identified networks comprised of edges mainly located between the subcortical-cerebellum network and networks including the frontoparietal default model and motor and visual networks. Discussion These results identified smoking severity-related structural covariance networks and provided a new insight into the neural underpinnings of smoking severity.
Collapse
|
8
|
Jacobson MM, Jenkins LM, Feldman DA, Crane NA, Langenecker SA. Reduced connectivity of the cognitive control neural network at rest in young adults who had their first drink of alcohol prior to age 18. Psychiatry Res Neuroimaging 2023; 332:111642. [PMID: 37086604 PMCID: PMC10247408 DOI: 10.1016/j.pscychresns.2023.111642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/21/2023] [Accepted: 04/05/2023] [Indexed: 04/24/2023]
Abstract
The cognitive control network (CCN) is an important network responsible for performing and modulating executive functions. In adolescents, alcohol use has been associated with weaker cognitive control, higher reward sensitivity, and later-in-life alcohol problems. Given that the CCN continues to develop into young adulthood, it is important to understand relations between early alcohol use, the CCN, and reward networks. Participants included individuals 18-23 years without alcohol use disorder. Based upon self-reported age of first alcoholic drink, participants were split into two groups: Early (onset) Drinkers (first drink < age 18, N = 52) and Late (onset) Drinkers (first drink > age 18, N = 44). All participants underwent an 8-minute resting-state fMRI scan. Seed regions of interest included the anterior dorsolateral prefrontal cortex (DLPFC), amygdala, and ventral striatum. Early Drinkers demonstrated significant reduced connectivity of CCN regions, including bilateral anterior DLPFC, compared to Late Drinkers. There were no significant differences between Early and Late Drinkers in connectivity between reward and CCN regions. These results suggest that individuals who begin drinking alcohol earlier in life may have alterations in the development of the CCN; however, longitudinal research is necessary to determine whether lower connectivity precedes or follows early alcohol use, and any other relevant factors.
Collapse
Affiliation(s)
- Maci M Jacobson
- Department of Psychiatry, The University of Utah, United States; Interdisciplinary Neuroscience Program, The University of Utah, United States.
| | - Lisanne M Jenkins
- Department of Psychiatry and Behavioral Sciences, Northwestern University, United States; Department of Psychiatry, The University of Illinois at Chicago, United States
| | | | - Natania A Crane
- Department of Psychiatry, The University of Illinois at Chicago, United States
| | - Scott A Langenecker
- Department of Psychiatry, The University of Utah, United States; Interdisciplinary Neuroscience Program, The University of Utah, United States; Department of Psychiatry, The University of Illinois at Chicago, United States
| |
Collapse
|
9
|
Zeng J, You L, Sheng H, Luo Y, Yang X. The differential neural substrates for reward choice under gain-loss contexts and risk in alcohol use disorder: Evidence from a voxel-based meta-analysis. Drug Alcohol Depend 2023; 248:109912. [PMID: 37182355 DOI: 10.1016/j.drugalcdep.2023.109912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/15/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Making a risky decision is a complex process that involves the evaluation of both the values of the options and the associated risk level; this process is distinct from reward processing in gain versus loss contexts. Although disrupted reward processing in mesolimbic dopamine circuitry is suggested to underlie pathological incentive processing in patients with alcohol use disorder (AUD), the differential neural processes subserving these motivational tendencies for risk situations or gain/loss choices in decision-making have not been identified. METHODS To examine the common or distinct neural mechanisms in the evaluation of risk versus outcomes for AUD, we conducted two separate coordinate-based meta-analyses of functional neuroimaging studies by using Seed-Based d Mapping software to evaluate 13 studies investigating gain and loss processing and 10 studies investigating risky decision-making. RESULTS During gain and loss processing, relative to healthy controls, AUD patients showed reduced activation in the mesocortical-limbic circuit, including the orbital prefrontal cortex (OFC), dorsal striatum, insula, hippocampus, cerebellum, cuneus cortex and superior temporal gyrus, but hyperactivation in the inferior temporal gyrus and paracentral lobule (extending to the middle cingulate cortex (MCC) and precuneus). During decision-making under risk, AUD patients exhibited hypoactivity of the prefrontal and cingulate cortices, including the posterior cingulate cortex (extending to the MCC), middle frontal gyrus, medial prefrontal cortex, dorsolateral prefrontal cortex, OFC and anterior cingulate cortex. CONCLUSIONS Our results extend existing neurological evidence by showing that a reduced response in the mesocortical-limbic circuit is found in gain versus loss processing, with decreased responsivity in cortical regions in risk decision-making. Our results implicate dissociable neural circuit responses for gain-loss processing and risk decision-making, which contribute to a better understanding of the pathophysiological mechanism underlying nondrug incentive and risk processing in individuals with AUD.
Collapse
Affiliation(s)
- Jianguang Zeng
- School of Economics and Business Administration, Chongqing University, Chongqing, China
| | - Lantao You
- School of Economics and Business Administration, Chongqing University, Chongqing, China
| | - Haoxuan Sheng
- School of Public Policy and Administration, Chongqing University, Chongqing, China
| | - Ya Luo
- Department of Psychiatry, State Key Lab of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Xun Yang
- School of Public Policy and Administration, Chongqing University, Chongqing, China.
| |
Collapse
|
10
|
Zeng J, You L, Yang F, Luo Y, Yu S, Yan J, Liu M, Yang X. A meta-analysis of the neural substrates of monetary reward anticipation and outcome in alcohol use disorder. Hum Brain Mapp 2023; 44:2841-2861. [PMID: 36852619 PMCID: PMC10089105 DOI: 10.1002/hbm.26249] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 01/23/2023] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
The capacity to anticipate and detect rewarding outcomes is fundamental for the development of adaptive decision-making and goal-oriented behavior. Delineating the neural correlates of different stages of reward processing is imperative for understanding the neurobiological mechanism underlying alcohol use disorder (AUD). To examine the neural correlates of monetary anticipation and outcome in AUD patients, we performed two separate voxel-wise meta-analyses of functional neuroimaging studies, including 12 studies investigating reward anticipation and 7 studies investigating reward outcome using the monetary incentive delay task. During the anticipation stage, AUD patients displayed decreased activation in response to monetary cues in mesocortical-limbic circuits and sensory areas, including the ventral striatum (VS), insula, hippocampus, inferior occipital gyrus, supramarginal gyrus, lingual gyrus and fusiform gyrus. During the outcome stage, AUD patients exhibited reduced activation in the dorsal striatum, VS and insula, and increased activation in the orbital frontal cortex and medial temporal area. Our findings suggest that different activation patterns are associated with nondrug rewards during different reward processing stages, potentially reflecting a changed sensitivity to monetary reward in AUD.
Collapse
Affiliation(s)
- Jianguang Zeng
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Lantao You
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Fan Yang
- Department of Ultrasonography, West China Second University HospitalSichuan UniversityChengduChina
- Chengdu Chenghua District Maternal and Child Health HospitalSichuan UniversityChengduChina
| | - Ya Luo
- Department of Psychiatry, State Key Lab of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Shuxian Yu
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Jiangnan Yan
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Mengqi Liu
- Department of RadiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xun Yang
- School of Public AffairsChongqing UniversityChongqingChina
| |
Collapse
|
11
|
Wu F, Dong P, Wu G, Deng J, Gao X, Song X, Yuan J, Sun H. The disruption of white matter integrity of systemic striatal circuits in alcohol-dependent males with physiological cue reactivity. Addict Biol 2023; 28:e13273. [PMID: 37016754 DOI: 10.1111/adb.13273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/05/2023] [Accepted: 02/23/2023] [Indexed: 04/06/2023]
Abstract
Alcohol dependence (AD) is a chronic and relapsing disorder. Conditioned cues associated with the rewarding properties of drugs could trigger motivational/physiological reactions and render subjects vulnerable to relapse. Striatal circuit dysfunction has been implicated in alcohol addiction behaviours. However, little is known about the striatal tracts structural connectivity changes underlying cue induced reactivity in AD. In our present study, we recruited 51 patients with AD; 31 individuals had physiological response. We used seed-based classification by probabilistic tractography with nine target masks to explore the white matter integrity of striatal circuits in physiological responders (N = 31), non-responders (N = 20), and healthy controls (N = 27). Compared with healthy controls, physiological responders showed lower fractional anisotropy (FA) and/or higher mean diffusivity in the striatum-dorsolateral prefrontal cortex (dlPFC), striatum-ventral lateral prefrontal cortex, striatum-supplementary motor area (SMA), and striatum-insular. Considering age and smoking are potential nuisances to diffusion parameters, an analysis of covariance also was conducted and similar results were found. We also found the cue-induced physiological response was negatively associated with the FA of the striatum-SMA (r = -0.287; p = 0.045) and left striatum-dlPFC (r = -0.253; p = 0.079) in AD. In our study, we found abnormal integrity of striatal circuit structural connectivity in AD with physiological cue reactivity, especially trajectory from prefrontal cortex and insular. We also found the FA of striatal tracks was negatively associated with the degree of cue reactivity. Our findings provide further evidence for reduced white matter integrity of striatal circuits for cue reactivity in male individuals with AD.
Collapse
Affiliation(s)
- Fei Wu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Ping Dong
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Guowei Wu
- Chinese Institute for Brain Research, Beijing, China
| | - Jiahui Deng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Xuejiao Gao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Xiaopeng Song
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Junliang Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Hongqiang Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| |
Collapse
|
12
|
Zhao Y, Skandali N, Bethlehem RAI, Voon V. Mesial Prefrontal Cortex and Alcohol Misuse: Dissociating Cross-sectional and Longitudinal Relationships in UK Biobank. Biol Psychiatry 2022; 92:907-916. [PMID: 35589437 DOI: 10.1016/j.biopsych.2022.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 02/24/2022] [Accepted: 03/12/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alcohol misuse is a major global public health issue. The disorder is characterized by aberrant neural networks interacting with environment and genetics. Dissecting the neural substrates and functional networks that relate to longitudinal changes in alcohol use from those that relate to alcohol misuse cross-sectionally is important to elucidate therapeutic approaches. METHODS To assess how neuroimaging data, including T1, resting-state functional magnetic resonance imaging, and diffusion-weighted imaging, relate to alcohol misuse cross-sectionally and longitudinally in the UK Biobank, this study analyzed range of alcohol misuse in a population-based normative sample of 24,784 participants, ages 45 to 81 years old, in a cross-sectional analysis and a sample of 3070 participants in a longitudinal analysis 2 years later. RESULTS Cross-sectional analysis showed that alcohol use is associated with a reduction in dorsal anterior cingulate cortex and dorsomedial prefrontal cortex gray matter concentration and functional resting-state connectivity (nodal degree: t24,422 = -12.99, p < 1 × 10-17). Reduced dorsal anterior cingulate cortex/dorsomedial prefrontal cortex functional connections to the ventrolateral prefrontal cortex, amygdala, and striatum relate to greater alcohol use. In a longitudinal analysis, higher resting-state nodal degree (t3036 = -3.27, p = .0011) and T1 gray matter concentration in the ventromedial prefrontal cortex relate to reduced alcohol intake frequency 2 years later. Higher ventromedial prefrontal cortex and frontoparietal executive network functional connectivity is associated with lower subsequent drinking longitudinally. CONCLUSIONS Dorsal versus ventromedial prefrontal regions are differentially related to alcohol misuse cross-sectionally or longitudinally in a large UK Biobank normative dataset. Our study provides a comprehensive understanding of the neurobiological substrates of alcohol use as a state or prospectively, thereby providing potential targets for clinical treatment.
Collapse
Affiliation(s)
- Ying Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom.
| | - Nikolina Skandali
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom; Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, United Kingdom
| | | | - Valerie Voon
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
13
|
Soleimani G, Towhidkhah F, Oghabian MA, Ekhtiari H. DLPFC stimulation alters large-scale brain networks connectivity during a drug cue reactivity task: A tDCS-fMRI study. Front Syst Neurosci 2022; 16:956315. [PMID: 36276607 PMCID: PMC9582757 DOI: 10.3389/fnsys.2022.956315] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/11/2022] [Indexed: 11/20/2022] Open
Abstract
Transcranial direct current stimulation (tDCS) is a promising intervention for reducing craving/consumption in individuals with substance use disorders. However, its exact mechanism of action has not yet been well explored. We aimed to examine the network-based effects of tDCS while people with methamphetamine use disorders (MUDs) were exposed to drug cues. In a randomized, double-blind sham-controlled trial with a crossover design, 15 participants with MUDs were recruited to receive 20 min of active/sham tDCS with an anode/cathode over F4/F3. MRI data, including structural and task-based functional MRI during a standard drug cue-reactivity task, were collected immediately before and after stimulation sessions. Craving scores were also recorded before and after MRI scans. Individualized head models were generated to determine brain regions with strong electric fields (EFs). Using atlas-based parcellation of head models, averaged EFs were extracted from the main nodes of three large-scale networks that showed abnormalities in MUDs; executive control (ECN), default mode (DMN), and ventral attention (VAN) networks. Main nodes with high EF intensity were used as seed regions for task-based functional connectivity (FC) [using generalized psychophysiological interaction (gPPI)] and activity [using a general linear model (GLM)] calculations. Subjective craving showed a significant reduction in immediate craving after active (-15.42 ± 5.42) compared to sham (-1 ± 2.63). In seed-to-whole brain results, the PFC node in ECN showed an enhanced PPI connectivity with precuneus and visual cortex; the cluster center in MNI (6, -84, -12); the PFC node in DMN showed a decreased PPI connectivity with contralateral parietal cortex;(-48, -60, 46). ROI-to-ROI results showed increased PPI connectivity within/between ECN-VAN while connectivity between ECN-DMN decreased. In line with connectivity, functional activity in the right PFC node in DMN decreased after tDCS while activity in PFC nodes of ECN/VAN increased. EF calculations in PFC nodes revealed that EF in DMN was outward, while the direction of EFs was inward in ECN/VAN. This study provides new insight into neural circuitry underlying MUDs that can be modulated by tDCS at the network level and specifically suggests that bilateral tDCS increases cortical excitability in ECN and VAN, while it has opposite effects on DMN that may be related to the direction of EFs.
Collapse
Affiliation(s)
- Ghazaleh Soleimani
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
- Iranian National Center for Addiction Studies, Tehran University of Medical Science, Tehran, Iran
| | - Farzad Towhidkhah
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Mohammad Ali Oghabian
- Neuroimaging and Analysis Group, Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Ekhtiari
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, United States
- Laureate Institute for Brain Research, Tulsa, OK, United States
| |
Collapse
|
14
|
Klein S, Krikova K, Antons S, Brand M, Klucken T, Stark R. Reward Responsiveness, Learning, and Valuation Implicated in Problematic Pornography Use — a Research Domain Criteria Perspective. CURRENT ADDICTION REPORTS 2022. [DOI: 10.1007/s40429-022-00423-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Abstract
Purpose of Review
Problematic pornography use (PPU) describes a pattern of behavior characterized by excessive time spent using or thinking about pornography and continued use despite negative consequences. To help advance the understanding of transdiagnostic underlying psychological and neurobiological mechanisms in PPU, we aim to review existing evidence on these mechanisms focusing on positive valence systems within the transdiagnostic Research Domain Criteria (RDoC) framework.
Recent Findings
Reward anticipation processes seem to be increased in individuals with PPU symptoms when they anticipate sexual stimuli compared with other rewards. Studies further suggest that the initial neural and attentional responses to sexual rewards compared with different control stimuli are also increased in individuals with PPU symptoms, as are conditioned responses in sexual reward learning paradigms. Sexual reward valuation studies point towards an increased neural value differentiation with increasing PPU symptoms.
Summary
The current state of evidence indicates that positive valence systems are altered in persons with PPU. This framework of organizing evidence may aid in elucidating PPU development and maintenance as well as planning future studies.
Collapse
|
15
|
Smart K, Worhunsky PD, Scheinost D, Angarita GA, Esterlis I, Carson RE, Krystal JH, O'Malley SS, Cosgrove KP, Hillmer AT. Multimodal neuroimaging of metabotropic glutamate 5 receptors and functional connectivity in alcohol use disorder. Alcohol Clin Exp Res 2022; 46:770-782. [PMID: 35342968 PMCID: PMC9117461 DOI: 10.1111/acer.14816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/15/2022] [Accepted: 03/19/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND People recovering from alcohol use disorder (AUD) show altered resting brain connectivity. The metabotropic glutamate 5 (mGlu5) receptor is an important regulator of synaptic plasticity potentially linked with synchronized brain activity and a target of interest in treating AUD. The goal of this work was to assess potential relationships of brain connectivity at rest with mGlu5 receptor availability in people with AUD at two time points early in abstinence. METHODS Forty-eight image data sets were acquired with a multimodal neuroimaging battery that included resting-state functional magnetic resonance imaging (fMRI) and mGlu5 receptor positron emission tomography (PET) with the radiotracer [18 F]FPEB. Participants with AUD (n = 14) were scanned twice, at approximately 1 and 4 weeks after beginning supervised abstinence. [18 F]FPEB PET results were published previously. Primary comparisons of fMRI outcomes were performed between the AUD group and healthy controls (HCs; n = 23) and assessed changes over time within the AUD group. Relationships between resting-state connectivity measures and mGlu5 receptor availability were explored within groups. RESULTS Compared to HCs, global functional connectivity of the orbitofrontal cortex was higher in the AUD group at 4 weeks of abstinence (p = 0.003), while network-level functional connectivity within the default mode network (DMN) was lower (p < 0.04). Exploratory multimodal analyses showed that mGlu5 receptor availability was correlated with global connectivity across all brain regions (HCs, r = 0.41; AUD group at 1 week of abstinence, r = 0.50 and at 4 weeks, r = 0.46; all p < 0.0001). Furthermore, a component of cortical and striatal mGlu5 availability was correlated with connectivity between the DMN and salience networks in HCs (r = 0.60, p = 0.003) but not in the AUD group (p > 0.3). CONCLUSIONS These preliminary findings of altered global and network connectivity during the first month of abstinence from drinking may reflect the loss of efficient network function, while exploratory relationships with mGlu5 receptor availability suggest a potential glutamatergic relationship with network coherence.
Collapse
Affiliation(s)
- Kelly Smart
- Yale PET Center, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Patrick D Worhunsky
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Dustin Scheinost
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Gustavo A Angarita
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Richard E Carson
- Yale PET Center, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - John H Krystal
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Kelly P Cosgrove
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ansel T Hillmer
- Yale PET Center, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| |
Collapse
|
16
|
Pandey AK, Ardekani BA, Byrne KNH, Kamarajan C, Zhang J, Pandey G, Meyers JL, Kinreich S, Chorlian DB, Kuang W, Stimus AT, Porjesz B. Statistical Nonparametric fMRI Maps in the Analysis of Response Inhibition in Abstinent Individuals with History of Alcohol Use Disorder. Behav Sci (Basel) 2022; 12:bs12050121. [PMID: 35621418 PMCID: PMC9137506 DOI: 10.3390/bs12050121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/26/2022] Open
Abstract
Inhibitory impairments may persist after abstinence in individuals with alcohol use disorder (AUD). Using traditional statistical parametric mapping (SPM) fMRI analysis, which requires data to satisfy parametric assumptions often difficult to satisfy in biophysical system as brain, studies have reported equivocal findings on brain areas responsible for response inhibition, and activation abnormalities during inhibition found in AUD persist after abstinence. Research is warranted using newer analysis approaches. fMRI scans were acquired during a Go/NoGo task from 30 abstinent male AUD and 30 healthy control participants with the objectives being (1) to characterize neuronal substrates associated with response inhibition using a rigorous nonparametric permutation-based fMRI analysis and (2) to determine whether these regions were differentially activated between abstinent AUD and control participants. A blood oxygen level dependent contrast analysis showed significant activation in several right cortical regions and deactivation in some left cortical regions during successful inhibition. The largest source of variance in activation level was due to group differences. The findings provide evidence of cortical substrates employed during response inhibition. The largest variance was explained by lower activation in inhibition as well as ventral attentional cortical networks in abstinent individuals with AUD, which were not found to be associated with length of abstinence, age, or impulsiveness.
Collapse
Affiliation(s)
- Ashwini Kumar Pandey
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
- Correspondence:
| | - Babak Assai Ardekani
- Center for Biomedical Imaging and Neuromodulation, The Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; (B.A.A.); (K.N.-H.B.)
| | - Kelly Nicole-Helen Byrne
- Center for Biomedical Imaging and Neuromodulation, The Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; (B.A.A.); (K.N.-H.B.)
| | - Chella Kamarajan
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
| | - Jian Zhang
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
| | - Gayathri Pandey
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
| | - Jacquelyn Leigh Meyers
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
| | - Sivan Kinreich
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
| | - David Balin Chorlian
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
| | - Weipeng Kuang
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
| | - Arthur T. Stimus
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
| | - Bernice Porjesz
- Henri Begleiter Neurodynamics Laboratory, Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #1203, Brooklyn, NY 11203, USA; (C.K.); (J.Z.); (G.P.); (J.L.M.); (S.K.); (D.B.C.); (W.K.); (A.T.S.); (B.P.)
| |
Collapse
|
17
|
Almeida‐Antunes N, Antón‐Toro L, Crego A, Rodrigues R, Sampaio A, López‐Caneda E. "It's a beer!": Brain functional hyperconnectivity during processing of alcohol-related images in young binge drinkers. Addict Biol 2022; 27:e13152. [PMID: 35229944 DOI: 10.1111/adb.13152] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/12/2022]
Abstract
Alcohol attentional bias has been pointed as a major marker of alcohol misuse. Recent evidence has revealed that brain functional connectivity (FC) may be a valuable index of the brain networks' integrity in young binge drinkers (BDs). However, there is no study to date examining the FC networks linked to the processing of alcohol-related images in this population. The present study aimed to explore the FC signatures underlying alcohol attention bias in young BDs. Thus, electroencephalographic (EEG) activity was recorded in 54 college students (55.5% females; 27 non/low-drinkers and 27 BDs) while performing a visual alcohol cue-reactivity task. We evaluated whole-brain FC profiles during the processing of alcoholic and non-alcoholic cues, as well as their potential relationship with craving and severity of alcohol use. Results showed that, at the behavioural level, BDs rated alcohol-related images as more pleasant/attractive than non/low-drinkers. Furthermore, at the electrophysiological level, BDs exhibited increased beta-band FC-particularly in the fronto-parieto-occipital network-when processing alcoholic cues. Conversely, they displayed reduced theta-band FC relatively to non/low-drinkers for non-alcoholic images. These hyper-/hypo-connectivity patterns were associated with higher alcohol craving levels. Findings are congruent with previous neurofunctional studies reporting an attentional bias towards alcohol-related information in BDs. These results may have important clinical implications as this neural reactivity to alcoholic cues may contribute to the maintenance and/or escalation of the drinking pattern. Finally, the present study constitutes the first evidence showing that FC networks may be a sensitive indicator to alcohol attentional bias in BDs.
Collapse
Affiliation(s)
- Natália Almeida‐Antunes
- Psychological Neuroscience Laboratory (PNL), Research Center in Psychology (CIPsi), School of Psychology University of Minho Braga Portugal
| | - Luis Antón‐Toro
- Department of Experimental Psychology Complutense University of Madrid (UCM) Madrid Spain
- Laboratory for Cognitive and Computational Neuroscience (UCM ‐ UPM) Center for Biomedical Technology (CBT) Madrid Spain
| | - Alberto Crego
- Psychological Neuroscience Laboratory (PNL), Research Center in Psychology (CIPsi), School of Psychology University of Minho Braga Portugal
| | - Rui Rodrigues
- Psychological Neuroscience Laboratory (PNL), Research Center in Psychology (CIPsi), School of Psychology University of Minho Braga Portugal
| | - Adriana Sampaio
- Psychological Neuroscience Laboratory (PNL), Research Center in Psychology (CIPsi), School of Psychology University of Minho Braga Portugal
| | - Eduardo López‐Caneda
- Psychological Neuroscience Laboratory (PNL), Research Center in Psychology (CIPsi), School of Psychology University of Minho Braga Portugal
| |
Collapse
|
18
|
Le TM, Malone T, Li CSR. Positive alcohol expectancy and resting-state functional connectivity of the insula in problem drinking. Drug Alcohol Depend 2022; 231:109248. [PMID: 34998254 PMCID: PMC8881788 DOI: 10.1016/j.drugalcdep.2021.109248] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 02/03/2023]
Abstract
Positive alcohol expectancy (AE), a significant predictor of excessive alcohol consumption, is associated with heightened drinking motivation and reduced control. As the insula interacts with the limbic and prefrontal structures to integrate stimulus saliency, interoception, and cognitive control, the region may play a unique role in modulating AE. Here, we examined resting-state functional connectivity of the right and left insula in relation to AE in 180 adult drinkers. Whole-brain multiple regressions and path analysis were performed to delineate the inter-relationship between AE, insular connectivity, and drinking severity. We found that heightened AE was associated with diminished right insular connectivity with regions involved in negative emotion processing and self-control, including the amygdala, putamen, and ventromedial prefrontal cortex. In contrast, there was a positive relationship between AE and right insular connectivity with regions implicated in motivated responses to alcohol stimuli, including the superior parietal lobule, postcentral and superior frontal gyri. Path analysis showed that the two sets of right insular connectivity exhibited opposing associations with AE and that their net strength (i.e., "control minus motivation") was negatively correlated with AE and drinking severity. Analyses of the left insula seed, in contrast, did not yield regional connectivity in significant correlation with AE. These findings highlight the roles of right insula connectivity in motivational and regulatory processes that may differentially modulate drinking behavior. Recruitment of the motivational circuit and/or disengagement of the affective control circuit would be associated with heightened AE and heavier alcohol consumption.
Collapse
Affiliation(s)
- Thang M. Le
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA,Correspondence: Thang M. Le, Ph.D., Connecticut Mental Health Center, S105, 34 Park Street, New Haven, CT 06519-1109, USA, , Phone: 203-974-7360
| | - Tessa Malone
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Chiang-Shan R. Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA,Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06520, USA,Wu Tsai Institute, Yale University, New Haven, CT, USA
| |
Collapse
|
19
|
Pfeifer P, Sebastian A, Buchholz HG, Kaller CP, Gründer G, Fehr C, Schreckenberger M, Tüscher O. Prefrontal and striatal dopamine D 2/D 3 receptors correlate with fMRI BOLD activation during stopping. Brain Imaging Behav 2022; 16:186-198. [PMID: 34403039 PMCID: PMC8825403 DOI: 10.1007/s11682-021-00491-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2021] [Indexed: 11/02/2022]
Abstract
D2-like dopamine receptors in animals and humans have been shown to be linked to impulsive behaviors that are highly relevant for several psychiatric disorders. Here, we investigate the relationship between the fronto-striatal D2/D3 dopamine receptor availability and response inhibition in a selected population of healthy OPRM1 G-allele carriers. Twenty-two participants successively underwent blood-oxygen level dependent functional magnetic resonance imaging (fMRI) while performing a stop-signal task and a separate positron emission tomography (PET) scan. Striatal and extrastriatal D2/D3 dopamine receptor availability was measured using the radiotracer [18F]fallypride. Caudate D2/D3 dopamine receptor availability positively correlated with stopping-related fronto-striatal fMRI activation. In addition, right prefrontal D2/D3 dopamine receptor availability correlated positively with stopping-related striatal fMRI BOLD signal. Our study partially replicates previous findings on correlations between striatal D2/D3 dopamine receptor availability and response inhibition in a population selected for its genetic determination of dopamine response to alcohol and as a modulator of impulse control via the endogenous opioid system. We confirm the important role of D2/D3 dopamine receptor availability in the fronto-striatal neural circuit for response inhibition. Moreover, we extend previous findings suggesting that dopamine receptor availability in the right inferior frontal cortex, a crucial region of the stopping network, is also strongly associated with stopping-related striatal fMRI activity in healthy OPRM1 G-allele carriers.
Collapse
Affiliation(s)
- Philippe Pfeifer
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland.
| | - Alexandra Sebastian
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
- Leibniz Institute for Resilience Research, Wallstraße 7, 55122, Mainz, Germany
| | - Hans Georg Buchholz
- Department of Nuclear Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Christoph P Kaller
- Department of Neurology and Neuroscience, University Medical Centre Freiburg, Freiburg, Germany
- Freiburg Brain Imaging Centre, University Medical Centre Freiburg, Freiburg, Germany
- Brain Links-BrainTools Cluster of Excellence, University Medical Centre Freiburg, Freiburg, Germany
| | - Gerhard Gründer
- Department of Molecular Neuroimaging, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Christoph Fehr
- Department for Psychiatry und Psychotherapy, Vitos Clinic for Psychiatry und Psychotherapy Hadamar/Weilmünster, Mönchberg 8, 65589, Hadamar, Germany
| | - Mathias Schreckenberger
- Department of Nuclear Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Oliver Tüscher
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
- Leibniz Institute for Resilience Research, Wallstraße 7, 55122, Mainz, Germany
| |
Collapse
|
20
|
Wu F, Dong P, Wu G, Deng J, Ni Z, Gao X, Li P, Li B, Yuan J, Sun H. Impulsive trait mediates the relationship between white matter integrity of prefrontal-striatal circuits and the severity of dependence in alcoholism. Front Psychiatry 2022; 13:985948. [PMID: 36159935 PMCID: PMC9490322 DOI: 10.3389/fpsyt.2022.985948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/15/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Alcohol dependence (AD) remains one of the major public health concerns. Impulsivity plays a central role in the transfer from recreational alcohol use to dependence and relapse. White matter dysfunction has been implicated in alcohol addiction behaviors and impulsivity. However, little is known about the role of systematic striatal structural connections underlying the mechanism of impulsive traits in AD. METHODS In our study, we used seed-based classification by probabilistic tractography with five target masks of striatal circuits to explore the differences in white matter integrity (fractional anisotropy, FA) in AD male patients (N = 51) and healthy controls (N = 27). We mainly explored the correlation between FA of the striatal circuits and impulsive traits (Barratt Impulsiveness Scale, BIS-11), and the mediation role of impulsivity in white matter integrity and the severity of alcohol dependence. RESULTS Compared with healthy controls, AD showed much lower FA in the left and right striatum-supplementary motor area (SMA) and left striatum-amygdala. We also found the decreased FA of right striatum-vlPFC was correlated with higher impulsivity. Besides, the relationship between reduced FA of right striatum-vlPFC and severity of dependence could be mediated by impulsivity. CONCLUSION In our study, we found disrupted white matter integrity in systematic striatal circuits in AD and the decreased FA of right striatum-vlPFC was correlated with higher impulsivity in AD. Our main findings provide evidence for reduced white matter integrity of systematic striatal circuits and the underlying mechanisms of impulsivity in male AD individuals.
Collapse
Affiliation(s)
- Fei Wu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Ping Dong
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Guowei Wu
- Chinese Institute for Brain Research, Beijing, China
| | - Jiahui Deng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Zhaojun Ni
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Xuejiao Gao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Peng Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Bing Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Junliang Yuan
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Hongqiang Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| |
Collapse
|
21
|
Picci G, Fishbein DH, VanMeter JW, Rose EJ. Effects of OPRM1 and DRD2 on brain structure in drug-naïve adolescents: Genetic and neural vulnerabilities to substance use. Psychopharmacology (Berl) 2022; 239:141-152. [PMID: 34816289 PMCID: PMC8776605 DOI: 10.1007/s00213-021-06030-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/15/2021] [Indexed: 01/03/2023]
Abstract
Genetic variants in the opioid receptor mu 1 (OPRM1) and dopamine receptor d2 (DRD2) genes are implicated in behavioral phenotypes related to substance use disorders (SUD). Despite associations among OPRM1 (rs179971) and DRD2 (rs6277) genes and structural alterations in neural reward pathways implicated in SUDs, little is known about the contribution of risk-related gene variants to structural neurodevelopment. In a 3-year longitudinal study of initially SU-naïve adolescents (N = 129; 70 females; 11-14 years old), participants underwent an MRI structural scan at baseline and provided genetic assays for OPRM1 and DRD2 with SU behavior assessed during follow-up visits. Baseline differences in key reward-related brain regions (i.e., bilateral caudate and cingulate cortex) were detected in those with genetic liability for SU in OPRM1 who went onto engage in SU at subsequent waves of data collection. In addition, main effects of OPRM1, DRD2, and SU were related to variability in structure of the putamen, anterior cingulate, and nucleus accumbens, respectively. These data provide preliminary evidence that genetic risk factors interact with future SU to confer structural variability prior to SU in regions commonly implicated in risk for SU and the development of SUDs.
Collapse
Affiliation(s)
- Giorgia Picci
- Institute for Human Neuroscience, Boys Town National Research Hospital, 378 Bucher Drive, Boys Town, NE, 68010, USA.
| | - Diana H Fishbein
- Department of Human Development and Family Studies, Program for Translational Research On Adversity and Neurodevelopment (P-TRAN), Edna Bennett Pierce Prevention Research Center, Penn State University, 218 Health and Human Development Building, University Park, PA, 16802, USA
| | - John W VanMeter
- Center for Functional and Molecular Imaging, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC, 20057, USA
| | - Emma J Rose
- Program for Translational Research On Adversity and Neurodevelopment (P-TRAN), Edna Bennett Pierce Prevention Research Center, Penn State University, 310A Biobehavioral Health Building, University Park, PA, 16802, USA
| |
Collapse
|
22
|
Moulin V, Framorando D, Gasser J, Dan-Glauser E. The Link Between Cannabis Use and Violent Behavior in the Early Phase of Psychosis: The Potential Role of Impulsivity. Front Psychiatry 2022; 13:746287. [PMID: 35392388 PMCID: PMC8980530 DOI: 10.3389/fpsyt.2022.746287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Recently, the literature has shown that Cannabis Use (CU) was a risk factor for Violent Behavior (VB) in patients with psychosis, and those in the early phase of psychosis (EPP). These findings are relevant because of the high prevalence of CU in this EPP, and the potential for prevention during this phase of illness. However, there is still a lack of clear explanations, supported by empirical evidence, about what underlies the link between CU and VB against other. METHOD This viewpoint reviews the scientific literature on the link between CU and VB, and the involvement of impulsivity in this relationship. This last point will be addressed at clinical and neurobiological levels. RESULTS Recent studies confirmed that CU is particularly high in the EPP, and is a risk factor for VB in the EPP and schizophrenia. Studies have also shown that impulsivity is a risk factor for VB in psychosis, is associated with CU, and may mediate the link between CU and VB. Research suggests a neurobiological mechanism, as CU affects the structures and function of frontal areas, known to play a role in impulsive behavior. CONCLUSION Scientific evidence support the hypothesis of an involvement of impulsivity as a variable that could mediate the link between CU and aggression, particularly, when CU has an early onset. However, this hypothesis should be confirmed with longitudinal studies and by taking into account confounding factors. The studies highlight the relevance of early prevention in the EPP, in addition to interventions focusing on psychotic disorders.
Collapse
Affiliation(s)
- Valerie Moulin
- Unit for Research in Legal Psychiatry and Psychology, Institute of Forensic Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - David Framorando
- Unit for Research in Legal Psychiatry and Psychology, Institute of Forensic Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Jacques Gasser
- Unit for Research in Legal Psychiatry and Psychology, Institute of Forensic Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Elise Dan-Glauser
- Institute of Psychology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
23
|
Ghin F, Beste C, Stock AK. Neurobiological mechanisms of control in alcohol use disorder - moving towards mechanism-based non-invasive brain stimulation treatments. Neurosci Biobehav Rev 2021; 133:104508. [PMID: 34942268 DOI: 10.1016/j.neubiorev.2021.12.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 12/14/2021] [Accepted: 12/19/2021] [Indexed: 12/13/2022]
Abstract
Alcohol use disorder (AUD) is characterized by excessive habitual drinking and loss of control over alcohol intake despite negative consequences. Both of these aspects foster uncontrolled drinking and high relapse rates in AUD patients. Yet, common interventions mostly focus on the phenomenological level, and prioritize the reduction of craving and withdrawal symptoms. Our review provides a mechanistic understanding of AUD and suggests alternative therapeutic approaches targeting the mechanisms underlying dysfunctional alcohol-related behaviours. Specifically, we explain how repeated drinking fosters the development of rigid drinking habits and is associated with diminished cognitive control. These behavioural and cognitive effects are then functionally related to the neurobiochemical effects of alcohol abuse. We further explain how alterations in fronto-striatal network activity may constitute the neurobiological correlates of these alcohol-related dysfunctions. Finally, we discuss limitations in current pharmacological AUD therapies and suggest non-invasive brain stimulation (like TMS and tDCS interventions) as a potential addition/alternative for modulating the activation of both cortical and subcortical areas to help re-establish the functional balance between controlled and automatic behaviour.
Collapse
Affiliation(s)
- Filippo Ghin
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; University Neuropsychology Center, Faculty of Medicine, TU Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; University Neuropsychology Center, Faculty of Medicine, TU Dresden, Germany
| | - Ann-Kathrin Stock
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; University Neuropsychology Center, Faculty of Medicine, TU Dresden, Germany; Biopsychology, Faculty of Psychology, TU Dresden, Dresden, Germany.
| |
Collapse
|
24
|
Nett KE, LaLumiere RT. Infralimbic cortex functioning across motivated behaviors: Can the differences be reconciled? Neurosci Biobehav Rev 2021; 131:704-721. [PMID: 34624366 PMCID: PMC8642304 DOI: 10.1016/j.neubiorev.2021.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/10/2021] [Accepted: 10/02/2021] [Indexed: 10/20/2022]
Abstract
The rodent infralimbic cortex (IL) is implicated in higher order executive functions such as reward seeking and flexible decision making. However, the precise nature of its role in these processes is unclear. Early evidence indicated that the IL promotes the extinction and ongoing inhibition of fear conditioning and cocaine seeking. However, evidence spanning other behavioral domains, such as natural reward seeking and habit-based learning, suggests a more nuanced understanding of IL function. As techniques have advanced and more studies have examined IL function, identifying a unifying explanation for its behavioral function has become increasingly difficult. Here, we discuss evidence of IL function across motivated behaviors, including associative learning, drug seeking, natural reward seeking, and goal-directed versus habit-based behaviors, and emphasize how context-specific encoding and heterogeneous IL neuronal populations may underlie seemingly conflicting findings in the literature. Together, the evidence suggests that a major IL function is to facilitate the encoding and updating of contingencies between cues and behaviors to guide subsequent behaviors.
Collapse
Affiliation(s)
- Kelle E Nett
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA 52242, United States.
| | - Ryan T LaLumiere
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA 52242, United States; Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, United States; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, United States
| |
Collapse
|
25
|
Alderson Myers AB, Arienzo D, Molnar SM, Marinkovic K. Local and network-level dysregulation of error processing is associated with binge drinking. NEUROIMAGE-CLINICAL 2021; 32:102879. [PMID: 34768146 PMCID: PMC8591397 DOI: 10.1016/j.nicl.2021.102879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 01/22/2023]
Abstract
Go/NoGo performance does not differ between binge (BDs) and light drinkers. BDs show greater BOLD activity to inhibition errors primarily in prefrontal areas. Greater functional connectivity in the frontal cortex correlates with drinking. Observed increase in error-related activity may serve a compensatory role. This is consistent with allostatic hyperexcitability reflecting neuroadaptation.
Binge drinking refers to the pattern of alcohol consumption that brings blood alcohol levels to or above legal intoxication levels. Commonly practiced by young adults, it is associated with neurofunctional alterations, raising health-related concerns. Executive deficits may contribute to the inability to refrain from excessive alcohol intake. As a facet of cognitive control, error processing allows for flexible modification of behavior to optimize future outcomes. It is highly relevant to addiction research, as a failure to inhibit excessive drinking results in relapses, which is a hallmark of alcohol use disorder. However, research on local and system-level neural underpinnings of inhibition failures as a function of binge drinking is limited. To address these gaps, functional magnetic resonance imaging (fMRI) was used to examine local changes and interregional functional connectivity during response inhibition errors on a Go/NoGo task. Young adult binge drinkers (BDs) performed equally well as light drinkers (LDs), a group of demographically matched individuals who drink regularly but in low-risk patterns. In contrast, BDs exhibited greater fMRI activity to inhibition errors contrasted with correct NoGo trials in the rostral anterior (rACC) and posterior cingulate cortices (PCC), as well as right middle frontal gyrus (R-MFG). Furthermore, BDs showed increased connectivity between the rACC and right lateral prefrontal cortex, in addition to greater connectivity between the R-MFG and the left ventrolateral and superior frontal cortices. Imaging indices were positively correlated only with alcohol-related measures, but not with those related to moods, disposition, or cognitive capacity. Taken together, greater error-related activity and expanded functional connectivity among prefrontal regions may serve a compensatory role to maintain efficiency of inhibitory control. Aligned with prominent models of addiction, these findings accentuate the importance of top-down control in maintaining low-risk drinking levels. They provide insight into potentially early signs of deteriorating cognitive control functions in BDs and may help guide intervention strategies aimed at preventing excessive drinking habits.
Collapse
Affiliation(s)
- Austin B Alderson Myers
- Department of Psychology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA.
| | - Donatello Arienzo
- Department of Psychology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA.
| | - Sean M Molnar
- Department of Psychology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA.
| | - Ksenija Marinkovic
- Department of Psychology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA; Department of Radiology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA.
| |
Collapse
|
26
|
Wang M, Zheng H, Zhou W, Jiang Q, Dong G. Persistent dependent behaviour is accompanied by dynamic switching between the ventral and dorsal striatal connections in internet gaming disorder. Addict Biol 2021; 26:e13046. [PMID: 33957705 DOI: 10.1111/adb.13046] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/11/2021] [Accepted: 04/12/2021] [Indexed: 01/02/2023]
Abstract
Cross-sectional studies have suggested that functional heterogeneity within the striatum in individuals with addictive behaviours may involve the transition from ventral to dorsal partitions; however, due to limitations of the cross-sectional design, whether the contribution of this transition to addiction was confused by individual differences remains unclear, especially for internet gaming disorder (IGD). Longitudinal functional magnetic resonance imaging (fMRI) data from 22 IGD subjects and 18 healthy controls were collected at baseline and more than 6 months later. We examined the connectivity features of subregions within the striatum between these two scans. Based on the results, we further performed dynamic causal modelling to explore the directional effect between regions and used these key features for data classification in machine learning to test the replicability of the results. Compared with controls, IGD subjects exhibited decreased functional connectivity between the left dorsal striatum (putamen) and the left insula, whereas connectivity between the right ventral striatum (nucleus accumbens [Nacc]) and the left insula was relatively stable over time. An inhibitory effective connectivity from the left putamen to the right Nacc was found in IGD subjects during the follow-up scan. Using the above features, the classification accuracy of the training model developed with the follow-up was better than that of the model based on the initial scan. Persistent IGD status was accompanied by a switch in the locus of control within the striatum, which provided new insights into association between IGD and drug addiction.
Collapse
Affiliation(s)
- Min Wang
- Center for Cognition and Brain Disorders The Affiliated Hospital of Hangzhou Normal University Hangzhou China
| | - Hui Zheng
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Weiran Zhou
- Center for Cognition and Brain Disorders The Affiliated Hospital of Hangzhou Normal University Hangzhou China
| | - Qing Jiang
- Department of Psychology Zhejiang Normal University Jinhua China
| | - Guang‐Heng Dong
- Center for Cognition and Brain Disorders The Affiliated Hospital of Hangzhou Normal University Hangzhou China
- Institute of Psychological Science Hangzhou Normal University Hangzhou China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments Hangzhou China
| |
Collapse
|
27
|
Fredriksson I, Tsai PJ, Shekara A, Duan Y, Applebey SV, Lu H, Bossert JM, Shaham Y, Yang Y. Orbitofrontal cortex and dorsal striatum functional connectivity predicts incubation of opioid craving after voluntary abstinence. Proc Natl Acad Sci U S A 2021; 118:e2106624118. [PMID: 34675078 PMCID: PMC8639358 DOI: 10.1073/pnas.2106624118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
We recently introduced a rat model of incubation of opioid craving after voluntary abstinence induced by negative consequences of drug seeking. Here, we used resting-state functional MRI to determine whether longitudinal functional connectivity changes in orbitofrontal cortex (OFC) circuits predict incubation of opioid craving after voluntary abstinence. We trained rats to self-administer for 14 d either intravenous oxycodone or palatable food. After 3 d, we introduced an electric barrier for 12 d that caused cessation of reward self-administration. We tested the rats for oxycodone or food seeking under extinction conditions immediately after self-administration training (early abstinence) and after electric barrier exposure (late abstinence). We imaged their brains before self-administration and during early and late abstinence. We analyzed changes in OFC functional connectivity induced by reward self-administration and electric barrier-induced abstinence. Oxycodone seeking was greater during late than early abstinence (incubation of oxycodone craving). Oxycodone self-administration experience increased OFC functional connectivity with dorsal striatum and related circuits that was positively correlated with incubated oxycodone seeking. In contrast, electric barrier-induced abstinence decreased OFC functional connectivity with dorsal striatum and related circuits that was negatively correlated with incubated oxycodone seeking. Food seeking was greater during early than late abstinence (abatement of food craving). Food self-administration experience and electric barrier-induced abstinence decreased or maintained functional connectivity in these circuits that were not correlated with abated food seeking. Opposing functional connectivity changes in OFC with dorsal striatum and related circuits induced by opioid self-administration versus voluntary abstinence predicted individual differences in incubation of opioid craving.
Collapse
Affiliation(s)
- Ida Fredriksson
- Behavioral Neuroscience Branch, Intramural Research Program/National Institute on Drug Abuse/NIH, Baltimore, MD 21224
- Center for Social and Affective Neuroscience, Linköping University, Linköping 581 83, Sweden
| | - Pei-Jung Tsai
- Neuroimaging Research Branch, Intramural Research Program/National Institute on Drug Abuse/NIH, Baltimore, MD 21224
| | - Aniruddha Shekara
- Behavioral Neuroscience Branch, Intramural Research Program/National Institute on Drug Abuse/NIH, Baltimore, MD 21224
| | - Ying Duan
- Neuroimaging Research Branch, Intramural Research Program/National Institute on Drug Abuse/NIH, Baltimore, MD 21224
| | - Sarah V Applebey
- Behavioral Neuroscience Branch, Intramural Research Program/National Institute on Drug Abuse/NIH, Baltimore, MD 21224
| | - Hanbing Lu
- Neuroimaging Research Branch, Intramural Research Program/National Institute on Drug Abuse/NIH, Baltimore, MD 21224
| | - Jennifer M Bossert
- Behavioral Neuroscience Branch, Intramural Research Program/National Institute on Drug Abuse/NIH, Baltimore, MD 21224
| | - Yavin Shaham
- Behavioral Neuroscience Branch, Intramural Research Program/National Institute on Drug Abuse/NIH, Baltimore, MD 21224;
| | - Yihong Yang
- Neuroimaging Research Branch, Intramural Research Program/National Institute on Drug Abuse/NIH, Baltimore, MD 21224
| |
Collapse
|
28
|
Butcher TJ, Chumin EJ, West JD, Dzemidzic M, Yoder KK. Cerebral Blood Flow in the Salience Network of Individuals with Alcohol Use Disorder. Alcohol Alcohol 2021; 57:445-451. [PMID: 34541599 DOI: 10.1093/alcalc/agab062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/27/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023] Open
Abstract
AIMS Magnetic resonance imaging (MRI) studies have identified structural and functional differences in salience network nodes of individuals with alcohol use disorders (AUDs) after chronic exposure to alcohol. However, no studies have investigated cerebral blood flow (CBF) in nontreatment-seeking (NTS) individuals with AUD. METHODS In this work, we sought to quantify putative CBF deficits in NTS individuals relative to social drinking (SD) controls and determine if CBF in the salience network is associated with AUD severity. Fifteen NTS (36.5 ± 11.2 years old, 30.0 ± 22.7 drinks/week) and 22 SD (35.6 ± 11.9 years old, 9.1 ± 5.7 drinks/week) underwent pseudocontinuous arterial spin labeling MRI. RESULTS Compared with social drinkers, NTS individuals had significantly lower CBF in the right and left dorsal anterior insula, and the left ventral anterior and posterior insula. The Alcohol Use Disorder Identification Test (AUDIT) score showed a significant negative relationship with CBF in the bilateral caudal anterior cingulate cortex. In addition, a significant negative correlation was present between number of standard drinks consumed per week and the left frontal opercular CBF. CONCLUSION These results provide evidence that insular CBF is negatively associated with heavy drinking, and that severity of alcohol use is related to CBF deficits in key nodes of the salience network. Longitudinal data are needed to understand if disruptions of CBF in the insula and the salience network are a predisposition for or a consequence of chronic AUD.
Collapse
Affiliation(s)
- Tarah J Butcher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Department of Radiology and Imaging Sciences, Indiana University Center for Neuroimaging, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Department of Psychiatry, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Department of Psychology, Indiana University Purdue University Indianapolis, 402 N. Blackford St, Indianapolis, IN 46202, USA
| | - Evgeny J Chumin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Department of Radiology and Imaging Sciences, Indiana University Center for Neuroimaging, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th St., Indianapolis, IN 46202, USA.,Department of Psychological and Brain Sciences, Indiana University, 1101 E 10th St, Bloomington, IN 47405, USA.,Indiana University Network Science Institute, Indiana University, 1001 IN-45, Bloomington, IN 47408, USA
| | - John D West
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Department of Radiology and Imaging Sciences, Indiana University Center for Neuroimaging, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th St., Indianapolis, IN 46202, USA
| | - Mario Dzemidzic
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Department of Radiology and Imaging Sciences, Indiana University Center for Neuroimaging, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Department of Neurology, Indiana University School of Medicine, 355. W. 16th St., Indianapolis, IN 46202, USA
| | - Karmen K Yoder
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Department of Radiology and Imaging Sciences, Indiana University Center for Neuroimaging, Indiana University School of Medicine, 355 W. 16th St., Indianapolis, IN 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th St., Indianapolis, IN 46202, USA.,Department of Psychology, Indiana University Purdue University Indianapolis, 402 N. Blackford St, Indianapolis, IN 46202, USA
| |
Collapse
|
29
|
Grodin EN, Burnette E, Towns B, Venegas A, Ray LA. Effect of alcohol, tobacco, and cannabis co-use on gray matter volume in heavy drinkers. PSYCHOLOGY OF ADDICTIVE BEHAVIORS 2021; 35:760-768. [PMID: 34435833 PMCID: PMC8484037 DOI: 10.1037/adb0000743] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Alcohol, tobacco, and cannabis are the three most frequently used drugs in the United States and co-use is common. Alcohol, tobacco, and cannabis use has been separately associated with altered brain structure, and alcohol and tobacco co-use results in decreases in gray matter volume. Less is known about the effect of alcohol and cannabis co-use, and alcohol, tobacco, and cannabis tri-use. Therefore, this study examined the effect of co- and tri-use on gray matter volume, a measure of brain cell density, in heavy drinkers. METHOD Heavy drinkers (n = 237; 152m/85f; age = 32.52; white = 111; black = 28; Latino = 9; American Indian = 2; Pacific Islander = 4; Asian = 59; mixed = 15; other = 9) were classified into four groups based on their alcohol, tobacco, and cannabis use: alcohol only users (n = 70), alcohol and tobacco co-users (n = 90), alcohol and cannabis co-users (n = 35), and alcohol, tobacco, and cannabis tri-users (n = 42). All participants completed a structural MRI scan. Voxel-based morphometry was conducted to evaluate the effect of co-use on gray matter volume, with alcohol only users as the reference group. Age, sex, and scanner were included as covariates. RESULTS Alcohol and tobacco co-users had significantly decreased left orbitofrontal gray matter volume relative to alcohol only users (Cohen's d = .79). There were no differences in gray matter volume between the alcohol only and alcohol and cannabis co-users, or between the alcohol only and tri-user groups. CONCLUSION The additive effect of tobacco co-use on gray matter volumes in heavy drinkers was limited and localized. The effect of tri-use of alcohol, tobacco, and cannabis may have interacted, such that overlapping cannabis and tobacco use masked volume differences present in separate co-using groups. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
- Erica N. Grodin
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
| | - Elizabeth Burnette
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
- Neuroscience Interdepartmental Program, University of California at Los Angeles, Los Angeles, CA
| | - Brandon Towns
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
| | - Alexandra Venegas
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
| | - Lara A. Ray
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA
| |
Collapse
|
30
|
Dong H, Wang M, Zhang J, Hu Y, Potenza MN, Dong G. Reduced frontostriatal functional connectivity and associations with severity of Internet gaming disorder. Addict Biol 2021; 26:e12985. [PMID: 33236526 DOI: 10.1111/adb.12985] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
Abstract
Cognitive, functional, and structural brain factors involving frontal executive and striatal reward networks have been implicated in Internet gaming disorder (IGD). However, frontostriatal network connectivity and its association with addiction severity are poorly understood in IGD. Resting-state fMRI data from 337 subjects (130 with IGD, 207 with recreational game use [RGU]) were collected. Striatal-cortical communications were measured with resting-state functional connectivity (FC) using coherent spontaneous fluctuations in the blood-oxygenation-level-dependent fMRI signal. Correlations were calculated between FC measures and IGD-related assessments (addiction severity and craving scores). Decreased FC was predominantly observed in IGD subjects, with IGD subjects showing decreased FC between the putamen and superior frontal gyrus (SFG), middle frontal gyrus (MFG), and inferior frontal gyrus (IFG) and the ventral striatum and IFG, superior temporal gyrus, and MFG. Disorder severity and craving scores were negatively correlated with FC between striatal and frontal brain regions. Associations between diminished FC in corticostriatal circuitry and clinical features (IGD craving, severity) suggest potential therapeutic targets for neuromodulation treatments. The extent to which frontostriatal circuits involving executive control over reward processes may be altered to treat IGD warrants additional study.
Collapse
Affiliation(s)
- Haohao Dong
- Center for Cognition and Brain Disorders The Affiliated Hospital of Hangzhou Normal University Hangzhou Zhejiang Province China
- Institute of Psychological Science Hangzhou Normal University Hangzhou Zhejiang Province China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments Hangzhou Zhejiang Province China
- Department of Psychology Zhejiang Normal University Jinhua China
| | - Min Wang
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments Hangzhou Zhejiang Province China
| | - Jialin Zhang
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments Hangzhou Zhejiang Province China
| | - Yuzheng Hu
- Department of Psychology and Behavioral Sciences Zhejiang University Hangzhou Zhejiang Province China
| | - Marc N. Potenza
- Department of Psychiatry, Department of Neuroscience, Child Study Center, and Connecticut Mental Health Center Yale University School of Medicine New Haven Connecticut USA
| | - Guang‐Heng Dong
- Center for Cognition and Brain Disorders The Affiliated Hospital of Hangzhou Normal University Hangzhou Zhejiang Province China
- Institute of Psychological Science Hangzhou Normal University Hangzhou Zhejiang Province China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments Hangzhou Zhejiang Province China
| |
Collapse
|
31
|
Weafer J, Gorka SM, Dzemidzic M, Kareken DA, Phan KL, de Wit H. Neural correlates of inhibitory control are associated with stimulant-like effects of alcohol. Neuropsychopharmacology 2021; 46:1442-1450. [PMID: 33947965 PMCID: PMC8208996 DOI: 10.1038/s41386-021-01014-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/03/2021] [Accepted: 04/06/2021] [Indexed: 11/08/2022]
Abstract
Poor inhibitory control and heightened feelings of stimulation after alcohol are two well-established risk factors for alcohol use disorder (AUD). Although these risk factors have traditionally been viewed as orthogonal, recent evidence suggests that the two are related and may share common neurobiological mechanisms. Here we examined the degree to which neural activity during inhibition was associated with subjective reports of stimulation following alcohol. To assess neural changes during inhibition, moderate alcohol drinkers performed a stop signal task during fMRI without drug. To assess subjective responses to alcohol they ingested alcohol (0.8 g/kg) or placebo beverages under double-blind conditions and provided subjective reports of stimulation and sedation. Feelings of stimulation following alcohol were inversely associated with activity in the supplementary motor area, insula, and middle frontal gyrus during inhibition (successful stop trials compared to go trials). Feelings of sedation did not correlate with brain activation. These results extend previous findings suggesting that poor inhibitory control is associated with more positive subjective responses to alcohol. These interrelated risk factors may contribute to susceptibility to future excessive alcohol use, and ultimately lead to neurobiological targets to prevent or treat AUD.
Collapse
Affiliation(s)
- Jessica Weafer
- Department of Psychology, University of Kentucky, Lexington, KY, USA.
| | - Stephanie M Gorka
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA
| | - Mario Dzemidzic
- Department of Neurology, Indiana University, Bloomington, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University, Bloomington, IN, USA
| | - David A Kareken
- Department of Neurology, Indiana University, Bloomington, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University, Bloomington, IN, USA
- Department of Psychiatry, Indiana University, Bloomington, IN, USA
- Stark Neurosciences Research Institute, Indianapolis, IN, USA
| | - K Luan Phan
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA
| | - Harriet de Wit
- Department of Psychiatry, University of Chicago, Chicago, IL, USA
| |
Collapse
|
32
|
Liu W, Peeters N, Fernández G, Kohn N. Common neural and transcriptional correlates of inhibitory control underlie emotion regulation and memory control. Soc Cogn Affect Neurosci 2021; 15:523-536. [PMID: 32507888 PMCID: PMC7328031 DOI: 10.1093/scan/nsaa073] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/05/2020] [Accepted: 05/22/2020] [Indexed: 12/30/2022] Open
Abstract
Inhibitory control is crucial for regulating emotions and may also enable memory control. However, evidence for their shared neurobiological correlates is limited. Here, we report meta-analyses of neuroimaging studies on emotion regulation, or memory control and link neural commonalities to transcriptional commonalities using the Allen Human Brain Atlas (AHBA). Based on 95 functional magnetic resonance imaging studies, we reveal a role of the right inferior parietal lobule embedded in a frontal–parietal–insular network during emotion regulation and memory control, which is similarly recruited during response inhibition. These co-activation patterns also overlap with the networks associated with ‘inhibition’, ‘cognitive control’ and ‘working memory’ when consulting the Neurosynth. Using the AHBA, we demonstrate that emotion regulation- and memory control-related brain activity patterns are associated with transcriptional profiles of a specific set of ‘inhibition-related’ genes. Gene ontology enrichment analysis of these ‘inhibition-related’ genes reveal associations with the neuronal transmission and risk for major psychiatric disorders as well as seizures and alcoholic dependence. In summary, this study identified a neural network and a set of genes associated with inhibitory control across emotion regulation and memory control. These findings facilitate our understanding of the neurobiological correlates of inhibitory control and may contribute to the development of brain stimulation and pharmacological interventions.
Collapse
Affiliation(s)
- Wei Liu
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6525 EN Nijmegen, The Netherlands
| | - Nancy Peeters
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6525 EN Nijmegen, The Netherlands
| | - Guillén Fernández
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6525 EN Nijmegen, The Netherlands
| | - Nils Kohn
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6525 EN Nijmegen, The Netherlands
| |
Collapse
|
33
|
Zhuang Q, Xu L, Zhou F, Yao S, Zheng X, Zhou X, Li J, Xu X, Fu M, Li K, Vatansever D, Kendrick KM, Becker B. Segregating domain-general from emotional context-specific inhibitory control systems - ventral striatum and orbitofrontal cortex serve as emotion-cognition integration hubs. Neuroimage 2021; 238:118269. [PMID: 34139360 DOI: 10.1016/j.neuroimage.2021.118269] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/10/2021] [Accepted: 06/13/2021] [Indexed: 10/21/2022] Open
Abstract
Inhibitory control hierarchically regulates cognitive and emotional systems in the service of adaptive goal-directed behavior across changing task demands and environments. While previous studies convergently determined the contribution of prefrontal-striatal systems to general inhibitory control, findings on the specific circuits that mediate emotional context-specific impact on inhibitory control remained inconclusive. Against this background we combined an evaluated emotional Go/No Go task with fMRI in a large cohort of subjects (N=250) to segregate brain systems and circuits that mediate domain-general from emotion-specific inhibitory control. Particularly during a positive emotional context, behavioral results showed a lower accuracy for No Go trials and a faster response time for Go trials. While the dorsal striatum and lateral frontal regions were involved in inhibitory control irrespective of emotional context, activity in the ventral striatum (VS) and medial orbitofrontal cortex (mOFC) varied as a function of emotional context. On the voxel-wise whole-brain network level, limbic and striatal systems generally exhibited highest changes in global brain connectivity during inhibitory control, while global brain connectivity of the left mOFC was less decreased during emotional contexts. Functional connectivity analyses moreover revealed that negative coupling between the VS with inferior frontal gyrus (IFG)/insula and mOFC varied as a function of emotional context. Together these findings indicate separable domain- general as well as emotional context-specific inhibitory brain systems which specifically encompass the VS and its connections with frontal regions.
Collapse
Affiliation(s)
- Qian Zhuang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Xu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Feng Zhou
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shuxia Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiao Zheng
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinqi Zhou
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jialin Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaolei Xu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Meina Fu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Keshuang Li
- School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
| | - Deniz Vatansever
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Keith M Kendrick
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China; Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China.
| | - Benjamin Becker
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
34
|
Hildebrandt MK, Dieterich R, Endrass T. Neural correlates of inhibitory control in relation to the degree of substance use and substance-related problems - A systematic review and perspective. Neurosci Biobehav Rev 2021; 128:1-11. [PMID: 34097979 DOI: 10.1016/j.neubiorev.2021.06.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/11/2021] [Accepted: 06/02/2021] [Indexed: 12/28/2022]
Abstract
Inhibitory control deficits are associated with substance use disorders (SUDs) and considered a risk factor. Most studies compare SUD groups with unaffected individuals, although the degree of substance use might relate to inhibitory control deficits and explain group differences. This raises the question to which extent these deficits are specifically linked to substance-related problems. We review studies reporting associations of inhibition-related neural activation (stop signal and go/nogo task) with continuous measures of the degree of substance use and substance-related problems, and with substance-related problems controlling for the degree of substance use. Results suggest negative associations between inhibition-related neural activation and the degree of substance use, but are inconclusive on the association with substance-related problems. Nonetheless, two studies reported significant associations of inhibition-related neural activation with substance-related problems controlling for the degree of substance use. Despite numerous studies showing alterations in inhibition-related neural activation in SUDs, the role of the degree of substance use needs further investigation and studies using dimensional approaches are necessary to uncover specific links to substance-related problems.
Collapse
Affiliation(s)
- Malin K Hildebrandt
- Technische Universität Dresden, Faculty of Psychology, Institute for Clinical Psychology and Psychotherapy, Addiction Research, Germany.
| | - Raoul Dieterich
- Technische Universität Dresden, Faculty of Psychology, Institute for Clinical Psychology and Psychotherapy, Addiction Research, Germany.
| | - Tanja Endrass
- Technische Universität Dresden, Faculty of Psychology, Institute for Clinical Psychology and Psychotherapy, Addiction Research, Germany.
| |
Collapse
|
35
|
McCane AM, Auterson CD, DeLory MJ, Lapish CC, Czachowski CL. Differential effects of quinine adulteration of alcohol on seeking and drinking. Alcohol 2021; 92:73-80. [PMID: 33465465 PMCID: PMC8026625 DOI: 10.1016/j.alcohol.2021.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 12/26/2022]
Abstract
Alcohol dependence is characterized by compulsive alcohol use. Alcohol-paired stimuli can drive compulsive alcohol use, induce craving, and lead to relapse. Alcohol dependence is highly heritable, and individuals with a family history are at elevated risk to develop an alcohol use disorder. Understanding the association between genetic vulnerability to alcohol dependence and neural alterations that promote an addiction phenotype are critical to the prevention and treatment of alcohol dependence. Here we use selectively bred alcohol-preferring P rats and their progenitor strain, Wistar rats, to investigate the relationship between genetic liability and alcohol-seeking and drinking behaviors in a discriminative stimuli paradigm. To further investigate strain differences in motivated responding, alcohol was adulterated with quinine, and intake and responding were assessed. While both strains learned to discriminate between stimuli that predicted alcohol availability, P rats learned faster and consumed more alcohol. Quinine adulteration reduced ethanol intake in both strains with no effect on ethanol-seeking measures. These data suggest genetic vulnerability to alcohol dependence is associated with increased motivated behaviors and highlight the utility of P rats in teasing apart the neural mechanisms associated with this phenotype. Additionally, these data suggest a dissociation between the neural systems that engage ethanol drinking versus compulsive ethanol seeking.
Collapse
Affiliation(s)
- Aqilah M McCane
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, United States.
| | - Curtis D Auterson
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Michael J DeLory
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Christopher C Lapish
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, United States; Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Cristine L Czachowski
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, United States; Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| |
Collapse
|
36
|
Chicchi Giglioli IA, Pérez Gálvez B, Gil Granados A, Alcañiz Raya M. The Virtual Cooking Task: A Preliminary Comparison Between Neuropsychological and Ecological Virtual Reality Tests to Assess Executive Functions Alterations in Patients Affected by Alcohol Use Disorder. CYBERPSYCHOLOGY, BEHAVIOR AND SOCIAL NETWORKING 2021; 24:673-682. [PMID: 33761276 DOI: 10.1089/cyber.2020.0560] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Alcohol use disorder (AUD) is a major global problem. Neuropsychological studies have shown that AUD causes deficits in executive functions (EFs), a set of higher order cognitive skills that govern individual behavior in everyday situations. Many standardized neuropsychological tests are used to evaluate EF. These are reliable and valid but have limitations in predicting real-life performance. To address this, we present a preliminary study to test the virtual cooking task (VCT) as an alternative to standardized neuropsychological tests. The VCT includes four subtasks developed to assess attentional, planning, and cognitive shifting abilities; it was tested in an immersive three-dimensional environment. To evaluate the VCT performance and standardized neuropsychological tests, data were gathered from a sample of healthy subjects (control group [CG]; n = 23) and AUD patients (n = 18). The standardized neuropsychological measures used consisted of questionnaires (Attentional Control Scale, Barratt Impulsiveness Scale, and Cognitive Flexibility Scale) and specific tests (Dot-probe task, Go/No-go test, Stroop test, the trail making test, and Tower of London test). The results showed significant higher correlations for AUD patients than for the CG for the VCT, questionnaires, and specific tests, mainly related to planning and cognitive shifting abilities. Furthermore, comparative analyses of the VCT performance showed that the AUD patients made more errors and had higher latency times than the CG. The present study provides initial evidence that a more ecologically valid assessment can be a useful tool to detect cognitive impairments in many neuropsychological and mental disorders, affecting daily activities.
Collapse
Affiliation(s)
- Irene Alice Chicchi Giglioli
- Department of Graphics Engineering, Instituto de Investigación e Innovación en Bioingeniería (I3B), Universitat Politècnica de València, València, Spain
| | - Bartolomé Pérez Gálvez
- Health Department, Unidad de Alcohología, Hospital Universitario de San Juan, Alicante, Spain.,Departamento de Medicina Clínica, Universidad Miguel Hernández, Elche, Spain
| | - Andrea Gil Granados
- Department of Graphics Engineering, Instituto de Investigación e Innovación en Bioingeniería (I3B), Universitat Politècnica de València, València, Spain
| | - Mariano Alcañiz Raya
- Department of Graphics Engineering, Instituto de Investigación e Innovación en Bioingeniería (I3B), Universitat Politècnica de València, València, Spain
| |
Collapse
|
37
|
Campbell EJ, Lawrence AJ. It's more than just interoception: The insular cortex involvement in alcohol use disorder. J Neurochem 2021; 157:1644-1651. [PMID: 33486788 DOI: 10.1111/jnc.15310] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/24/2022]
Abstract
Understanding brain structures and circuits impacted by alcohol use disorder is critical for improving our future prevention techniques and treatment options. A brain region that has recently gained traction for its involvement in substance use disorder is the insular cortex. This brain region is multi-functional and spatially complex, resulting in a relative lack of understanding of the involvement of the insular cortex in alcohol use disorder. Here we discuss the role of the insular cortex in alcohol use disorder, particularly during periods of abstinence and in response to alcohol and alcohol-related cues and contexts. We also discuss a broader role of the insular in alcohol-associated risky decision making and impulse control. Finally, we canvas potential challenges associated with targeting the insular cortex to treat individuals with alcohol use disorder.
Collapse
Affiliation(s)
- Erin J Campbell
- The Florey Institute of Neuroscience and Mental Health, Parkville, Vic, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Vic, Australia
| | - Andrew J Lawrence
- The Florey Institute of Neuroscience and Mental Health, Parkville, Vic, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Vic, Australia
| |
Collapse
|
38
|
Britton MK, Porges EC, Bryant V, Cohen RA. Neuroimaging and Cognitive Evidence for Combined HIV-Alcohol Effects on the Central Nervous System: A Review. Alcohol Clin Exp Res 2021; 45:290-306. [PMID: 33296091 PMCID: PMC9486759 DOI: 10.1111/acer.14530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/29/2020] [Indexed: 12/27/2022]
Abstract
Alcohol use disorder (AUD) among people living with HIV (PLWH) is a significant public health concern. Despite the advent of effective antiretroviral therapy, up to 50% of PLWH still experience worsened neurocognition, which comorbid AUD exacerbates. We report converging lines of neuroimaging and neuropsychological evidence linking comorbid HIV/AUD to dysfunction in brain regions linked to executive function, learning and memory, processing speed, and motor control, and consequently to impairment in daily life. The brain shrinkage, functional network alterations, and brain metabolite disruption seen in individuals with HIV/AUD have been attributed to several interacting pathways: viral proteins and EtOH are directly neurotoxic and exacerbate each other's neurotoxic effects; EtOH reduces antiretroviral adherence and increases viral replication; AUD and HIV both increase gut microbial translocation, promoting systemic inflammation and HIV transport into the brain by immune cells; and HIV may compound alcohol's damaging effects on the liver, further increasing inflammation. We additionally review the neurocognitive effects of aging, Hepatitis C coinfection, obesity, and cardiovascular disease, tobacco use, and nutritional deficiencies, all of which have been shown to compound cognitive changes in HIV, AUD, and in their comorbidity. Finally, we examine emerging questions in HIV/AUD research, including genetic and cognitive protective factors, the role of binge drinking in HIV/AUD-linked cognitive decline, and whether neurocognitive and brain functions normalize after drinking cessation.
Collapse
Affiliation(s)
- Mark K. Britton
- University of Florida, Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, Cognitive Aging and Memory Clinical Translational Research Program; 1225 Center Drive, Gainesville, Florida 32607
| | - Eric C. Porges
- University of Florida, Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, Cognitive Aging and Memory Clinical Translational Research Program; 1225 Center Drive, Gainesville, Florida 32607
| | - Vaughn Bryant
- University of Florida, Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, Cognitive Aging and Memory Clinical Translational Research Program; 1225 Center Drive, Gainesville, Florida 32607
- University of Florida, Department of Epidemiology, 2004 Mowry Road, Gainesville, FL 32610
| | - Ronald A. Cohen
- University of Florida, Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, Cognitive Aging and Memory Clinical Translational Research Program; 1225 Center Drive, Gainesville, Florida 32607
| |
Collapse
|
39
|
Strosche A, Zhang X, Kirsch M, Hermann D, Ende G, Kiefer F, Vollstädt‐Klein S. Investigation of brain functional connectivity to assess cognitive control over cue-processing in Alcohol Use Disorder. Addict Biol 2021; 26:e12863. [PMID: 31908107 DOI: 10.1111/adb.12863] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 10/19/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022]
Abstract
Alcohol Use Disorder has been associated with impairments of functional connectivity between neural networks underlying reward processing and cognitive control. Evidence for aberrant functional connectivity between the striatum, insula, and frontal cortex in alcohol users exists at rest, but not during cue-exposure. In this study, we investigated functional connectivity changes during a cue-reactivity task across different subgroups of alcohol consumers. Ninety-six participants (ranging from light social to heavy social drinkers and nonabstinent dependent to abstinent dependent drinkers) were examined. A functional magnetic resonance imaging cue-reactivity paradigm was administered, during which alcohol-related and neutral stimuli were presented. Applying psychophysiological interaction analyses, we found: (a) Abstinent alcohol-dependent patients compared with non-abstinent dependent drinkers showed a greater increase of functional connectivity of the ventral striatum and anterior insula with the anterior cingulate cortex and dorsolateral prefrontal cortex during the presentation of alcohol cues compared with neutral cues. (b) Subjective craving correlated positively with functional connectivity change between the posterior insula and the medial orbitofrontal cortex and negatively with functional connectivity change between the ventral striatum and the anterior cingulate cortex, dorsolateral prefrontal cortex, and lateral orbitofrontal cortex. (c) Compulsivity of alcohol use correlated positively with functional connectivity change between the dorsolateral prefrontal cortex and the ventral striatum, anterior insula, and posterior insula. Results suggest increased cognitive control over cue-processing in abstinent alcohol-dependent patients, compensating high levels of cue-provoked craving and compulsive use. Clinical trial registration details: ClinicalTrials.gov ID: NCT00926900.
Collapse
Affiliation(s)
- Alicia Strosche
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim University of Heidelberg Heidelberg Germany
| | - Xiaochu Zhang
- CAS Key Laboratory of Brain Function and Disease and School of Life Sciences University of Science and Technology of China Hefei Anhui China
- Hefei Medical Research Center on Alcohol Addiction Anhui Mental Health Center Hefei China
- Academy of Psychology and Behavior Tianjin Normal University Tianjin China
| | - Martina Kirsch
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim University of Heidelberg Heidelberg Germany
| | - Derik Hermann
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim University of Heidelberg Heidelberg Germany
| | - Gabriele Ende
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim University of Heidelberg Heidelberg Germany
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim University of Heidelberg Heidelberg Germany
| | - Sabine Vollstädt‐Klein
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim University of Heidelberg Heidelberg Germany
| |
Collapse
|
40
|
León-Domínguez U, Solís-Marcos I, López-Delgado CA, Martín JMBY, León-Carrión J. A Frontal Neuropsychological Profile in Fitness to Drive. ACCIDENT; ANALYSIS AND PREVENTION 2020; 148:105807. [PMID: 33069156 DOI: 10.1016/j.aap.2020.105807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/24/2020] [Accepted: 09/27/2020] [Indexed: 06/11/2023]
Abstract
Traffic accidents are a global concern due to the elevated mortality rates of both drivers and pedestrians. The World Health Organization declared 2011-2020 as the Decade of Action for Road Safety, endorsing initiatives to reduce traffic-related deaths. Yet, despite these incentives, fatal accidents still occur. Different studies have linked deficits in executive functions to risky driving attitudes and crashes. The present study focuses on demographic, cognitive and personality factors, related to the prefrontal cortex, that are characteristic of drivers prone to risky behavior behind the wheel. The penalty Points System was used to classify drivers as "safe", with no point loss over a two-year period, or "risky", with full point loss during the same interval. A neuropsychological assessment of prefrontal cognitive functions was carried out on each group to identify variables associated with safe and risky behavior. Neuropsychological indexes were obtained from a continuous performance task without cue (Simple Attention), a continuous performance task with cue (Conditioned Attention), the Tower of Hanoi test and the Neurologically-related Changes in Personality Inventory (NECHAPI). A Discriminant Analysis (DA) found that education level, reaction times in Simple and Conditioned Attention, learning errors in the Tower of Hanoi and vulnerability in the personality test, best predicted whether drivers were likely to be in the safe or risky group. Finally, a cross-validation analysis performed on the same sample correctly classified 87.5% of the drivers. These data suggest that prefrontal dysfunction contributes to risky behavior behind the wheel. The inclusion of cognitive programs to identify and train drivers with this propensity could reduce risky driving, and consequently, save lives on the road.
Collapse
Affiliation(s)
- Umberto León-Domínguez
- Human Cognition and Brain Research lab, School of Psychology, University of Monterrey, San Pedro Garza, García, Mexico.
| | - Ignacio Solís-Marcos
- The Swedish National Road and Transport Research Institute (VTI) Linköping, Sweden
| | | | | | - José León-Carrión
- Department of Experimental Psychology, University of Seville, Seville, Spain; Center for Brain Injury Rehabilitation (CRECER), Seville, Spain
| |
Collapse
|
41
|
Luciana M. Risks versus consequences of adolescent and young adult substance use: A focus on executive control. CURRENT ADDICTION REPORTS 2020; 7:453-463. [PMID: 33816055 PMCID: PMC8014909 DOI: 10.1007/s40429-020-00341-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW This review examines the role of executive control processes in the liability for substance misuse and whether substance use, once initiated, leads to subsequent decrements as proposed by neurotoxicity models of substance use disorder (SUD). RECENT FINDINGS As indicated by a number of recent meta-analyses, executive control processes, which include working memory, cognitive flexibility and numerous aspects of attentional, behavioral and emotional control, are impaired in the context of active SUD. Longitudinal studies of behaviorally disinhibited children, individuals with familial risks for SUD, and twins within whom genetic versus environmental influences on behavior can be modeled robustly indicate that relatively poor control is a vulnerability factor for early substance use initiation, binge patterns of use, and subsequent SUD. Evidence of further declines in executive control, once substance use is initiated, is mixed, although a growing number of neuroimaging studies indicate that frontostriatal, frontolimbic, and frontocerebellar systems are altered as a consequence of use. SUMMARY Together these patterns suggest strategies for identifying children and adolescents at high risk for SUD, avenues through which substance-related neurotoxicities can be more reliably detected, and the need to structure prevention efforts in a manner that is developmentally appropriate and perhaps personalized to individual vulnerabilities.
Collapse
Affiliation(s)
- Monica Luciana
- Department of Psychology, University of Minnesota, Minneapolis MN, 55455 USA
| |
Collapse
|
42
|
Zhao Y, Sallie SN, Cui H, Zeng N, Du J, Yuan T, Li D, De Ridder D, Zhang C. Anterior Cingulate Cortex in Addiction: New Insights for Neuromodulation. Neuromodulation 2020; 24:S1094-7159(21)00082-9. [PMID: 33090660 DOI: 10.1111/ner.13291] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Substance use disorder (SUD) is characterized by compulsive use of addictive substances with considerable impact on both the medical system and society as a whole. The craving of substances leads to relapse in the majority of patients within one year of traditional treatments. In recent decades, neuromodulation approaches have emerged as potential novel treatments of SUD, but the ideal neural target remains contentious. MATERIALS AND METHODS In this review, we discuss new insights on the anterior cingulate cortex (ACC) as a neuromodulation target for SUD. RESULTS AND CONCLUSION First, we illustrate that the ACC serves as a central "hub" in addiction-related neural networks of cognitive functions, including, but not limited to, decision-making, cognitive inhibition, emotion, and motivation. Then, we summarize the literature targeting the ACC to treat SUDs via available neuromodulation approaches. Finally, we propose potential directions to improve the effect of stimulating the ACC in SUD treatment. We emphasize that the ACC can be divided into at least four sub-regions, which have distinctive functions and connections. Studies focusing on these sub-regions may help to develop more precise and effective ACC stimulation according to patients' symptom profiles and cognitive deficits.
Collapse
Affiliation(s)
- Yijie Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Samantha N Sallie
- Department of Psychiatry, University of Cambridge, Level E4, Addenbrooke's Hospital, Cambridge, UK
| | - Hailun Cui
- Department of Psychiatry, University of Cambridge, Level E4, Addenbrooke's Hospital, Cambridge, UK
| | - Ningning Zeng
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiang Du
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dianyou Li
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dirk De Ridder
- Department of Surgical Sciences, Section of Neurosurgery, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
43
|
Gerchen MF, Weiss F, Kirsch M, Rentsch A, Halli P, Kiefer F, Kirsch P. Dynamic frontostriatal functional peak connectivity (in alcohol use disorder). Hum Brain Mapp 2020; 42:36-46. [PMID: 32885886 PMCID: PMC7721230 DOI: 10.1002/hbm.25201] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/31/2020] [Accepted: 08/23/2020] [Indexed: 01/25/2023] Open
Abstract
Alcohol use disorder (AUD) is associated with changes in frontostriatal connectivity, but functional magnetic resonance imaging (fMRI) functional connectivity (FC) approaches are usually not adapted to these circuits. We developed a circuit‐specific fMRI analysis approach to detect dynamic changes in frontostriatal FC inspired by medial‐ventral‐rostral to lateral‐dorsal‐caudal frontostriatal gradients originally identified in nonhuman primate tract‐tracing data. In our PeaCoG (“peak connectivity on a gradient”) approach we use information about the location of strongest FC on empirical frontostriatal connectivity gradients. We have recently described a basic PeaCoG version with conventional FC, and now developed a dynamic PeaCoG approach with sliding‐window FC. In resting state data of n = 66 AUD participants and n = 40 healthy controls we continue here the analyses that we began with the basic version. Our former result of an AUD‐associated ventral shift in right orbitofrontal cortex PeaCoG is consistently detected in the dynamic approach. Temporospatial variability of dynamic PeaCoG in the left dorsolateral prefrontal cortex is reduced in AUD and associated with self‐efficacy to abstain and days of abstinence. Our method has the potential to provide insight into the dynamics of frontostriatal circuits, which has so far been relatively unexplored, and into their role in mental disorders and normal cognition.
Collapse
Affiliation(s)
- Martin Fungisai Gerchen
- Department of Clinical Psychology, Central Institute of Mental Health, Heidelberg University/Medical Faculty Mannheim, Mannheim, Germany.,Department of Psychology, Heidelberg University, Heidelberg, Germany.,Bernstein Center for Computational Neuroscience Heidelberg/Mannheim, Mannheim, Germany
| | - Franziska Weiss
- Department of Clinical Psychology, Central Institute of Mental Health, Heidelberg University/Medical Faculty Mannheim, Mannheim, Germany
| | - Martina Kirsch
- Department of Addiction Behavior and Addiction Medicine, Central Institute of Mental Health, Heidelberg University/Medical Faculty Mannheim, Mannheim, Germany
| | - Alena Rentsch
- Department of Clinical Psychology, Central Institute of Mental Health, Heidelberg University/Medical Faculty Mannheim, Mannheim, Germany
| | - Patrick Halli
- Department of Clinical Psychology, Central Institute of Mental Health, Heidelberg University/Medical Faculty Mannheim, Mannheim, Germany
| | - Falk Kiefer
- Department of Addiction Behavior and Addiction Medicine, Central Institute of Mental Health, Heidelberg University/Medical Faculty Mannheim, Mannheim, Germany
| | - Peter Kirsch
- Department of Clinical Psychology, Central Institute of Mental Health, Heidelberg University/Medical Faculty Mannheim, Mannheim, Germany.,Department of Psychology, Heidelberg University, Heidelberg, Germany.,Bernstein Center for Computational Neuroscience Heidelberg/Mannheim, Mannheim, Germany
| |
Collapse
|
44
|
Sion A, Bruña Fernández R, Martínez Maldonado A, Domínguez Centeno I, Torrado‐Carvajal A, Rubio G, Pereda E, Jurado‐Barba R. Resting‐state connectivity and network parameter analysis in alcohol‐dependent males. A simultaneous EEG‐MEG study. J Neurosci Res 2020; 98:1857-1876. [DOI: 10.1002/jnr.24673] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 05/15/2020] [Accepted: 05/25/2020] [Indexed: 02/04/2023]
Affiliation(s)
- Ana Sion
- 12 de Octubre Biomedical Research Institute Madrid Spain
| | - Ricardo Bruña Fernández
- Laboratory of Cognitive and Computational Neuroscience Center for Biomedical Technology (CTB) Madrid Spain
- Department of Experimental Psychology Universidad Complutense de Madrid Madrid Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Madrid Spain
| | | | - Isabel Domínguez Centeno
- 12 de Octubre Biomedical Research Institute Madrid Spain
- Psychology Department, Health Science Faculty Camilo José Cela University Madrid Spain
| | - Angel Torrado‐Carvajal
- Athinoula A. Martinos Center for Biomedical Imaging Department of Radiology Massachusetts General Hospital and Harvard Medical School Boston MA USA
- Medical Image Analysis and Biometry Laboratory Universidad Rey Juan Carlos Madrid Spain
| | - Gabriel Rubio
- 12 de Octubre Biomedical Research Institute Madrid Spain
- 12 de Octubre Hospital Madrid Spain
- Medicine Faculty Complutense de Madrid University Madrid Spain
- Addictive Disorders Network (Red de Trastornos adictivos, RETIS) Carlos III Institute Madrid Spain
| | - Ernesto Pereda
- Laboratory of Cognitive and Computational Neuroscience Center for Biomedical Technology (CTB) Madrid Spain
- Department of Industrial Engineering & IUNE Universidad de la Laguna San Cristóbal de La Laguna Spain
| | - Rosa Jurado‐Barba
- 12 de Octubre Biomedical Research Institute Madrid Spain
- Psychology Department, Health Science Faculty Camilo José Cela University Madrid Spain
| |
Collapse
|
45
|
Courtney AL, Casey BJ, Rapuano KM. A Neurobiological Model of Alcohol Marketing Effects on Underage Drinking. J Stud Alcohol Drugs Suppl 2020:68-80. [PMID: 32079563 PMCID: PMC7064001 DOI: 10.15288/jsads.2020.s19.68] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 01/29/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Although an association between exposure to alcohol advertising and underage drinking is well documented, the underlying neurobiological contributions to this association remain largely unexplored. From an epidemiological perspective, identifying the neurobiological plausibility of this exposure-outcome association is a crucial step toward establishing marketing as a contributor to youth drinking and informing public policy interventions to decrease this influence. METHOD We conducted a critical review of the literature on neurobiological risk factors and adolescent brain development, social influences on drinking, and neural contributions to reward sensitization and risk taking. By drawing from these separate areas of research, we propose a unified, neurobiological model of alcohol marketing effects on underage drinking. RESULTS We discuss and extend the literature to suggest that responses in prefrontal-reward circuitry help establish alcohol advertisements as reward-predictive cues that may reinforce consumption upon exposure. We focus on adolescence as a sensitive window of development during which youth are particularly susceptible to social and reward cues, which are defining characteristics of many alcohol advertisements. As a result, alcohol marketing may promote positive associations early in life that motivate social drinking, and corresponding neurobiological changes may contribute to later patterns of alcohol abuse. CONCLUSIONS The neurobiological model proposed here, which considers neurodevelopmental risk factors, social influences, and reward sensitization to alcohol cues, suggests that exposure to alcohol marketing could plausibly influence underage drinking by sensitizing prefrontal-reward circuitry.
Collapse
Affiliation(s)
| | - B. J. Casey
- Department of Psychology, Yale University, New Haven, Connecticut
| | | |
Collapse
|
46
|
Hagerty SL, YorkWilliams SL, Bidwell LC, Weiland BJ, Sabbineni A, Blaine SK, Bryan AD, Hutchison KE. DRD2 methylation is associated with executive control network connectivity and severity of alcohol problems among a sample of polysubstance users. Addict Biol 2020; 25:e12684. [PMID: 30370960 PMCID: PMC7326368 DOI: 10.1111/adb.12684] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/07/2018] [Accepted: 09/30/2018] [Indexed: 12/21/2022]
Abstract
Chronic exposure to alcohol and other drugs of abuse has been associated with deleterious consequences, including functional connectivity deficits within neural networks associated with executive control. Altered functional connectivity within the executive control network (ECN) might underlie the progressive inability to control consumption of alcohol and other drugs as substance use disorders progress. Genetic and epigenetic factors have been associated with substance use disorders (SUDs). For example, dopamine receptor 2 (DRD2) functioning has been associated with alcohol use disorder (AUD) and related phenotypes, including correlates of executive functioning. The present study aims to explore the relationship between a continuous measure of alcohol-related problems, epigenetic markers (methylation) within the DRD2 gene, and functional connectivity within the ECN among a sample of polysubstance users. A community sample of 658 subjects, whose consumption of alcohol, nicotine, and cannabis span across a spectrum of quantity and frequency of use, were obtained across previous studies in polysubstance using populations. Resting state functional magnetic resonance imaging was analyzed to identify intrinsic connectivity networks using a priori regions of interest. Methylation measurement of functionally relevant sites within the DRD2 gene was achieved via pyrosequencing. Regression-based models, including mediation and moderation models, tested the association between DRD2 methylation, functional connectivity within intrinsic neural networks (including the ECN), and severity of alcohol problems. Results suggest that average DRD2 methylation was negatively associated with right ECN (RECN) and left ECN (LECN) connectivity, but not associated with other networks tested, and DRD2 methylation was significantly associated with alcohol problems severity. Mediation models were not supported, although moderation models suggested that connectivity between edges within the RECN moderated the relationship between DRD2 methylation and AUD severity. Results support a theoretical model in which epigenetic factors are associated with neurobiological correlates of alcohol consumption among a sample of polysubstance users.
Collapse
Affiliation(s)
- Sarah L. Hagerty
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado
| | - Sophie L. YorkWilliams
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado
| | - L. Cinnamon Bidwell
- Institute of Cognitive Science, University of Colorado Boulder, Boulder, Colorado
| | - Barbara J. Weiland
- Institute of Cognitive Science, University of Colorado Boulder, Boulder, Colorado
| | - Amithrupa Sabbineni
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado
| | - Sara K. Blaine
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Angela D. Bryan
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado
| | - Kent E. Hutchison
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado
| |
Collapse
|
47
|
Liu X, Zhou H, Jiang C, Xue Y, Zhou Z, Wang J. Cognitive Control Deficits in Alcohol Dependence Are a Trait- and State-Dependent Biomarker: An ERP Study. Front Psychiatry 2020; 11:606891. [PMID: 33363489 PMCID: PMC7752997 DOI: 10.3389/fpsyt.2020.606891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 11/16/2022] Open
Abstract
Alcohol dependence (AD) presents cognitive control deficits. Event-related potential (ERP) P300 reflects cognitive control-related processing. The aim of this study was to investigate whether cognitive control deficits are a trait biomarker or a state biomarker in AD. Participants included 30 AD patients and 30 healthy controls (HCs). All participants were measured with P300 evoked by a three-stimulus auditory oddball paradigm at a normal state (time 1, i.e., just after the last alcohol intake) and abstinence (time 2, i.e., just after a 4-week abstinence). The results showed that for P3a and P3b amplitude, the interaction effect for group × time point was significant, the simple effect for group at time 1 level and time 2 level was significant, and the simple effect for time point at AD group level was significant; however, the simple effect for time point at HC group level was not significant. Above results indicated that compared to HCs, AD patients present reductions of P3a/3b amplitude, and after 4-week alcohol abstinence, although P3a/3b amplitudes were improved, they were still lower than those of HCs. For P3a and P3b latencies, no significant differences were observed. These findings conclude that AD patients present cognitive control deficits that are reflected by P3a/3b and that cognitive control deficits in AD are trait- and state-dependent. The implication of these findings is helpful to understand the psychological and neural processes for AD, and these findings suggest that improving the cognitive control function may impact the treatment effect for AD.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of Substance Dependence, The Affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| | - Hongliang Zhou
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Chenguang Jiang
- Department of Substance Dependence, The Affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| | - Yanling Xue
- Department of Substance Dependence, The Affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| | - Zhenhe Zhou
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| | - Jun Wang
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| |
Collapse
|
48
|
Chang J, Hu J, Li CSR, Yu R. Neural correlates of enhanced response inhibition in the aftermath of stress. Neuroimage 2020; 204:116212. [PMID: 31546050 PMCID: PMC7509808 DOI: 10.1016/j.neuroimage.2019.116212] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 09/12/2019] [Accepted: 09/18/2019] [Indexed: 12/25/2022] Open
Abstract
Life stress has been shown to impact cognitive functions, including inhibitory control. However, the immediate effects of acute stress on inhibitory control and the underlying neural mechanisms remain unclear. In a behavioral pilot study (N = 30) and a within-subject functional magnetic resonance imaging study (N = 30), we examined how acute stress induced by Trier Social Stress Test influenced inhibitory control in a stop signal task. Behavioral results across two studies showed that stress consistently improved inhibitory control. Shorter stop signal reaction time (SSRT) in stress as compared with control condition was associated with stronger connectivity between the superior/middle frontal gyrus (SFG/MFG) and striatum. Dynamic causal modeling revealed distinct best models under stress and control condition, with an enhanced interaction between the SFG/MFG and the striatum after stress exposure. This research identified the SFG/MFG-striatum network as a key circuit underlying acute stress-elicited enhancement of inhibitory control in a stop signal task.
Collapse
Affiliation(s)
- Jingjing Chang
- Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, School of Psychology, South China Normal University, Guangzhou, China
| | - Jianping Hu
- Laboratory for Behavioral and Regional Finance, Guangdong University of Finance, Guangzhou, China
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Rongjun Yu
- Department of Psychology, National University of Singapore, Singapore.
| |
Collapse
|
49
|
Wang Y, Yan KJ, Fan CX, Luo XN, Zhou Y. Altered functional connectivity of the nucleus accumbens subdivisions in amphetamine-type stimulant abusers: a resting-state fMRI study. BMC Neurosci 2019; 20:66. [PMID: 31888484 PMCID: PMC6937793 DOI: 10.1186/s12868-019-0548-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/20/2019] [Indexed: 12/18/2022] Open
Abstract
Background The growing abuse of amphetamine-type stimulants leads to new challenges to human health. A possible addiction mechanism has been proposed by altered functional architecture of the nucleus accumbens (NAc) during resting state. NAc contains different subdivisions and they may play different roles in addiction. The aim of the present study was to examine whether there are common or distinct patterns of functional connectivity of the NAc subdivisions in amphetamine-type stimulant abusers (ATSAs). Methods The present study recruited 17 male ATSAs and 22 healthy male controls. All the subjects underwent resting-state functional magnetic resonance imaging (fMRI) with their eyes closed. The NAc was divided into core-like and shell-like subdivisions. We used seed-based resting-state functional connectivity (RSFC) analyses to identify differences in brain functional architecture between ATSAs and healthy controls (HCs). Results ATSAs had lower positive RSFCs with all of the NAc subdivisions over the left orbital part of superior frontal gyrus and higher positive RSFCs with the NAc subdivisions over the left opercular part of inferior frontal gyrus than HCs, which indicates common abnormalities across the NAc subdivisions in ATSAs. In addition, the RSFCs between the NAc subdivisions and the left orbital part of superior frontal gyrus were negatively correlated with the addiction severity in ATSAs. Conclusion These results provide evidence that there are common RSFC patterns of the NAc subdivisions in ATSAs. The abnormality indicated by disrupted functional connectivity between the NAc subdivisions and prefrontal cortex suggests abnormal interaction between the rewarding process and cognitive control in ATSAs. Our results shed insight on the neurobiological mechanisms of ATSA and suggest potential novel therapeutic targets for treatment and intervention of ATSAs.
Collapse
Affiliation(s)
- Yun Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Kai-Juan Yan
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Chen-Xiao Fan
- The Second Hospital of Jinhua City, Jinhua, Zhejiang, China
| | - Xiao-Nian Luo
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yuan Zhou
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing, China. .,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China. .,Key Laboratory of Behavioral Science & Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, China. .,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
50
|
de Laat B, Goldberg A, Shi J, Tetrault JM, Nabulsi N, Zheng MQ, Najafzadeh S, Gao H, Kapinos M, Ropchan J, O'Malley SS, Huang Y, Morris ED, Krishnan-Sarin S. The Kappa Opioid Receptor Is Associated With Naltrexone-Induced Reduction of Drinking and Craving. Biol Psychiatry 2019; 86:864-871. [PMID: 31399255 DOI: 10.1016/j.biopsych.2019.05.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/23/2019] [Accepted: 05/28/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Naltrexone is a nonselective opioid receptor antagonist used as a treatment for alcohol use disorder. However, only modest clinical effects have been observed, possibly because of limited knowledge about the biological variables affecting the efficacy of naltrexone. We investigated the potential role of the kappa opioid receptor (KOR) in the therapeutic effect of naltrexone. METHODS A total of 48 non-treatment-seeking heavy drinkers (16 women) who met DSM-IV criteria for alcohol dependence participated in two alcohol drinking paradigms (ADPs) separated by a week of open-label naltrexone (100 mg daily). Craving, assessed with the Alcohol Urge Questionnaire and the Yale Craving Scale, and drinking behavior were recorded in each ADP. Prior to naltrexone initiation, KOR availability was determined in the amygdala, hippocampus, pallidum, striatum, cingulate cortex, and prefrontal cortex using positron emission tomography with [11C]LY2795050. RESULTS Participants reported lower levels of craving (Yale Craving Scale: -11 ± 1, p < .0001; Alcohol Urge Questionnaire: -6 ± 0.6, p < .0001) and consumed fewer drinks (-3.7 ± 4, p < .0001) during the second ADP following naltrexone therapy. The observed reduction in drinking was negatively associated with baseline KOR availability in the striatum (p = .005), pallidum (p = .023), and cingulate cortex (p = .018). Voxelwise analysis identified clusters in the bilateral insula, prefrontal, and cingulate cortex associated with the reduction in drinking (p < .0001). In addition, KOR availability in all evaluated brain regions was associated with craving measured in both ADPs. CONCLUSIONS The KOR is implicated in drinking and craving following naltrexone therapy in alcohol use disorder.
Collapse
Affiliation(s)
- Bart de Laat
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut.
| | - Alissa Goldberg
- Department of Psychiatry, Yale University, New Haven, Connecticut
| | - Julia Shi
- Department of Internal Medicine, Yale University, New Haven, Connecticut
| | | | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Ming-Qiang Zheng
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Soheila Najafzadeh
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Hong Gao
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Michael Kapinos
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Jim Ropchan
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | | | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Evan D Morris
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut; Department of Psychiatry, Yale University, New Haven, Connecticut; Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | | |
Collapse
|