1
|
Fu J, Xu X, Dong YS, Wang M, Zhou Z, Hu Y, Li Q, Liu S, He W, Dong GH. Efficacy and neural mechanisms of approach bias modification training in patients with internet gaming disorder: A randomized clinical trial. J Affect Disord 2025; 376:355-365. [PMID: 39955074 DOI: 10.1016/j.jad.2025.02.026] [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: 08/30/2024] [Revised: 02/01/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Internet gaming disorder (IGD) is a prevalent behavioral addiction linked to neural alterations and significant negative outcomes. Approach bias modification (ApBM) training aims to correct imbalances in reflective and impulsive systems, reducing cravings and addictions. This study examined the effectiveness of ApBM training in IGD patients and explored the brain response changes associated with the intervention. METHODS Fifty-one patients with IGD were randomly assigned to an ApBM group (n = 26) or a sham-ApBM group (n = 25). Resting-state functional magnetic resonance imaging scans and behavioral assessments, including Internet Addiction Test scores, DSM-5 criteria, game craving levels, and automatic approach bias, were conducted before and after a ten-day training with five sessions. An analysis of variance (ANOVA) was employed to assess time (pre- and post-test) × group (ApBM group vs. sham-ApBM group) effects on behavioral measures. Functional connectivity (FC) analyses focused on regions of interest identified through regional homogeneity and degree centrality calculations. Additionally, we analyzed the relationship between neuroimaging variables and intervention outcomes. RESULTS Significant group × time interactions were found for automatic approach bias, Internet Addiction Test scores, DSM-5 criteria, and game craving levels. Post-training, these measures significantly decreased in the ApBM group but showed no significant changes in the sham-ApBM group. FC analysis revealed increased connectivity within executive control regions, enhanced connectivity between executive control and reward-related regions, and decreased connectivity within reward-related regions, exclusively in the ApBM group. CONCLUSIONS ApBM training effectively reduces gaming cravings in patients with IGD, enhancing executive control and mitigating impulsive behaviors.
Collapse
Affiliation(s)
- Jiejie Fu
- Department of Psychology, Yunnan Normal University, Kunming, Yunnan Province, China; Center for Cognition and Brain Disorders, the Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Xuefeng Xu
- Department of Psychology, Yunnan Normal University, Kunming, Yunnan Province, China
| | - Yi-Sheng Dong
- Department of Psychology and Behavioral Sciences, Zhejiang University, Hangzhou, China
| | - Min Wang
- Department of Psychology, Yunnan Normal University, Kunming, Yunnan Province, China
| | - Zhangzhushan Zhou
- Department of Psychology, Yunnan Normal University, Kunming, Yunnan Province, China
| | - Yijun Hu
- Department of Psychology, Yunnan Normal University, Kunming, Yunnan Province, China
| | - Qinxuan Li
- Department of Psychology, Yunnan Normal University, Kunming, Yunnan Province, China
| | - Shengjia Liu
- Department of Psychology, Yunnan Normal University, Kunming, Yunnan Province, China
| | - Weijie He
- Department of Psychology, Yunnan Normal University, Kunming, Yunnan Province, China
| | - Guang-Heng Dong
- Department of Psychology, Yunnan Normal University, Kunming, Yunnan Province, China.
| |
Collapse
|
2
|
Zhang W, Yang X, He B, He A, Li J, Zhang X, Zhang Y. Theobromine synergizes with nicotine to enhance animal motor abilities and cognitive function. Behav Pharmacol 2025:00008877-990000000-00125. [PMID: 40167572 DOI: 10.1097/fbp.0000000000000821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The societal consensus on the need for smoking cessation is well-established, yet the number of tobacco users continues to rise. This trend is largely driven by the potent physiological effects of nicotine, which significantly increase the likelihood of tobacco use initiation. Certain compounds found in everyday foods, such as theobromine and caffeine in cocoa products, also exhibit psychostimulatory properties. However, comprehensive studies on the effects of consuming these compounds in the context of tobacco use are limited. This study investigates the role of theobromine, a primary active component in cocoa, in modulating the positive physiological effects of nicotine and explores the underlying mechanisms. Our findings reveal that while low doses of theobromine do not alter nicotine's addictive properties, they amplify its positive physiological effects. Notably, theobromine's impact on nicotine varies significantly between the hippocampus and cerebellum, highlighting region-specific interactions.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Beijing Life Science Academy (BLSA), Beijing
- Key Laboratory of Tobacco Flavor Basic Research of CNTC, Zhengzhou Tobacco Research Institute of China National Tobacco Corporation, Zhengzhou
| | - Xiao Yang
- Key Laboratory of Tobacco Flavor Basic Research of CNTC, Zhengzhou Tobacco Research Institute of China National Tobacco Corporation, Zhengzhou
| | - Baojiang He
- Key Laboratory of Tobacco Flavor Basic Research of CNTC, Zhengzhou Tobacco Research Institute of China National Tobacco Corporation, Zhengzhou
| | - Aimin He
- Technology Center, China Tobacco Hebei Industrial Co., Ltd., Shijiazhuang, China
| | - Jiehui Li
- Technology Center, China Tobacco Hebei Industrial Co., Ltd., Shijiazhuang, China
| | - Xiaojing Zhang
- Technology Center, China Tobacco Hebei Industrial Co., Ltd., Shijiazhuang, China
| | - Yanfang Zhang
- Technology Center, China Tobacco Hebei Industrial Co., Ltd., Shijiazhuang, China
| |
Collapse
|
3
|
Lacomba‐Arnau E, Martínez‐Molina A, Barrós‐Loscertales A. Structural Cerebellar and Lateral Frontoparietal Networks are altered in CUD: An SBM Analysis. Addict Biol 2025; 30:e70021. [PMID: 40072344 PMCID: PMC11899759 DOI: 10.1111/adb.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 03/14/2025]
Abstract
Repetitive drug use results in enduring structural and functional changes in the brain. Addiction research has consistently revealed significant modifications in key brain networks related to reward, habit, salience, executive function, memory and self-regulation. Techniques like Voxel-based Morphometry have highlighted large-scale structural differences in grey matter across distinct groups. Source-based Morphometry (SBM) takes this a step further by incorporating the Independent Component Analysis to detect shared patterns of grey matter variation, all without requiring prior selection of regions of interest. However, SBM has yet to be employed in the study of structural alteration patterns related to cocaine addiction. Therefore, we performed this analysis to explore alterations in structural covariance specific to cocaine addiction. Our study involved 40 individuals diagnosed with Cocaine Use Disorder (CUD) and 40 matched healthy controls. Participants with CUD completed clinical questionnaires assessing the severity of their dependence and other relevant clinical variables. Following the adjustment for age-related effects, we observed notable disparities between groups in two structural independent components, which we identified as the structural cerebellar network and the structural lateral frontoparietal network, which display opposing trends. Specifically, the individuals with CUD exhibited a heightened contribution to the cerebellar network but simultaneously demonstrated a reduced contribution to the lateral frontoparietal network compared to the healthy controls. These findings unveil distinctive covariance patterns of neuroregulation linked with cocaine addiction, which indicates an interruption in the typical structural development in an affected lateral frontoparietal network, while suggesting an extended pattern of neuroregulation within the cerebellar network in individuals with CUD.
Collapse
Affiliation(s)
- Elena Lacomba‐Arnau
- Departament de Psicologia, Sociologia i Treball SocialUniversitat de LleidaLleidaSpain
- Department of Precision HealthLuxembourg Institute of HealthStrassenLuxembourg
| | | | | |
Collapse
|
4
|
Gironda SC, Centanni SW, Weiner JL. Early life psychosocial stress increases binge-like ethanol consumption and CSF1R inhibition prevents stress-induced alterations in microglia and brain macrophage population density. Brain Behav Immun Health 2025; 43:100933. [PMID: 39896839 PMCID: PMC11787031 DOI: 10.1016/j.bbih.2024.100933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/04/2024] [Accepted: 12/21/2024] [Indexed: 02/04/2025] Open
Abstract
Early life stress (ELS) has lasting consequences on microglia and brain macrophage function. During ELS, microglia and brain macrophages alter their engagement with synapses leading to changes in neuronal excitability. Further, ELS can induce innate immune memory formation in microglia and brain macrophages resulting in altered responsivity to future environmental stimuli. These alterations can result in lasting adaptations in circuit function and may mediate the relationship between ELS and the risk to develop alcohol use disorder (AUD). Whether microglia and brain macrophages truly mediate this relationship remains elusive. Here, we report: 1) an ELS model, psychosocial stress (PSS), increases binge-like ethanol consumption in early adulthood. 2) Repeated binge-like ethanol consumption increases microglia and brain macrophage population densities across the brain. 3) PSS may elicit innate immune memory formation in microglia and brain macrophages leading to altered population densities following repeated binge-like ethanol consumption. 4) Microglia and brain macrophage inhibition trended towards preventing PSS-evoked changes in binge-like ethanol consumption and normalized microglia and brain macrophage population densities. Therefore, our study suggests that acutely inhibiting microglia and brain macrophage function during periods of early life PSS may prevent innate immune memory formation and assist in reducing the risk to develop AUD.
Collapse
Affiliation(s)
| | - Samuel W. Centanni
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC, 27101, USA
| | | |
Collapse
|
5
|
Oka T, Kubo T, Kobayashi N, Murakami M, Chiba T, Cortese A. Decoding and modifying dynamic attentional bias in gaming disorder. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230090. [PMID: 39428882 PMCID: PMC11491851 DOI: 10.1098/rstb.2023.0090] [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: 11/06/2023] [Revised: 02/26/2024] [Accepted: 05/15/2024] [Indexed: 10/22/2024] Open
Abstract
With the spread of smartphones and computer games, concerns have escalated regarding the rising prevalence of gaming disorder. Patients often display attentional biases, unconsciously turning their attention towards gaming-related stimuli. However, attempts to discover and ameliorate these attentional deficits have yielded inconsistent outcomes, potentially due to the dynamic nature of attentional bias. This study investigated neural mechanisms underlying attentional bias state by combining neuroimaging (functional magnetic resonance imaging -fMRI) with an approach-avoidance task tailored to an individual's gaming preference. We conducted a multivariate pattern analysis of endogenous brain activity in 21 participants with probable gaming disorder. Our analyses revealed that activity patterns in the insula tracked temporal attentional bias states specific to gaming stimuli. A broad network of frontal and parietal regions instead appeared to predict a general temporal attentional bias state. Finally, we conducted a proof-of-concept study for 'just-in-time' attentional bias training through fMRI-decoded neurofeedback of insula activity patterns, named decoded attentional bias training (DecABT). Our preliminary results suggest that DecABT may help to decrease the attractiveness of gaming stimuli via a insula- and precuneus-based neural mechanism. This work provides new evidence for the insula as an endogenous regulator of attentional bias states in gaming disorder and a starting point to develop novel, individualized therapeutic approaches to treat addiction.This article is part of the theme issue 'Neurofeedback: new territories and neurocognitive mechanisms of endogenous neuromodulation'.
Collapse
Affiliation(s)
- Taiki Oka
- Department of Decoded Neurofeedback, Computational Neuroscience Laboratories, Advanced Telecommunications Research Institute International, Kyoto, Japan
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Clinical Psychology, Graduate School of Human Sciences, Osaka University, Suita, Japan
| | - Takatomi Kubo
- Department of Decoded Neurofeedback, Computational Neuroscience Laboratories, Advanced Telecommunications Research Institute International, Kyoto, Japan
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Nara, Japan
| | - Nao Kobayashi
- Healthcare Medical Group, Life Science Laboratories, KDDI Research, Inc., Saitama, Japan
| | - Misa Murakami
- Department of Decoded Neurofeedback, Computational Neuroscience Laboratories, Advanced Telecommunications Research Institute International, Kyoto, Japan
| | - Toshinori Chiba
- Department of Decoded Neurofeedback, Computational Neuroscience Laboratories, Advanced Telecommunications Research Institute International, Kyoto, Japan
- Department of Psychiatry, Self-Defense Forces Hanshin Hospital, Kawanishi, Japan
| | - Aurelio Cortese
- Department of Decoded Neurofeedback, Computational Neuroscience Laboratories, Advanced Telecommunications Research Institute International, Kyoto, Japan
| |
Collapse
|
6
|
Rodrigues RPS, Sousa SS, López-Caneda E, Almeida-Antunes N, González‑Villar AJ, Sampaio A, Crego A. Associative memory in alcohol-related contexts: An fMRI study with young binge drinkers. J Psychopharmacol 2024; 38:972-985. [PMID: 39373255 PMCID: PMC11528936 DOI: 10.1177/02698811241282624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
BACKGROUND Alcohol-related cues are known to influence craving levels, a hallmark of alcohol misuse. Binge drinking (BD), a pattern of heavy alcohol use, has been associated with cognitive and neurofunctional alterations, including alcohol attentional bias, memory impairments, as well as disrupted activity in prefrontal- and reward-related regions. However, literature is yet to explore how memories associated with alcohol-related cues are processed by BDs, and how the recall of this information may influence their reward processing. AIMS The present functional magnetic resonance imaging (fMRI) study aimed to investigate the neurofunctional signatures of BD during an associative memory task. METHOD In all, 36 university students, 20 BDs and 16 alcohol abstainers, were asked to memorize neutral objects paired with either alcohol or non-alcohol-related contexts. Subsequently, neutral stimuli were presented, and participants were asked to classify them as being previously paired with alcohol- or non-alcohol-related contexts. RESULTS While behavioral performance was similar in both groups, during the recall of alcohol-related cues, BDs showed increased brain activation in two clusters including the thalamus, globus pallidus and dorsal striatum, and cerebellum and occipital fusiform gyrus, respectively. CONCLUSION These findings suggest that BDs display augmented brain activity in areas responsible for mental imagery and reward processing when trying to recall alcohol-related cues, which might ultimately contribute to alcohol craving, even without being directly exposed to an alcohol-related context. These results highlight the importance of considering how alcohol-related contexts may influence alcohol-seeking behavior and, consequently, the maintenance or increase in alcohol use.
Collapse
|
7
|
Hoffman LJ, Foley JM, Leong JK, Sullivan-Toole H, Elliott BL, Olson IR. A Virtual In Vivo Dissection and Analysis of Socioaffective Symptoms Related to Cerebellum-Midbrain Reward Circuitry in Humans. J Neurosci 2024; 44:e1031242024. [PMID: 39256045 PMCID: PMC11466071 DOI: 10.1523/jneurosci.1031-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
Emerging research in nonhuman animals implicates cerebellar projections to the ventral tegmental area (VTA) in appetitive behaviors, but these circuits have not been characterized in humans. Here, we mapped cerebello-VTA white matter connectivity in a cohort of men and women using probabilistic tractography on diffusion imaging data from the Human Connectome Project. We uncovered the topographical organization of these connections by separately tracking from parcels of cerebellar lobule VI, crus I/II, vermis, paravermis, and cerebrocerebellum. Results revealed that connections between the cerebellum and VTA predominantly originate in the right cerebellar hemisphere, interposed nucleus, and paravermal cortex and terminate mostly ipsilaterally. Paravermal crus I sends the most connections to the VTA compared with other lobules. We discovered a mediolateral gradient of connectivity, such that the medial cerebellum has the highest connectivity with the VTA. Individual differences in microstructure were associated with measures of negative affect and social functioning. By splitting the tracts into quarters, we found that the socioaffective effects were driven by the third quarter of the tract, corresponding to the point at which the fibers leave the deep nuclei. Taken together, we produced detailed maps of cerebello-VTA structural connectivity for the first time in humans and established their relevance for trait differences in socioaffective regulation.
Collapse
Affiliation(s)
- Linda J Hoffman
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania 19122
| | - Julia M Foley
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania 19122
| | - Josiah K Leong
- Department of Psychological Science, University of Arkansas, Fayetteville, Arkansas 72701
| | - Holly Sullivan-Toole
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania 19122
| | - Blake L Elliott
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania 19122
| | - Ingrid R Olson
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania 19122
| |
Collapse
|
8
|
Manto M, Adamaszek M, Apps R, Carlson E, Guarque-Chabrera J, Heleven E, Kakei S, Khodakhah K, Kuo SH, Lin CYR, Joshua M, Miquel M, Mitoma H, Larry N, Péron JA, Pickford J, Schutter DJLG, Singh MK, Tan T, Tanaka H, Tsai P, Van Overwalle F, Yamashiro K. Consensus Paper: Cerebellum and Reward. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2169-2192. [PMID: 38769243 DOI: 10.1007/s12311-024-01702-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
Cerebellum is a key-structure for the modulation of motor, cognitive, social and affective functions, contributing to automatic behaviours through interactions with the cerebral cortex, basal ganglia and spinal cord. The predictive mechanisms used by the cerebellum cover not only sensorimotor functions but also reward-related tasks. Cerebellar circuits appear to encode temporal difference error and reward prediction error. From a chemical standpoint, cerebellar catecholamines modulate the rate of cerebellar-based cognitive learning, and mediate cerebellar contributions during complex behaviours. Reward processing and its associated emotions are tuned by the cerebellum which operates as a controller of adaptive homeostatic processes based on interoceptive and exteroceptive inputs. Lobules VI-VII/areas of the vermis are candidate regions for the cortico-subcortical signaling pathways associated with loss aversion and reward sensitivity, together with other nodes of the limbic circuitry. There is growing evidence that the cerebellum works as a hub of regional dysconnectivity across all mood states and that mental disorders involve the cerebellar circuitry, including mood and addiction disorders, and impaired eating behaviors where the cerebellum might be involved in longer time scales of prediction as compared to motor operations. Cerebellar patients exhibit aberrant social behaviour, showing aberrant impulsivity/compulsivity. The cerebellum is a master-piece of reward mechanisms, together with the striatum, ventral tegmental area (VTA) and prefrontal cortex (PFC). Critically, studies on reward processing reinforce our view that a fundamental role of the cerebellum is to construct internal models, perform predictions on the impact of future behaviour and compare what is predicted and what actually occurs.
Collapse
Affiliation(s)
- Mario Manto
- Service de Neurologie, Médiathèque Jean Jacquy, CHU-Charleroi, 6000, Charleroi, Belgium.
- Service Des Neurosciences, Université de Mons, 7000, Mons, Belgium.
- Unité Des Ataxies Cérébelleuses, CHU-Charleroi, Service Des Neurosciences, University of Mons, 7000, Mons, Belgium.
| | - Michael Adamaszek
- Department of Clinical and Cognitive Neurorehabilitation, Klinik Bavaria Kreischa, 01731, Kreischa, Germany
| | - Richard Apps
- School of Physiology, Pharmacology & Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Erik Carlson
- Department of Psychiatry and Behavioural Sciences, University of Washington, Seattle, WA, 98108, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
| | - Julian Guarque-Chabrera
- Área de Psicobiología, Facultat de Ciències de La Salut, Universitat Jaume I, 12071, Castellón de La Plana, Spain
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, 10461, USA
| | - Elien Heleven
- Faculty of Psychology and Center for Neuroscience, Vrije Universiteit Brussel, 1050, Brussels, Belgium
| | - Shinji Kakei
- Department of Anatomy and Physiology, Jissen Women's University, Tokyo, 191-8510, Japan
| | - Kamran Khodakhah
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, 10461, USA
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
- Initiative of Columbia Ataxia and Tremor, Columbia University Medical Center, New York, NY, 10032, USA
| | - Chi-Ying R Lin
- Alzheimer's Disease and Memory Disorders Center, Department of Neurology, Baylor College of Medicine, Houston, 77030 TX, USA
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, 77030 TX, USA
| | - Mati Joshua
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, Israel
| | - Marta Miquel
- Área de Psicobiología, Facultat de Ciències de La Salut, Universitat Jaume I, 12071, Castellón de La Plana, Spain
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, 10461, USA
| | - Hiroshi Mitoma
- Department of Medical Education, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Noga Larry
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, Israel
| | - Julie Anne Péron
- Clinical and Experimental Neuropsychology Laboratory, Department of Psychology and Educational Sciences, University of Geneva, 1205, Geneva, Switzerland
| | - Jasmine Pickford
- School of Physiology, Pharmacology & Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Dennis J L G Schutter
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Heidelberglaan 1, 3584 CS, Utrecht, The Netherlands
| | - Manpreet K Singh
- Psychiatry and Behavioral Sciences, University of California Davis, 2230 Stockton Blvd, Sacramento, CA, 95817, USA
| | - Tommy Tan
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Hirokazu Tanaka
- Faculty of Information Technology, Tokyo City University, Tokyo, 158-8557, Japan
| | - Peter Tsai
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, 75235, USA
- Departments of Neuroscience, Pediatrics, Psychiatry, UT Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Frank Van Overwalle
- Faculty of Psychology and Center for Neuroscience, Vrije Universiteit Brussel, 1050, Brussels, Belgium
| | - Kunihiko Yamashiro
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, 75235, USA
| |
Collapse
|
9
|
Ma L, Tao Q, Dang J, Sun J, Niu X, Zhang M, Kang Y, Wang W, Cheng J, Han S, Zhang Y. Altered local intrinsic neural activity and molecular architecture in internet use disorders. Brain Res Bull 2024; 216:111052. [PMID: 39173776 DOI: 10.1016/j.brainresbull.2024.111052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/20/2024] [Accepted: 08/17/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Internet gaming disorder (IGD) is mainly characterized by its core dysfunction in higher-order brain cortices involved in inhibitory control, whose neurobiological basis remains unclear. Then, we will investigate local intrinsic neural activity (INA) alterations in IGD, ascertain whether these potential alterations are related to clinical characteristics, and further explore the underlying molecular architecture. METHOD In this study, we performed the fractional amplitude of low-frequency fluctuation (fALFF) and regional homogeneity (ReHo) derived from resting-state functional magnetic resonance imaging (rs-fMRI) to explore the impact of IGD on local INA. Correlation analysis revealed the relationship between ReHo and fALFF in terms of group differences and clinical characteristics. Moreover, correlations between fALFF, ReHo, and PET- and SPECT-driven maps were investigated to elucidate the specific molecular architecture alternations in IGD. Finally, receiver operating characteristic curve (ROC) analysis was used to show the potential abilities of fALFF and ReHo in distinguishing individuals with IGD (IGDs) from healthy controls (HCs). RESULT Compared with HCs, IGDs revealed increased ReHo and fALFF in the prefrontal cortex. Significantly decreased ReHo was observed in the temporal lobe, occipital lobe, and cerebellum. In addition, the ReHo values in the cerebellum_7b_R were positively correlated with internet addiction severity. ROC curve analysis showed that ReHo and fALFF-altered brain regions could effectively distinguish IGDs from HCs. More importantly, cross-modal correlations revealed local INA changes in brain regions associated with the monoamine neurotransmitter system and the less studied cholinergic/GABAergic system. CONCLUSION These results suggest that local functional impairments are shown in the audiovisual and inhibitory control circuits in IGDs. This may be associated with underlying neurotransmitter system alterations. Therefore, this study provides the possibility of GABAergic receptor agonists and cholinergic receptor inhibitors for the treatment of IGD.
Collapse
Affiliation(s)
- Longyao Ma
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Qiuying Tao
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Jinghan Dang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Jieping Sun
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Xiaoyu Niu
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Mengzhe Zhang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Yimeng Kang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Weijian Wang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China.
| |
Collapse
|
10
|
Banei F, Aliaghaei A, Meftahi GH. The effect of chronic administration of oxycodone on the behavioral functions and histopathology in the cerebellum and striatum of adult male rats. 3 Biotech 2024; 14:225. [PMID: 39247457 PMCID: PMC11379841 DOI: 10.1007/s13205-024-04062-y] [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: 06/24/2024] [Accepted: 08/21/2024] [Indexed: 09/10/2024] Open
Abstract
Oxycodone is widely used for pain management and acts via binding to mu- and kappa opioid receptors. It was shown that extended oxycodone usage can result from the demyelination and degeneration of neurons through the stress response, which triggers apoptotic signaling pathways. The striatum and cerebellum are recognized as significant contributors to addiction; however, there is no report on the effect of oxycodone on the cerebellum and striatum and motor coordination. We treated rats daily with oxycodone at 15 mg/kg doses for thirty days. Motor performance and electromyography activity were then evaluated. Stereological methods were performed to assess the number of neurons in the cerebellum and striatum as well as immunohistochemistry for microgliosis and astrogliosis. Furthermore, the Sholl analysis method was utilized to evaluate the cellular structure of both microglia and astrocytes. Results of the rotarod test for motor coordination show no significant (P < 0.05) difference between the oxycodone subjects and those in the control group. In addition, open-field assessments indicated that the application of oxycodone did not alter the amount of distance covered (as an indicator of locomotion) or time spent in the central area (as an indicator of anxiety) (P < 0.001). The electromyography (EMG) test result showed that oxycodone caused a delay in the reaction of the muscular nerves (P < 0.001). Data and results from our experiment revealed that administering oxycodone did not affect astrogliosis and the number of neurons in the cerebellum and striatum (P < 0.05). In contrast, it altered neuromuscular function. In addition, oxycodone administration activated microglia in the cerebellum and striatum. In conclusion, we encourage more research on the adverse effects of oxycodone on the brain.
Collapse
Affiliation(s)
- Farzin Banei
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Present Address: Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
11
|
Genç B, Aslan K, Bako D, Delibalta S, Bilgici MNC. Cortical and subcortical structural changes in pediatric patients with infratentorial tumors. Diagn Interv Radiol 2024; 30:328-334. [PMID: 38836466 PMCID: PMC11590737 DOI: 10.4274/dir.2024.242652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
PURPOSE This study aimed to detect supratentorial cortical and subcortical morphological changes in pediatric patients with infratentorial tumors. METHODS The study included 24 patients aged 4-18 years who were diagnosed with primary infratentorial tumors and 41 age- and gender-matched healthy controls. Synthetic magnetization-prepared rapid gradient echo images of brain magnetic resonance imaging were generated using deep learning algorithms applied to T2-axial images. The cortical thickness, surface area, volume, and local gyrification index (LGI), as well as subcortical gray matter volumes, were automatically calculated. Surface-based morphometry parameters for the patient and control groups were compared using the general linear model, and volumes between subcortical structures were compared using the t-test and Mann-Whitney U test. RESULTS In the patient group, cortical thinning was observed in the left supramarginal, and cortical thickening was observed in the left caudal middle frontal (CMF), left fusiform, left lateral orbitofrontal, left lingual gyrus, right CMF, right posterior cingulate, and right superior frontal (P < 0.050). The patient group showed a volume reduction in the pars triangularis, paracentral, precentral, and supramarginal gyri of the left hemisphere (P < 0.05). A decreased surface area was observed in the bilateral superior frontal and cingulate gyri (P < 0.05). The patient group exhibited a decreased LGI in the right precentral and superior temporal gyri, left supramarginal, and posterior cingulate gyri and showed an increased volume in the bilateral caudate nucleus and hippocampus, while a volume reduction was observed in the bilateral putamen, pallidum, and amygdala (P < 0.05). The ventricular volume and tumor volume showed a positive correlation with the cortical thickness in the bilateral CMF while demonstrating a negative correlation with areas exhibiting a decreased LGI (P < 0.05). CONCLUSION Posterior fossa tumors lead to widespread morphological changes in cortical structures, with the most prominent pattern being hypogyria. CLINICAL SIGNIFICANCE This study illuminates the neurological impacts of infratentorial tumors in children, providing a foundation for future therapeutic strategies aimed at mitigating these adverse cortical and subcortical changes and improving patient outcomes.
Collapse
Affiliation(s)
- Barış Genç
- Ondokuz Mayıs University Faculty of Medicine, Department of Neuroradiology, Samsun, Türkiye
| | - Kerim Aslan
- Ondokuz Mayıs University Faculty of Medicine, Department of Neuroradiology, Samsun, Türkiye
| | - Derya Bako
- Ondokuz Mayıs University Faculty of Medicine, Department of Neuroradiology, Samsun, Türkiye
| | - Semra Delibalta
- Acıbadem University, Atakent Hospital, Clinic of Radiology, İstanbul, Türkiye
| | | |
Collapse
|
12
|
Ahmadi S, Majidi M, Koraei M, Vasef S. The Inflammation/NF-κB and BDNF/TrkB/CREB Pathways in the Cerebellum Are Implicated in the Changes in Spatial Working Memory After Both Morphine Dependence and Withdrawal in Rat. Mol Neurobiol 2024; 61:6721-6733. [PMID: 38347284 DOI: 10.1007/s12035-024-03993-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/27/2024] [Indexed: 08/22/2024]
Abstract
We aimed to explore the impact of the cerebellum on the decline in spatial working memory following morphine dependence and withdrawal. Two groups of male Wistar rats received intraperitoneal injections of either saline (1 ml/kg) or morphine (10 mg/kg) twice daily for 10 days, serving as the control and dependent groups. Additionally, a withdrawal group underwent a 30-day withdrawal period after the dependence phase. Spatial working memory was assessed using a Y maze test. ELISA and western blot were used to assess protein levels in the cerebellum. On day 1, morphine impaired spatial working memory, deteriorated further after 10 days of morphine use, and nearly returned to its initial level following a 30-day withdrawal period. On day 10, significant increases in TNF-α, IL-1β, and CXCL12 and a notable decrease in IL-10 levels were detected in the morphine-dependent group, which did not completely restore in the withdrawal group. The protein levels of CXCR4, TLR4, P2X7R, and NF-κB sharply increased in the morphine-dependent group. However, these levels almost returned to normal after withdrawal. In the morphine-dependent group, BDNF decreased, while TrkB and CREB1 increased noticeably. Nevertheless, after withdrawal, TrkB and CREB1 but not BDNF levels returned to normal. In the morphine-dependent group, both CACNA1 and KCNMA1 decreased significantly and after withdrawal, only KCNMA1 showed partial restoration, while CACNA1 did not. It can be concluded that inflammation/NF-κB and BDNF/TrkB/CREB pathways play key roles in neural adaptation within the cerebellum, contributing to the decline in spatial working memory after both morphine dependence and withdrawal.
Collapse
Affiliation(s)
- Shamseddin Ahmadi
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran.
| | - Mohammad Majidi
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| | - Maryam Koraei
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| | - Samira Vasef
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| |
Collapse
|
13
|
Gironda SC, Centanni SW, Weiner JL. Early life psychosocial stress increases binge-like ethanol consumption and CSF1R inhibition prevents stress-induced alterations in microglia and brain macrophage population density. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.27.605403. [PMID: 39211115 PMCID: PMC11361020 DOI: 10.1101/2024.07.27.605403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Early life stress (ELS) has lasting consequences on microglia and brain macrophage function. During ELS, microglia and brain macrophages alter their engagement with synapses leading to changes in neuronal excitability. Further, ELS can induce innate immune memory formation in microglia and brain macrophages resulting in altered responsivity to future environmental stimuli. These alterations can result in lasting adaptations in circuit function and may mediate the relationship between ELS and the risk to develop alcohol use disorder (AUD). Whether microglia and brain macrophages truly mediate this relationship remains elusive. Here, we report: 1) an ELS model, psychosocial stress (PSS), increases binge-like ethanol consumption in early adulthood. 2) Repeated binge-like ethanol consumption increases microglia and brain macrophage population densities across the brain. 3) PSS may elicit innate immune memory formation in microglia and brain macrophages leading to altered population densities following repeated binge-like ethanol consumption. 4) Microglia and brain macrophage inhibition trended towards preventing PSS-evoked changes in binge-like ethanol consumption and normalized microglia and brain macrophage population densities. Therefore, our study suggests that acutely inhibiting microglia and brain macrophage function during periods of early life PSS may prevent innate immune memory formation and assist in reducing the risk to develop AUD. Highlights An early life psychosocial stress (PSS) exposure increases ethanol consumptionMicroglial inhibition during PSS trends towards reducing ethanol consumptionBinge ethanol consumption increases microglial count and alters cell proximityEarly life PSS alters microglial responsivity to binge ethanol consumptionMicroglial inhibition may prevent microglial innate immune memory formation.
Collapse
|
14
|
Chen P, Wang J, Tang G, Chen G, Xiao S, Guo Z, Qi Z, Wang J, Wang Y. Large-scale network abnormality in behavioral addiction. J Affect Disord 2024; 354:743-751. [PMID: 38521138 DOI: 10.1016/j.jad.2024.03.034] [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: 08/22/2023] [Revised: 03/01/2024] [Accepted: 03/09/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Researchers have endeavored to ascertain the network dysfunction associated with behavioral addiction (BA) through the utilization of resting-state functional connectivity (rsFC). Nevertheless, the identification of aberrant patterns within large-scale networks pertaining to BA has proven to be challenging. METHODS Whole-brain seed-based rsFC studies comparing subjects with BA and healthy controls (HC) were collected from multiple databases. Multilevel kernel density analysis was employed to ascertain brain networks in which BA was linked to hyper-connectivity or hypo-connectivity with each prior network. RESULTS Fifty-six seed-based rsFC publications (1755 individuals with BA and 1828 HC) were included in the meta-analysis. The present study indicate that individuals with BAs exhibit (1) hypo-connectivity within the fronto-parietal network (FN) and hypo- and hyper-connectivity within the ventral attention network (VAN); (2) hypo-connectivity between the FN and regions of the VAN, hypo-connectivity between the VAN and regions of the FN and default mode network (DMN), hyper-connectivity between the DMN and regions of the FN; (3) hypo-connectivity between the reward system and regions of the sensorimotor network (SS), DMN and VAN; (4) hypo-connectivity between the FN and regions of the SS, hyper-connectivity between the VAN and regions of the SS. CONCLUSIONS These findings provide impetus for a conceptual framework positing a model of BA characterized by disconnected functional coordination among large-scale networks.
Collapse
Affiliation(s)
- Pan Chen
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Junjing Wang
- Department of Applied Psychology, Guangdong University of Foreign Studies, Guangzhou 510006, China
| | - Guixian Tang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Guanmao Chen
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Shu Xiao
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Zixuan Guo
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Zhangzhang Qi
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Jurong Wang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Ying Wang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China.
| |
Collapse
|
15
|
Hoffman LJ, Foley JM, Leong JK, Sullivan-Toole H, Elliott BL, Olson IR. An in vivo Dissection, and Analysis of Socio-Affective Symptoms related to Cerebellum-Midbrain Reward Circuitry in Humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.29.560239. [PMID: 38798382 PMCID: PMC11118266 DOI: 10.1101/2023.09.29.560239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Emerging research in non-human animals implicates cerebellar projections to the ventral tegmental area (VTA) in appetitive behaviors, but these circuits have not been characterized in humans. Here, we mapped cerebello-VTA white-matter connectivity in humans using probabilistic tractography on diffusion imaging data from the Human Connectome Project. We uncovered the topographical organization of these connections by separately tracking from parcels of cerebellar lobule VI, crus I/II, vermis, paravermis, and cerebrocerebellum. Results revealed that connections from the cerebellum to the VTA predominantly originate in the right hemisphere, interposed nucleus, and paravermal cortex, and terminate mostly ipsilaterally. Paravermal crus I sends the most connections to the VTA compared to other lobules. We discovered a medial-to-lateral gradient of connectivity, such that the medial cerebellum has the highest connectivity with the VTA. Individual differences in microstructure were associated with measures of negative affect and social functioning. By splitting the tracts into quarters, we found that the socio-affective effects were driven by the third quarter of the tract, corresponding to the point at which the fibers leave the deep nuclei. Taken together, we produced detailed maps of cerebello-VTA structural connectivity for the first time in humans and established their relevance for trait differences in socio-affective regulation.
Collapse
Affiliation(s)
- Linda J. Hoffman
- Temple University, Department of Psychology and Neuroscience, Philadelphia, PA, USA
| | - Julia M. Foley
- Temple University, Department of Psychology and Neuroscience, Philadelphia, PA, USA
| | - Josiah K. Leong
- University of Arkansas, Department of Psychological Science, Fayetteville, AR, USA
| | - Holly Sullivan-Toole
- Temple University, Department of Psychology and Neuroscience, Philadelphia, PA, USA
| | - Blake L. Elliott
- Temple University, Department of Psychology and Neuroscience, Philadelphia, PA, USA
| | - Ingrid R. Olson
- Temple University, Department of Psychology and Neuroscience, Philadelphia, PA, USA
| |
Collapse
|
16
|
Li CN, Keay KA, Henderson LA, Mychasiuk R. Re-examining the Mysterious Role of the Cerebellum in Pain. J Neurosci 2024; 44:e1538232024. [PMID: 38658164 PMCID: PMC11044115 DOI: 10.1523/jneurosci.1538-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 04/26/2024] Open
Abstract
Pain is considered a multidimensional experience that embodies not merely sensation, but also emotion and perception. As is appropriate for this complexity, pain is represented and processed by an extensive matrix of cortical and subcortical structures. Of these structures, the cerebellum is gaining increasing attention. Although association between the cerebellum and both acute and chronic pain have been extensively detailed in electrophysiological and neuroimaging studies, a deep understanding of what functions are mediated by these associations is lacking. Nevertheless, the available evidence implies that lobules IV-VI and Crus I are especially pertinent to pain processing, and anatomical studies reveal that these regions connect with higher-order structures of sensorimotor, emotional, and cognitive function. Therefore, we speculate that the cerebellum exerts a modulatory role in pain via its communication with sites of sensorimotor, executive, reward, and limbic function. On this basis, in this review, we propose numerous ways in which the cerebellum might contribute to both acute and chronic pain, drawing particular attention to emotional and cognitive elements of pain. In addition, we emphasise the importance of advancing our knowledge about the relationship between the cerebellum and pain by discussing novel therapeutic opportunities that capitalize on this association.
Collapse
Affiliation(s)
- Crystal N Li
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Kevin A Keay
- School of Medical Sciences (Neuroscience) and Brain and Mind Centre, University of Sydney, NSW 2006, Australia
| | - Luke A Henderson
- School of Medical Sciences (Neuroscience) and Brain and Mind Centre, University of Sydney, NSW 2006, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| |
Collapse
|
17
|
Kebschull JM, Casoni F, Consalez GG, Goldowitz D, Hawkes R, Ruigrok TJH, Schilling K, Wingate R, Wu J, Yeung J, Uusisaari MY. Cerebellum Lecture: the Cerebellar Nuclei-Core of the Cerebellum. CEREBELLUM (LONDON, ENGLAND) 2024; 23:620-677. [PMID: 36781689 PMCID: PMC10951048 DOI: 10.1007/s12311-022-01506-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/10/2022] [Indexed: 02/15/2023]
Abstract
The cerebellum is a key player in many brain functions and a major topic of neuroscience research. However, the cerebellar nuclei (CN), the main output structures of the cerebellum, are often overlooked. This neglect is because research on the cerebellum typically focuses on the cortex and tends to treat the CN as relatively simple output nuclei conveying an inverted signal from the cerebellar cortex to the rest of the brain. In this review, by adopting a nucleocentric perspective we aim to rectify this impression. First, we describe CN anatomy and modularity and comprehensively integrate CN architecture with its highly organized but complex afferent and efferent connectivity. This is followed by a novel classification of the specific neuronal classes the CN comprise and speculate on the implications of CN structure and physiology for our understanding of adult cerebellar function. Based on this thorough review of the adult literature we provide a comprehensive overview of CN embryonic development and, by comparing cerebellar structures in various chordate clades, propose an interpretation of CN evolution. Despite their critical importance in cerebellar function, from a clinical perspective intriguingly few, if any, neurological disorders appear to primarily affect the CN. To highlight this curious anomaly, and encourage future nucleocentric interpretations, we build on our review to provide a brief overview of the various syndromes in which the CN are currently implicated. Finally, we summarize the specific perspectives that a nucleocentric view of the cerebellum brings, move major outstanding issues in CN biology to the limelight, and provide a roadmap to the key questions that need to be answered in order to create a comprehensive integrated model of CN structure, function, development, and evolution.
Collapse
Affiliation(s)
- Justus M Kebschull
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | - Filippo Casoni
- Division of Neuroscience, San Raffaele Scientific Institute, and San Raffaele University, Milan, Italy
| | - G Giacomo Consalez
- Division of Neuroscience, San Raffaele Scientific Institute, and San Raffaele University, Milan, Italy
| | - Daniel Goldowitz
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Richard Hawkes
- Department of Cell Biology & Anatomy and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Tom J H Ruigrok
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Karl Schilling
- Department of Anatomy, Anatomy & Cell Biology, Rheinische Friedrich-Wilhelms-Universität, 53115, Bonn, Federal Republic of Germany
| | - Richard Wingate
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Joshua Wu
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Joanna Yeung
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Marylka Yoe Uusisaari
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-Son, Kunigami-Gun, Okinawa, 904-0495, Japan.
| |
Collapse
|
18
|
Rice LC, Rochowiak RN, Plotkin MR, Rosch KS, Mostofsky SH, Crocetti D. Sex Differences and Behavioral Associations with Typically Developing Pediatric Regional Cerebellar Gray Matter Volume. CEREBELLUM (LONDON, ENGLAND) 2024; 23:589-600. [PMID: 37382829 PMCID: PMC10986327 DOI: 10.1007/s12311-023-01569-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/15/2023] [Indexed: 06/30/2023]
Abstract
The cerebellum contributes to motor and higher-order control throughout neurodevelopment, with marked growth during childhood. Few studies have investigated differential associations of cerebellar morphometry with function in males and females. The present study examines sex differences in regional cerebellar gray matter volume (GMV) and the moderating effect of sex on the relationship between GMV and motor, cognitive, and emotional functions in a large cohort of typically developing (TD) children. Participants included 371 TD children (123 females, age 8-12 years). A convolutional neural network-based approach was employed for cerebellar parcellation. Volumes were harmonized using ComBat to adjust for hardware-induced variations. Regression analyses examined the effect of sex on GMV and whether sex moderated the relationship between GMV and motor, cognitive, and emotional functions. Males showed larger GMV in right lobules I-V, bilateral lobules VI, crus II/VIIb, and VIII, left lobule X, and vermis regions I-V and VIII-X. Greater motor function correlated with less vermis VI-VII GMV in females. Greater cognitive function correlated with greater left lobule VI GMV in females and less left lobule VI GMV in males. Finally, greater internalizing symptoms correlated with greater bilateral lobule IX GMV in females but less in males. These findings reveal sexually dimorphic patterns of cerebellar structure and associations with motor, cognitive, and emotional functions. Males generally show larger GMV than females. Larger GMV was associated with better cognitive functioning for females and better motor/emotional functioning for males.
Collapse
Affiliation(s)
- Laura C Rice
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, 716 N. Broadway, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Rebecca N Rochowiak
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, 716 N. Broadway, Baltimore, MD, 21205, USA
| | - Micah R Plotkin
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, 716 N. Broadway, Baltimore, MD, 21205, USA
| | - Keri S Rosch
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, 716 N. Broadway, Baltimore, MD, 21205, USA
- Neuropsychology Department, Kennedy Krieger Institute, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Stewart H Mostofsky
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, 716 N. Broadway, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Deana Crocetti
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, 716 N. Broadway, Baltimore, MD, 21205, USA.
| |
Collapse
|
19
|
Ge F, Wang Z, Yu W, Yuan X, Cai Q, Wang G, Li X, Xu X, Yang P, Fan Y, Chang J, Guan X. Activating Lobule VI PC TH+-Med Pathway in Cerebellum Blocks the Acquisition of Methamphetamine Conditioned Place Preference in Mice. J Neurosci 2024; 44:e1312232024. [PMID: 38331582 PMCID: PMC10941241 DOI: 10.1523/jneurosci.1312-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Cerebellum has been implicated in drug addiction; however, its underlying cellular populations and neuronal circuitry remain largely unknown. In the current study, we identified a neural pathway from tyrosine hydroxylase (TH)-positive Purkinje cells (PCTH+) in cerebellar lobule VI to calcium/calmodulin-dependent protein kinase II (CaMKII)-positive glutamatergic neurons in the medial cerebellar nucleus (MedCaMKII), forming the lobule VI PCTH+-MedCaMKII pathway in male mice. In naive male mice, inhibition of PCTH+ neurons activated Med neurons. During conditioned place preference (CPP) training, exposure to methamphetamine (METH) inhibited lobule VI PCTH+ neurons while excited MedCaMKII neurons in mice. Silencing MedCaMKII using a tetanus toxin light chain (tettox) suppressed the acquisition of METH CPP in mice but resulted in motor coordination deficits in naive mice. In contrast, activating lobule VI PCTH+ terminals within Med inhibited the activity of Med neurons and subsequently blocked the acquisition of METH CPP in mice without affecting motor coordination, locomotor activity, and sucrose reinforcements in naive mice. Our findings identified a novel lobule VI PCTH+-MedCaMKII pathway within the cerebellum and explored its role in mediating the acquisition of METH-preferred behaviors.
Collapse
Affiliation(s)
- Feifei Ge
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zilin Wang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wen Yu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiya Yuan
- The first Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Qinglong Cai
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guanxiong Wang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiang Li
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xing Xu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ping Yang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiasong Chang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
20
|
Fuchs BA, Pearce AL, Rolls BJ, Wilson SJ, Rose EJ, Geier CF, Garavan H, Keller KL. The Cerebellar Response to Visual Portion Size Cues Is Associated with the Portion Size Effect in Children. Nutrients 2024; 16:738. [PMID: 38474866 DOI: 10.3390/nu16050738] [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: 12/20/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The neural mechanisms underlying susceptibility to eating more in response to large portions (i.e., the portion size effect) remain unclear. Thus, the present study examined how neural responses to portion size relate to changes in weight and energy consumed as portions increase. Associations were examined across brain regions traditionally implicated in appetite control (i.e., an appetitive network) as well as the cerebellum, which has recently been implicated in appetite-related processes. Children without obesity (i.e., BMI-for-age-and-sex percentile < 90; N = 63; 55% female) viewed images of larger and smaller portions of food during fMRI and, in separate sessions, ate four meals that varied in portion size. Individual-level linear and quadratic associations between intake (kcal, grams) and portion size (i.e., portion size slopes) were estimated. The response to portion size in cerebellar lobules IV-VI was associated with the quadratic portion size slope estimated from gram intake; a greater response to images depicting smaller compared to larger portions was associated with steeper increases in intake with increasing portion sizes. Within the appetitive network, neural responses were not associated with portion size slopes. A decreased cerebellar response to larger amounts of food may increase children's susceptibility to overeating when excessively large portions are served.
Collapse
Affiliation(s)
- Bari A Fuchs
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Alaina L Pearce
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Barbara J Rolls
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Stephen J Wilson
- Department of Psychology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Emma J Rose
- Department of Psychology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Charles F Geier
- Human Development and Family Science, University of Georgia, Athens, GA 31793, USA
| | - Hugh Garavan
- Department of Psychological Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Kathleen L Keller
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Food Science, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
21
|
Zhu T, Wang W, Chen Y, Kranzler HR, Li CSR, Bi J. Machine Learning of Functional Connectivity to Biotype Alcohol and Nicotine Use Disorders. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:326-336. [PMID: 37696489 PMCID: PMC10976073 DOI: 10.1016/j.bpsc.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/23/2023] [Accepted: 08/28/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND Magnetic resonance imaging provides noninvasive tools to investigate alcohol use disorder (AUD) and nicotine use disorder (NUD) and neural phenotypes for genetic studies. A data-driven transdiagnostic approach could provide a new perspective on the neurobiology of AUD and NUD. METHODS Using samples of individuals with AUD (n = 140), individuals with NUD (n = 249), and healthy control participants (n = 461) from the UK Biobank, we integrated clinical, neuroimaging, and genetic markers to identify biotypes of AUD and NUD. We partitioned participants with AUD and NUD based on resting-state functional connectivity (FC) features associated with clinical metrics. A multitask artificial neural network was trained to evaluate the cluster-defined biotypes and jointly infer AUD and NUD diagnoses. RESULTS Three biotypes-primary NUD, mixed NUD/AUD with depression and anxiety, and mixed AUD/NUD-were identified. Multitask classifiers incorporating biotype knowledge achieved higher area under the curve (AUD: 0.76, NUD: 0.74) than single-task classifiers without biotype differentiation (AUD: 0.61, NUD: 0.64). Cerebellar FC features were important in distinguishing the 3 biotypes. The biotype of mixed NUD/AUD with depression and anxiety demonstrated the largest number of FC features (n = 5), all related to the visual cortex, that significantly differed from healthy control participants and were validated in a replication sample (p < .05). A polymorphism in TNRC6A was associated with the mixed AUD/NUD biotype in both the discovery (p = 7.3 × 10-5) and replication (p = 4.2 × 10-2) sets. CONCLUSIONS Biotyping and multitask learning using FC features can characterize the clinical and genetic profiles of AUD and NUD and help identify cerebellar and visual circuit markers to differentiate the AUD/NUD group from the healthy control group. These markers support a new growing body of literature.
Collapse
Affiliation(s)
- Tan Zhu
- Department of Computer Science and Engineering, School of Engineering, University of Connecticut, Storrs, Connecticut
| | - Wuyi Wang
- Data Analytics Department, Yale New Haven Health System, New Haven, Connecticut
| | - Yu Chen
- Department of Psychiatry, School of Medicine, Yale University, New Haven, Connecticut
| | - Henry R Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Chiang-Shan R Li
- Department of Psychiatry, School of Medicine, Yale University, New Haven, Connecticut; Department of Neuroscience, School of Medicine, Yale University, New Haven, Connecticut; Wu Tsai Institute, Yale University, New Haven, Connecticut
| | - Jinbo Bi
- Department of Computer Science and Engineering, School of Engineering, University of Connecticut, Storrs, Connecticut.
| |
Collapse
|
22
|
Washburn S, Oñate M, Yoshida J, Vera J, Bhuvanasundaram R, Khatami L, Nadim F, Khodakhah K. The cerebellum directly modulates the substantia nigra dopaminergic activity. Nat Neurosci 2024; 27:497-513. [PMID: 38272967 PMCID: PMC11441724 DOI: 10.1038/s41593-023-01560-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 12/13/2023] [Indexed: 01/27/2024]
Abstract
Evidence of direct reciprocal connections between the cerebellum and basal ganglia has challenged the long-held notion that these structures function independently. While anatomical studies have suggested the presence of cerebellar projections to the substantia nigra pars compacta (SNc), the nature and function of these connections (Cb-SNc) is unknown. Here we show, in mice, that Cb-SNc projections form monosynaptic glutamatergic synapses with dopaminergic and non-dopaminergic neurons in the SNc. Optogenetic activation of Cb-SNc axons in the SNc is associated with increased SNc activity, elevated striatal dopamine levels and increased locomotion. During behavior, Cb-SNc projections are bilaterally activated before ambulation and unilateral lever manipulation. Cb-SNc projections show prominent activation for water reward and higher activation for sweet water, suggesting that the pathway also encodes reward value. Thus, the cerebellum directly, rapidly and effectively modulates basal ganglia dopamine levels and conveys information related to movement initiation, vigor and reward processing.
Collapse
Affiliation(s)
- Samantha Washburn
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maritza Oñate
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Junichi Yoshida
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jorge Vera
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Leila Khatami
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Farzan Nadim
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, USA
| | - Kamran Khodakhah
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA.
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
23
|
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
|
24
|
Miller AP, Gizer IR. Neurogenetic and multi-omic sources of overlap among sensation seeking, alcohol consumption, and alcohol use disorder. Addict Biol 2024; 29:e13365. [PMID: 38380706 PMCID: PMC10882188 DOI: 10.1111/adb.13365] [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: 05/30/2023] [Revised: 09/26/2023] [Accepted: 11/26/2023] [Indexed: 02/22/2024]
Abstract
Sensation seeking is bidirectionally associated with levels of alcohol consumption in both adult and adolescent samples, and shared neurobiological and genetic influences may in part explain these associations. Links between sensation seeking and alcohol use disorder (AUD) may primarily manifest via increased alcohol consumption rather than through direct effects on increasing problems and consequences. Here the overlap among sensation seeking, alcohol consumption, and AUD was examined using multivariate modelling approaches for genome-wide association study (GWAS) summary statistics in conjunction with neurobiologically informed analyses at multiple levels of investigation. Meta-analytic and genomic structural equation modelling (GenomicSEM) approaches were used to conduct GWAS of sensation seeking, alcohol consumption, and AUD. Resulting summary statistics were used in downstream analyses to examine shared brain tissue enrichment of heritability and genome-wide evidence of overlap (e.g., stratified GenomicSEM, RRHO, genetic correlations with neuroimaging phenotypes), and to identify genomic regions likely contributing to observed genetic overlap across traits (e.g., H-MAGMA and LAVA). Across approaches, results supported shared neurogenetic architecture between sensation seeking and alcohol consumption characterised by overlapping enrichment of genes expressed in midbrain and striatal tissues and variants associated with increased cortical surface area. Alcohol consumption and AUD evidenced overlap in relation to variants associated with decreased frontocortical thickness. Finally, genetic mediation models provided evidence of alcohol consumption mediating associations between sensation seeking and AUD. This study extends previous research by examining critical sources of neurogenetic and multi-omic overlap among sensation seeking, alcohol consumption, and AUD which may underlie observed phenotypic associations.
Collapse
Affiliation(s)
- Alex P. Miller
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Ian R. Gizer
- Department of Psychological SciencesUniversity of MissouriColumbiaMissouriUSA
| |
Collapse
|
25
|
Luo D, He W, Shen D, Tang B, Tao H, Tang Q, Lai M, Liu J, Liu Y, Xu J, Meng J, Li J. Alterations in the brain functional network of abstinent male individuals with methamphetamine use disorder. Cereb Cortex 2024; 34:bhad523. [PMID: 38300175 DOI: 10.1093/cercor/bhad523] [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: 09/06/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 02/02/2024] Open
Abstract
Methamphetamine is a highly addictive psychostimulant drug that is abused globally and is a serious threat to health worldwide. Unfortunately, the specific mechanism underlying addiction remains unclear. Thus, this study aimed to investigate the characteristics of functional connectivity in the brain network and the factors influencing methamphetamine use disorder in patients using magnetic resonance imaging. We included 96 abstinent male participants with methamphetamine use disorder and 46 age- and sex-matched healthy controls for magnetic resonance imaging. Compared with healthy controls, participants with methamphetamine use disorder had greater impulsivity, fewer small-world attributes of the resting-state network, more nodal topological attributes in the cerebellum, greater functional connectivity strength within the cerebellum and between the cerebellum and brain, and decreased frontoparietal functional connectivity strength. In addition, after controlling for covariates, the partial correlation analysis showed that small-world properties were significantly associated with methamphetamine use frequency, psychological craving, and impulsivity. Furthermore, we revealed that the small-word attribute significantly mediated the effect of methamphetamine use frequency on motor impulsivity in the methamphetamine use disorder group. These findings may further improve our understanding of the neural mechanism of impulse control dysfunction underlying methamphetamine addiction and assist in exploring the neuropathological mechanism underlying methamphetamine use disorder-related dysfunction and rehabilitation.
Collapse
Affiliation(s)
- Dan Luo
- Mental Health Center, West China Hospital of Sichuan University, No. 28 Dian Xin Nan Jie, Wuhou District, Chengdu, China
| | - Wanlin He
- Radiology Department, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region (Hospital. C.T.), No. 20, Xi Mian Qiao Heng Jie, Wuhou District, Chengdu, China
| | - Danlin Shen
- Mental Health Center, West China Hospital of Sichuan University, No. 28 Dian Xin Nan Jie, Wuhou District, Chengdu, China
| | - Bin Tang
- Chengdu Compulsory Detoxification Center, No. 9 Xue Fu Lu, Shuangliu District, Chengdu, China
| | - Hongge Tao
- Chengdu Compulsory Detoxification Center, No. 9 Xue Fu Lu, Shuangliu District, Chengdu, China
| | - Qiao Tang
- Mental Health Center, West China Hospital of Sichuan University, No. 28 Dian Xin Nan Jie, Wuhou District, Chengdu, China
| | - Mingfeng Lai
- Mental Health Center, West China Hospital of Sichuan University, No. 28 Dian Xin Nan Jie, Wuhou District, Chengdu, China
| | - Jun Liu
- Sichuan Drug Rehabilitation Administration, No. 90 Shu Tong Jie, Jinniu District, Chengdu, China
| | - Yishan Liu
- Sichuan Drug Rehabilitation Administration, No. 90 Shu Tong Jie, Jinniu District, Chengdu, China
| | - Jiajun Xu
- Mental Health Center, West China Hospital of Sichuan University, No. 28 Dian Xin Nan Jie, Wuhou District, Chengdu, China
| | - Jinli Meng
- Radiology Department, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region (Hospital. C.T.), No. 20, Xi Mian Qiao Heng Jie, Wuhou District, Chengdu, China
| | - Jing Li
- Mental Health Center, West China Hospital of Sichuan University, No. 28 Dian Xin Nan Jie, Wuhou District, Chengdu, China
| |
Collapse
|
26
|
Kupnicka P, Listos J, Tarnowski M, Kolasa A, Kapczuk P, Surówka A, Kwiatkowski J, Janawa K, Chlubek D, Baranowska-Bosiacka I. The Effect of Prenatal and Neonatal Fluoride Exposure to Morphine-Induced Neuroinflammation. Int J Mol Sci 2024; 25:826. [PMID: 38255899 PMCID: PMC10815549 DOI: 10.3390/ijms25020826] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/01/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Physical dependence is associated with the formation of neuroadaptive changes in the central nervous system (CNS), both at the molecular and cellular levels. Various studies have demonstrated the immunomodulatory and proinflammatory properties of morphine. The resulting neuroinflammation in drug dependence exacerbates substance abuse-related behaviors and increases morphine tolerance. Studies prove that fluoride exposure may also contribute to the development of neuroinflammation and neurodegenerative changes. Morphine addiction is a major social problem. Neuroinflammation increases tolerance to morphine, and neurodegenerative effects caused by fluoride in structures related to the development of dependence may impair the functioning of neuronal pathways, change the concentration of neurotransmitters, and cause memory and learning disorders, which implies this element influences the development of dependence. Therefore, our study aimed to evaluate the inflammatory state of selected brain structures in morphine-dependent rats pre-exposed to fluoride, including changes in cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) expression as well as microglial and astroglial activity via the evaluation of Iba1 and GFAP expression. We provide evidence that both morphine administration and fluoride exposure have an impact on the inflammatory response by altering the expression of COX-1, COX-2, ionized calcium-binding adapter molecule (Iba1), and glial fibrillary acidic protein (GFAP) in brain structures involved in dependence development, such as the prefrontal cortex, striatum, hippocampus, and cerebellum. We observed that the expression of COX-1 and COX-2 in morphine-dependent rats is influenced by prior fluoride exposure, and these changes vary depending on the specific brain region. Additionally, we observed active astrogliosis, as indicated by increased GFAP expression, in all brain structures of morphine-dependent rats, regardless of fluoride exposure. Furthermore, the effect of morphine on Iba1 expression varied across different brain regions, and fluoride pre-exposure may influence microglial activation. However, it remains unclear whether these changes are a result of the direct or indirect actions of morphine and fluoride on the factors analyzed.
Collapse
Affiliation(s)
- Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| | - Agnieszka Kolasa
- Department of Histology and Embryology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Anna Surówka
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Jakub Kwiatkowski
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Kamil Janawa
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
27
|
Lv Q, Zhang M, Jiang H, Liu Y, Zhao S, Xu X, Zhang W, Chen T, Su H, Zhang J, Wang H, Zhang J, Feng Y, Li Y, Li B, Zhao M, Wang Z. Metabolic and functional substrates of impulsive decision-making in individuals with heroin addiction after prolonged methadone maintenance treatment. Neuroimage 2023; 283:120421. [PMID: 37879424 DOI: 10.1016/j.neuroimage.2023.120421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/08/2023] [Accepted: 10/23/2023] [Indexed: 10/27/2023] Open
Abstract
Elevated impulsivity has been frequently reported in individuals with opioid addiction receiving methadone maintenance therapy (MMT), but the underlying neural mechanisms and cognitive subprocesses are not fully understood. We acquired functional magnetic resonance imaging (fMRI) data from 37 subjects with heroin addiction receiving long-term MMT and 33 healthy controls who performed a probabilistic reversal learning task, and measured their resting-state brain glucose using fluorine-18-fluorodeoxyglucose positron emission tomography (18F-FDG PET). Subjects receiving MMT exhibited significantly elevated self-reported impulsivity, and computational modeling revealed a marked impulsive decision bias manifested as switching more frequently without available evidence. Moreover, this impulsive decision bias was associated with the dose and duration of methadone use, irrelevant to the duration of heroin use. During the task, the switch-related hypoactivation in the left rostral middle frontal gyrus was correlated with the impulsive decision bias while the function of reward sensitivity was intact in subjects receiving MMT. Using prior brain-wide receptor density data, we found that the highest variance of regional metabolic abnormalities was explained by the spatial distribution of μ-opioid receptors among 10 types of neurotransmitter receptors. Heightened impulsivity in individuals receiving prolonged MMT is manifested as atypical choice bias and noise in decision-making processes, which is further driven by deficits in top-down cognitive control, other than reward sensitivity. Our findings uncover multifaceted mechanisms underlying elevated impulsivity in subjects receiving MMT, which might provide insights for developing complementary therapies to improve retention during MMT.
Collapse
Affiliation(s)
- Qian Lv
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental Health, IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Miao Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haifeng Jiang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yilin Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, China
| | - Shaoling Zhao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, China
| | - Xiaomin Xu
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenlei Zhang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tianzhen Chen
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hang Su
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiangtao Zhang
- Tongde Hospital of Zhejiang Province (Zhejiang Mental Health Center), Zhejiang Office of Mental Health, Hangzhou, China
| | - Heqiu Wang
- Tongde Hospital of Zhejiang Province (Zhejiang Mental Health Center), Zhejiang Office of Mental Health, Hangzhou, China
| | - Jianmin Zhang
- Tongde Hospital of Zhejiang Province (Zhejiang Mental Health Center), Zhejiang Office of Mental Health, Hangzhou, China
| | - Yuanjing Feng
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Yongqiang Li
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Min Zhao
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Zheng Wang
- School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental Health, IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
28
|
Mohammadpanah M, Farrokhi S, Sani M, Moghaddam MH, Bayat AH, Boroujeni ME, Abdollahifar MA, Fathi M, Vakili K, Nikpour F, Omran HS, Ahmadirad H, Ghorbani Z, Peyvandi AA, Aliaghaei A. Exposure to Δ9-tetrahydrocannabinol leads to a rise in caspase-3, morphological changes in microglial, and astrocyte reactivity in the cerebellum of rats. Toxicol Res (Camb) 2023; 12:1077-1094. [PMID: 38145099 PMCID: PMC10734605 DOI: 10.1093/toxres/tfad098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/21/2023] [Accepted: 10/06/2023] [Indexed: 12/26/2023] Open
Abstract
The present study aimed to elucidate the effect of 10 mg/kg Δ9-tetrahydrocannabinol (THC) on cerebellar neuronal and glial morphology, apoptosis and inflammatory gene expression using a series of histological assays including stereology, Sholl analysis, immunofluorescence and real-time qPCR in male Wistar rats. A decrease in the number of Purkinje neurons and the thickness of the granular layer in the cerebellum was reported in THC-treated rats. Increased expression of Iba-1 and arborization of microglial processes were evidence of microgliosis and morphological changes in microglia. In addition, astrogliosis and changes in astrocyte morphology were other findings associated with THC administration. THC also led to an increase in caspase-3 positive cells and a decrease in autophagy and inflammatory gene expression such as mTOR, BECN1 and LAMP2. However, there were no significant changes in the volume of molecular layers and white matter, the spatial arrangement of granular layers and white matter, or the spatial arrangement of granular layers and white matter in the cerebellum. Taken together, our data showed both neuroprotective and neurodegenerative properties of THC in the cerebellum, which require further study in the future.
Collapse
Affiliation(s)
- Mojtaba Mohammadpanah
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sheida Farrokhi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Sani
- Department of Educational Neuroscience, Aras International Campus, University of Tabriz, Tabriz, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Amir-Hossein Bayat
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahdi Eskandarian Boroujeni
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nikpour
- Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Salehi Omran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Ahmadirad
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeynab Ghorbani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Asghar Peyvandi
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Zeng Y, Wu GR, Xue Y, Baeken C, Wei L. The moderating effect of resting heart rate variability on the relationship between internet addiction tendency and brain morphology. Addict Biol 2023; 28:e13340. [PMID: 37855073 DOI: 10.1111/adb.13340] [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: 06/12/2023] [Revised: 08/09/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023]
Abstract
Previous neuroimaging studies have investigated brain morphology associated with internet addiction tendency (IAT) in healthy subjects. However, whether resting vagally-mediated heart rate variability (HRV) exerting influences on the association of IAT and brain morphology remains unclear. This study used voxel-based morphometry (VBM) and multiple regression analyses to assess the interaction effect of IAT and resting vagally-mediated HRV on regional grey matter volumes in 82 healthy subjects. To further illustrate the observed interaction effect, the moderated hierarchical regression analysis was performed. The results showed that resting vagally-mediated HRV moderated the relationship between IAT scores and grey matter volume (GMV) in the precuneus and cerebellum. Specifically, individuals with higher resting vagally-mediated HRV showed a significant positive relationship between IAT scores and GMV in the precuneus, whereas individuals with lower resting vagally-mediated HRV showed a significant negative relationship between IAT scores and GMV in the precuneus. In addition, IAT scores were negatively correlated with GMV in the cerebellum among individuals with lower resting vagally-mediated HRV, but not among individuals with higher resting vagally-mediated HRV. These findings have demonstrated a moderating role of resting vagally-mediated HRV on the association of IAT and brain morphology.
Collapse
Affiliation(s)
- Yuandong Zeng
- School of Psychology, Jiangxi Normal University, Nanchang, China
| | - Guo-Rong Wu
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Southwest University, Chongqing, China
- Faculty of Medicine and Health Sciences, Department of Head and Skin, Ghent Experimental Psychiatry (GHEP) lab, Ghent University, Ghent, Belgium
| | - Yingying Xue
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Southwest University, Chongqing, China
| | - Chris Baeken
- Faculty of Medicine and Health Sciences, Department of Head and Skin, Ghent Experimental Psychiatry (GHEP) lab, Ghent University, Ghent, Belgium
- Department of Psychiatry, University Hospital (UZBrussel), Brussels, Belgium
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Luqing Wei
- School of Psychology, Jiangxi Normal University, Nanchang, China
| |
Collapse
|
30
|
Mundorf A, Siebert A, Desmond JE, Peterburs J. The role of the cerebellum in internet gaming disorder-A systematic review. Addict Biol 2023; 28:e13331. [PMID: 37753565 PMCID: PMC10662475 DOI: 10.1111/adb.13331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/30/2023] [Accepted: 08/15/2023] [Indexed: 09/28/2023]
Abstract
Recent studies increasingly highlight involvement of the cerebellum in drug craving and addiction. However, its exact role, that is, whether the cerebellum is a critical component of a brain network underlying addictive behaviour, or whether it rather is a facilitator or mediator, is still unclear. Findings concerning the newly recognized internet gaming disorder (IGD) suggest that changes in cerebellar connectivity and functioning are associated with behavioural/non-substance addiction. Here, we systematically review the literature on IGD and cerebellar involvement following the PRISMA guidelines. A total of 13 neuroimaging studies met the inclusion criteria. Studies utilized a broad range of diagnostic instruments and resulting cut-off criteria, rendering it difficult to compare findings. Results on altered cerebro-cerebellar connectivity in patients with IGD are mixed; most studies report altered or increased functional connectivity. Moreover, decreased cerebellar grey matter volume is reported. Studies have further indicated that differential activation patterns in the cerebellum may enable discrimination between healthy subjects and subjects with IGD, even allowing for prediction of treatment outcomes. Given the strong connectivity between the cerebellum and cerebral regions, the cerebellum may act as an intermediary between regions involved in craving and addiction and consequently affect symptoms of IGD. Results suggest differential involvement of the cerebellar lobes, emphasizing a need for high-resolution parcellation of the cerebellum in future studies. However, the studies included in the present review have small sample sizes and include mostly male participants. Thus, results may have limited generalizability yet highlight a crucial role of the cerebellum in IGD that needs further investigation.
Collapse
Affiliation(s)
- Annakarina Mundorf
- Institute for Systems Medicine & Department of Human Medicine, MSH Medical School Hamburg, Germany
| | - Annabelle Siebert
- Institute for Systems Medicine & Department of Human Medicine, MSH Medical School Hamburg, Germany
| | - John E. Desmond
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jutta Peterburs
- Institute for Systems Medicine & Department of Human Medicine, MSH Medical School Hamburg, Germany
| |
Collapse
|
31
|
Zamudio PA, Gioia D, Glaser C, Woodward JJ. Chemogenetic Perturbation of the Posterior But Not Anterior Cerebellum Reduces Voluntary Ethanol Consumption. eNeuro 2023; 10:ENEURO.0037-23.2023. [PMID: 37679043 PMCID: PMC10512884 DOI: 10.1523/eneuro.0037-23.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/09/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
The cerebellum communicates with brain areas critically involved in control of goal-directed behaviors including the prefrontal and orbitofrontal cortices and midbrain and basal ganglia structures. In particular, the posterior cerebellum is important for cognitive flexibility and has been implicated in alcohol and drug-related memory. We hypothesized that the cerebellum, through its multiple connections to reward-related brain circuitry, regulates alcohol consumption. To test this, we expressed inhibitory designer receptors exclusively activated by designer drugs (DREADDs) in molecular layer interneurons (MLIs) in anterior (IV-V) or posterior (VI-VIII) cerebellar lobules of male and female mice and activated them during alcohol drinking sessions. In a home-cage drinking paradigm, alcohol consumption was significantly decreased by clozapine-N-oxide (CNO) or deschloroclozapine (DCZ) administration in male mice expressing DREADDs in posterior but not anterior lobules. CNO/DCZ injections did not affect drinking in DREADD expressing female mice or in male mice expressing the control vector. Activation of DREADDs expressed in anterior or posterior lobules had no effect on sucrose or quinine consumption in male or female mice. During operant self-administration sessions, DCZ decreased the number of licks and bouts in male but not female mice expressing DREADDs in posterior lobules with no effect in control vector mice. Performance on an accelerated rotarod was unaffected by chemogenetic manipulation while distance traveled in the open field was decreased by DREADD activation in anterior but not posterior lobules. These results indicate that neuronal activity within the posterior cerebellar cortex plays an important role in the control of alcohol consumption in male mice.
Collapse
Affiliation(s)
- Paula A Zamudio
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Dominic Gioia
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Christina Glaser
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
32
|
Valeri J, Gisabella B, Pantazopoulos H. Dynamic regulation of the extracellular matrix in reward memory processes: a question of time. Front Cell Neurosci 2023; 17:1208974. [PMID: 37396928 PMCID: PMC10311570 DOI: 10.3389/fncel.2023.1208974] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Substance use disorders are a global health problem with increasing prevalence resulting in significant socioeconomic burden and increased mortality. Converging lines of evidence point to a critical role of brain extracellular matrix (ECM) molecules in the pathophysiology of substance use disorders. An increasing number of preclinical studies highlight the ECM as a promising target for development of novel cessation pharmacotherapies. The brain ECM is dynamically regulated during learning and memory processes, thus the time course of ECM alterations in substance use disorders is a critical factor that may impact interpretation of the current studies and development of pharmacological therapies. This review highlights the evidence for the involvement of ECM molecules in reward learning, including drug reward and natural reward such as food, as well as evidence regarding the pathophysiological state of the brain's ECM in substance use disorders and metabolic disorders. We focus on the information regarding time-course and substance specific changes in ECM molecules and how this information can be leveraged for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
33
|
Melchor-Eixea I, Guarque-Chabrera J, Sanchez-Hernandez A, Ibáñez-Marín P, Pastor R, Miquel M. Putting forward a model for the role of the cerebellum in cocaine-induced pavlovian memory. Front Syst Neurosci 2023; 17:1154014. [PMID: 37388941 PMCID: PMC10303950 DOI: 10.3389/fnsys.2023.1154014] [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: 01/30/2023] [Accepted: 05/25/2023] [Indexed: 07/01/2023] Open
Abstract
Substance Use Disorder (SUD) involves emotional, cognitive, and motivational dysfunction. Long-lasting molecular and structural changes in brain regions functionally and anatomically linked to the cerebellum, such as the prefrontal cortex, amygdala, hippocampus, basal ganglia, and ventral tegmental area, are characteristic of SUD. Direct and indirect reciprocal connectivity between the cerebellum and these brain regions can explain cerebellar roles in Pavlovian and reinforcement learning, fear memory, and executive functions. It is increasingly clear that the cerebellum modulates brain functions altered in SUD and other neuropsychiatric disorders that exhibit comorbidity with SUD. In the present manuscript, we review and discuss this evidence and present new research exploring the role of the cerebellum in cocaine-induced conditioned memory using chemogenetic tools (designer receptor exclusively activated by designer drug, DREADDs). Our preliminary data showed that inactivation of a region that includes the interposed and lateral deep cerebellar nuclei reduces the facilitating effect of a posterior vermis lesion on cocaine-induced preference conditioning. These findings support our previous research and suggest that posterior vermis damage may increase drug impact on the addiction circuitry by regulating activity in the DCN. However, they raise further questions that will also be discussed.
Collapse
|
34
|
Liang G, Li X, Yuan H, Sun M, Qin S, Wei B. Abnormal static and dynamic amplitude of low-frequency fluctuations in multiple brain regions of methamphetamine abstainers. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:13318-13333. [PMID: 37501489 DOI: 10.3934/mbe.2023593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Methamphetamine (meth) addiction is a significant social and public health problem worldwide. The relapse rate of meth abstainers is significantly high, but the underlying physiological mechanisms are unclear. Therefore, in this study, we performed resting-state functional magnetic resonance imaging (rs-fMRI) analysis to detect differences in the spontaneous neural activity between the meth abstainers and the healthy controls, and identify the physiological mechanisms underlying the high relapse rate among the meth abstainers. The fluctuations and time variations in the blood oxygenation level-dependent (BOLD) signal of the local brain activity was analyzed from the pre-processed rs-fMRI data of 11 meth abstainers and 11 healthy controls and estimated the amplitude of low-frequency fluctuations (ALFF) and the dynamic ALFF (dALFF). In comparison with the healthy controls, meth abstainers showed higher ALFF in the anterior central gyrus, posterior central gyrus, trigonal-inferior frontal gyrus, middle temporal gyrus, dorsolateral superior frontal gyrus, and the insula, and reduced ALFF in the paracentral lobule and middle occipital gyrus. Furthermore, the meth abstainers showed significantly reduced dALFF in the supplementary motor area, orbital inferior frontal gyrus, middle frontal gyrus, medial superior frontal gyrus, middle occipital gyrus, insula, middle temporal gyrus, anterior central gyrus, and the cerebellum compared to the healthy controls ($ P < 0.05 $). These data showed abnormal spontaneous neural activity in several brain regions related to the cognitive, executive, and other social functions in the meth abstainers and potentially represent the underlying physiological mechanisms that are responsible for the high relapse rate. In conclusion, a combination of ALFF and dALFF analytical methods can be used to estimate abnormal spontaneous brain activity in the meth abstainers and make a more reasonable explanation for the high relapse rate of meth abstainers.
Collapse
Affiliation(s)
- Guixiang Liang
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
| | - Xiang Li
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
| | - Hang Yuan
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
| | - Min Sun
- Affiliation Shandong Detoxification Monitoring and Treatment Institute, Zibo 255000, China
| | - Sijun Qin
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
| | - Benzheng Wei
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266000, China
| |
Collapse
|
35
|
Lorenzetti V, Gaillard A, Beyer E, Kowalczyk M, Kamboj SK, Manning V, Gleeson J. Do mindfulness-based interventions change brain function in people with substance dependence? A systematic review of the fMRI evidence. BMC Psychiatry 2023; 23:407. [PMID: 37286936 DOI: 10.1186/s12888-023-04789-7] [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: 12/19/2022] [Accepted: 04/14/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND Substance use disorders (SUDs) affect ~ 35 million people globally and are associated with strong cravings, stress, and brain alterations. Mindfulness-based interventions (MBIs) can mitigate the adverse psychosocial outcomes of SUDs, but the underlying neurobiology is unclear. Emerging findings were systematically synthesised from fMRI studies about MBI-associated changes in brain function in SUDs and their associations with mindfulness, drug quantity, and craving. METHODS PsycINFO, Medline, CINAHL, PubMed, Scopus, and Web of Science were searched. Seven studies met inclusion criteria. RESULTS Group by time effects indicated that MBIs in SUDs (6 tobacco and 1 opioid) were associated with changes in the function of brain pathways implicated in mindfulness and addiction (e.g., anterior cingulate cortex and striatum), which correlated with greater mindfulness, lower craving and drug quantity. CONCLUSIONS The evidence for fMRI-related changes with MBI in SUD is currently limited. More fMRI studies are required to identify how MBIs mitigate and facilitate recovery from aberrant brain functioning in SUDs.
Collapse
Affiliation(s)
- Valentina Lorenzetti
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Level 5 Daniel Mannix Building, 115 Victoria Parade, Fitzroy, VIC, 3065, Australia.
| | - Alexandra Gaillard
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Level 5 Daniel Mannix Building, 115 Victoria Parade, Fitzroy, VIC, 3065, Australia
- Centre for Mental Health, Swinburne University of Technology, Hawthorn, Australia
| | - Emillie Beyer
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Level 5 Daniel Mannix Building, 115 Victoria Parade, Fitzroy, VIC, 3065, Australia
| | - Magdalena Kowalczyk
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Level 5 Daniel Mannix Building, 115 Victoria Parade, Fitzroy, VIC, 3065, Australia
| | - Sunjeev K Kamboj
- Research Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Victoria Manning
- Monash Addiction Research Centre, Eastern Health Clinical School, Monash University, Melbourne, Australia
- Turning Point, Eastern Health, Monash University, Melbourne, Australia
| | - John Gleeson
- Digital Innovations in Mental Health and Well-being Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Australian Catholic University, Melbourne, VIC, Australia
| |
Collapse
|
36
|
Miller AP, Gizer IR. Neurogenetic and multi-omic sources of overlap among sensation seeking, alcohol consumption, and alcohol use disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.30.23290733. [PMID: 37333128 PMCID: PMC10274973 DOI: 10.1101/2023.05.30.23290733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Sensation seeking is bidirectionally associated with levels of alcohol consumption in both adult and adolescent samples and shared neurobiological and genetic influences may in part explain this association. Links between sensation seeking and alcohol use disorder (AUD) may primarily manifest via increased alcohol consumption rather than through direct effects on increasing problems and consequences. Here the overlap between sensation seeking, alcohol consumption, and AUD was examined using multivariate modeling approaches for genome-wide association study (GWAS) summary statistics in conjunction with neurobiologically-informed analyses at multiple levels of investigation. Meta-analytic and genomic structural equation modeling (GenomicSEM) approaches were used to conduct GWAS of sensation seeking, alcohol consumption, and AUD. Resulting summary statistics were used in downstream analyses to examine shared brain tissue enrichment of heritability and genome-wide evidence of overlap (e.g., stratified GenomicSEM, RRHO, genetic correlations with neuroimaging phenotypes) and to identify genomic regions likely contributing to observed genetic overlap across traits (e.g., HMAGMA, LAVA). Across approaches, results supported shared neurogenetic architecture between sensation seeking and alcohol consumption characterized by overlapping enrichment of genes expressed in midbrain and striatal tissues and variants associated with increased cortical surface area. Alcohol consumption and AUD evidenced overlap in relation to variants associated with decreased frontocortical thickness. Finally, genetic mediation models provided evidence of alcohol consumption mediating associations between sensation seeking and AUD. This study extends previous research by examining critical sources of neurogenetic and multi-omic overlap among sensation seeking, alcohol consumption, and AUD which may underlie observed phenotypic associations.
Collapse
Affiliation(s)
- Alex P. Miller
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Ian R. Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
37
|
Francis AN, Camprodon JA, Filbey F. Functional hyperconnectivity between corticocerebellar networks and altered decision making in young adult cannabis users: Evidence from 7T and multivariate pattern analysis. Psychiatry Res Neuroimaging 2023; 331:111613. [PMID: 36924741 DOI: 10.1016/j.pscychresns.2023.111613] [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: 12/01/2022] [Revised: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 03/18/2023]
Abstract
Decision-making (DM) impairments are important predictors of cannabis initiation and continued use. In cannabis users, how decision-making abnormalities related to structural and functional connectivity in the brain are not fully understood. We employed a three-method multimodal image analysis and multivariate pattern analysis (MVPA) on high dimensional 7 tesla MRI images examining functional connectivity, white matter microstructure and gray matter volume in a group of cannabis users and non-users. Neuroimaging and cognitive analyses were performed on 92 CU and 92 age- matched NU from a total of 187 7T scans. CU were selected on the basis of their scores on the Semi-Structured Assessment for the Genetics of Alcoholism. The groups were first compared on a decision-making test and then on ICA based functional connectivity between corticocerebellar networks. An MVPA was done as a confirmatory analysis. The anatomy of these networks was then assessed using Diffusion Tensor imaging (DTI) and cortical volume analyses. Cannabis Users had significantly higher scores on the Iowa Gambling task (IGT) [Gambling task Percentage larger] and significantly lower scores on the [Gambling task reward Percentage smaller]. Left accumbens (L NAc) volume was significantly larger in cannabis users. DTI analysis between the groups yielded no significant (FWE corrected) differences. Resting state FC analysis of the left Cerebellum region 9 showed enhanced functional connectivity with the right nucleus accumbens and left pallidum and left putamen in CU. In addition, posterior cerebellum showed enhanced functional connectivity (FWE corrected) with 2 nodes of the DMN and left and right paracingulate (sub genual ACC) and the sub callosal cortex in CU. IGT percentage larger scores correlated with posterior cerebellar functional connectivity in non-user women. A multivariate pattern analysis confirmed this cerebellar hyperconnectivity in both groups. Our results demonstrate for the first time that deficits in DM observed in cannabis users are mirrored in hyper connectivity in corticocerebellar networks. Cortical volumes of some of the nodes of these networks showed increases in users. However, the underlying white matter was largely intact in CU. The observed DM deficits and hyper connectivity in resting networks may contribute to difficulties in quitting and/or facilitating relapse.
Collapse
Affiliation(s)
- Alan N Francis
- Department of Neuroscience, University of Texas Rio Grande Valley, TX, United States.
| | - Joan A Camprodon
- Dept of Psychiatry, Massachusetts General Hospital, Harvard Medical School, United States
| | - Francesca Filbey
- Center for Brain Health, School of Behavioral & Brain Science, University of Texas, Dallas, United States
| |
Collapse
|
38
|
Zanin JP, Pandya MA, Espinoza D, Friedman WJ, Shiflett MW. Excess cerebellar granule neurons induced by the absence of p75NTR during development elicit social behavior deficits in mice. Front Mol Neurosci 2023; 16:1147597. [PMID: 37305555 PMCID: PMC10249730 DOI: 10.3389/fnmol.2023.1147597] [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: 01/18/2023] [Accepted: 04/24/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Recently, the cerebellum has been implicated with non-motor functions, including cognitive and emotional behavior. Anatomical and functional studies demonstrate bidirectional cerebellar connections with brain regions involved in social cognition. Cerebellar developmental abnormalities and injury are often associated with several psychiatric and mental disorders including autism spectrum disorders and anxiety. The cerebellar granule neurons (CGN) are essential for cerebellar function since they provide sensorimotor, proprioceptive, and contextual information to Purkinje cells to modify behavior in different contexts. Therefore, alterations to the CGN population are likely to compromise cerebellar processing and function. Previously we demonstrated that the p75 neurotrophin receptor (p75NTR) was fundamental for the development of the CGN. In the absence of p75NTR, we observed increased proliferation of the granule cell precursors (GCPs), followed by increased GCP migration toward the internal granule layer. The excess granule cells were incorporated into the cerebellar network, inducing alterations in cerebellar circuit processing. Methods In the present study, we used two conditional mouse lines to specifically delete the expression of p75NTR in CGN. In both mouse lines, deletion of the target gene was under the control of the transcription factor Atoh-1 promotor, however, one of the lines was also tamoxifen-inducible. Results We observed a loss of p75NTR expression from the GCPs in all cerebellar lobes. Compared to control animals, both mouse lines exhibited a reduced preference for social interactions when presented with a choice to interact with a mouse or an object. Open-field locomotor behavior and operant reward learning were unaffected in both lines. Lack of preference for social novelty and increased anxiety-related behavior was present in mice with constitutive p75NTR deletion; however, these effects were not present in the tamoxifen-inducible mice with p75NTR deletion that more specifically targeted the GCPs. Discussion Our findings demonstrate that alterations to CGN development by loss of p75NTR alter social behavior, and contribute to the increasing evidence that the cerebellum plays a role in non-motor-related behaviors, including social behavior.
Collapse
Affiliation(s)
- Juan Pablo Zanin
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Mansi A. Pandya
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Diego Espinoza
- Department of Psychology, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Wilma J. Friedman
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Michael W. Shiflett
- Department of Psychology, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
39
|
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
|
40
|
Rodríguez-Borillo O, Roselló-Jiménez L, Guarque-Chabrera J, Palau-Batet M, Gil-Miravet I, Pastor R, Miquel M, Font L. Neural correlates of cocaine-induced conditioned place preference in the posterior cerebellar cortex. Front Behav Neurosci 2023; 17:1174189. [PMID: 37179684 PMCID: PMC10169591 DOI: 10.3389/fnbeh.2023.1174189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/06/2023] [Indexed: 05/15/2023] Open
Abstract
Introduction Addictive drugs are potent neuropharmacological agents capable of inducing long-lasting changes in learning and memory neurocircuitry. With repeated use, contexts and cues associated with consumption can acquire motivational and reinforcing properties of abused drugs, triggering drug craving and relapse. Neuroplasticity underlying drug-induced memories takes place in prefrontal-limbic-striatal networks. Recent evidence suggests that the cerebellum is also involved in the circuitry responsible for drug-induced conditioning. In rodents, preference for cocaine-associated olfactory cues has been shown to correlate with increased activity at the apical part of the granular cell layer in the posterior vermis (lobules VIII and IX). It is important to determine if the cerebellum's role in drug conditioning is a general phenomenon or is limited to a particular sensory modality. Methods The present study evaluated the role of the posterior cerebellum (lobules VIII and IX), together with the medial prefrontal cortex (mPFC), ventral tegmental area (VTA), and nucleus accumbens (NAc) using a cocaine-induced conditioned place preference procedure with tactile cues. Cocaine CPP was tested using ascending (3, 6, 12, and 24 mg/kg) doses of cocaine in mice. Results Compared to control groups (Unpaired and Saline animals), Paired mice were able to show a preference for the cues associated with cocaine. Increased activation (cFos expression) of the posterior cerebellum was found in cocaine CPP groups and showed a positive correlation with CPP levels. Such increases in cFos activity in the posterior cerebellum significantly correlated with cFos expression in the mPFC. Discussion Our data suggest that the dorsal region of the cerebellum could be an important part of the network that mediates cocaine-conditioned behavior.
Collapse
Affiliation(s)
| | | | - Julian Guarque-Chabrera
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Spain
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - María Palau-Batet
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Spain
| | - Isis Gil-Miravet
- Unitat Predepartamental de Medicina, Universitat Jaume I, Castellón de la Plana, Spain
| | - Raúl Pastor
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Spain
| | - Marta Miquel
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Spain
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - Laura Font
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Spain
| |
Collapse
|
41
|
Chen F, Wang X, Jang SK, Quach BC, Weissenkampen JD, Khunsriraksakul C, Yang L, Sauteraud R, Albert CM, Allred NDD, Arnett DK, Ashley-Koch AE, Barnes KC, Barr RG, Becker DM, Bielak LF, Bis JC, Blangero J, Boorgula MP, Chasman DI, Chavan S, Chen YDI, Chuang LM, Correa A, Curran JE, David SP, de las Fuentes L, Deka R, Duggirala R, Faul JD, Garrett ME, Gharib SA, Guo X, Hall ME, Hawley NL, He J, Hobbs BD, Hokanson JE, Hsiung CA, Hwang SJ, Hyde TM, Irvin MR, Jaffe AE, Johnson EO, Kaplan R, Kardia SLR, Kaufman JD, Kelly TN, Kleinman JE, Kooperberg C, Lee IT, Levy D, Lutz SM, Manichaikul AW, Martin LW, Marx O, McGarvey ST, Minster RL, Moll M, Moussa KA, Naseri T, North KE, Oelsner EC, Peralta JM, Peyser PA, Psaty BM, Rafaels N, Raffield LM, Reupena MS, Rich SS, Rotter JI, Schwartz DA, Shadyab AH, Sheu WHH, Sims M, Smith JA, Sun X, Taylor KD, Telen MJ, Watson H, Weeks DE, Weir DR, Yanek LR, Young KA, Young KL, Zhao W, Hancock DB, Jiang B, Vrieze S, Liu DJ. Multi-ancestry transcriptome-wide association analyses yield insights into tobacco use biology and drug repurposing. Nat Genet 2023; 55:291-300. [PMID: 36702996 PMCID: PMC9925385 DOI: 10.1038/s41588-022-01282-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/08/2022] [Indexed: 01/27/2023]
Abstract
Most transcriptome-wide association studies (TWASs) so far focus on European ancestry and lack diversity. To overcome this limitation, we aggregated genome-wide association study (GWAS) summary statistics, whole-genome sequences and expression quantitative trait locus (eQTL) data from diverse ancestries. We developed a new approach, TESLA (multi-ancestry integrative study using an optimal linear combination of association statistics), to integrate an eQTL dataset with a multi-ancestry GWAS. By exploiting shared phenotypic effects between ancestries and accommodating potential effect heterogeneities, TESLA improves power over other TWAS methods. When applied to tobacco use phenotypes, TESLA identified 273 new genes, up to 55% more compared with alternative TWAS methods. These hits and subsequent fine mapping using TESLA point to target genes with biological relevance. In silico drug-repurposing analyses highlight several drugs with known efficacy, including dextromethorphan and galantamine, and new drugs such as muscle relaxants that may be repurposed for treating nicotine addiction.
Collapse
Affiliation(s)
- Fang Chen
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Xingyan Wang
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Seon-Kyeong Jang
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | | | - J Dylan Weissenkampen
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, Penn State College of Medicine, Hershey, PA, USA
| | | | - Lina Yang
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Renan Sauteraud
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Christine M Albert
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Duke Comprehensive Sickle Cell Center, Duke University Medical Center, Durham, NC, USA
| | - Kathleen C Barnes
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - R Graham Barr
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Diane M Becker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Joshua C Bis
- Department of Medicine, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - John Blangero
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Meher Preethi Boorgula
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sameer Chavan
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Yii-Der I Chen
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Lee-Ming Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Adolfo Correa
- Department of Medicine, Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS, USA
| | - Joanne E Curran
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Sean P David
- University of Chicago, Chicago, IL, USA
- NorthShore University Health System, Evanston, IL, USA
| | - Lisa de las Fuentes
- Department of Medicine, Division of Biostatistics and Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Ranjan Deka
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Ravindranath Duggirala
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Jessica D Faul
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI, USA
| | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Sina A Gharib
- Department of Medicine, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Computational Medicine Core at Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Xiuqing Guo
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Michael E Hall
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Nicola L Hawley
- Department of Epidemiology (Chronic Disease), School of Public Health, Yale University, New Haven, CT, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Brian D Hobbs
- Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John E Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Chao A Hsiung
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Jen Hwang
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mental Health and Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Human Genetics and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, The Bronx, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Joel D Kaufman
- Departments of Environmental & Occupational Health Sciences, Medicine, and Epidemiology, University of Washington Seattle, Seattle, WA, USA
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - I-Te Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Daniel Levy
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sharon M Lutz
- Department of Population Medicine, Harvard Pilgrim Health Care, Boston, MA, USA
| | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Lisa W Martin
- Division of Cardiology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Olivia Marx
- Department of Biomedical Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Stephen T McGarvey
- Department of Epidemiology, International Health Institute, Brown University School of Public Health, Providence, RI, USA
| | - Ryan L Minster
- Department of Human Genetics and Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew Moll
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Karine A Moussa
- Penn State Huck Institutes of Life Sciences, Penn State College of Medicine, University Park, PA, USA
| | - Take Naseri
- Ministry of Health, Government of Samoa, Apia, Samoa
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elizabeth C Oelsner
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Juan M Peralta
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Bruce M Psaty
- Department of Medicine, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Nicholas Rafaels
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | | | - Aladdin H Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, USA
| | | | - Mario Sims
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI, USA
| | - Xiao Sun
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Kent D Taylor
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Marilyn J Telen
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Harold Watson
- Faculty of Medical Sciences, University of the West Indies, Cave Hill Campus, Barbados
| | - Daniel E Weeks
- Department of Human Genetics and Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - David R Weir
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI, USA
| | - Lisa R Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kendra A Young
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI, USA
| | | | - Bibo Jiang
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA.
| | - Scott Vrieze
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA.
| | - Dajiang J Liu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
42
|
Szabo E, Timmers I, Borsook D, Simons LE, Sieberg CB. Altered anterior insula functional connectivity in adolescent and young women with endometriosis-associated pain: Pilot resting-state fMRI study. Eur J Paediatr Neurol 2022; 41:80-90. [PMID: 36375399 PMCID: PMC9722632 DOI: 10.1016/j.ejpn.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/12/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Endometriosis is the leading cause of chronic pelvic pain. Alterations in brain functional connectivity have been reported in adult women with endometriosis-associated pain (EAP), however, it is still unknown if similar patterns of changes exist in adolescents. METHODS In this pilot study, resting-state fMRI scans were obtained from 11 adolescent and young women with EAP and 14 healthy female controls. Using a seed-to-voxel approach, we investigated functional connectivity between the anterior insula, medial prefrontal cortex, and the rest of the brain. Furthermore, we explored whether potential functional connectivity differences were correlated with clinical characteristics including disease duration, pain intensity, and different psychosocial factors (pain catastrophizing, fear of pain, functional disability, anxiety, and depression). RESULTS Our findings revealed that patients with EAP demonstrated significantly decreased connectivity between the right anterior insula and two clusters: one in the right cerebellum, and one in the left middle frontal gyrus compared to controls. Additionally, functional connectivity between the right anterior insula and the right cerebellum was positively associated with pain intensity levels. In patients with EAP, brain changes were also correlated with state anxiety and fear of pain. CONCLUSIONS Our results are relevant not only for understanding the brain characteristics underlying EAP at a younger age, but also in enhancing future pain treatment efforts by supporting the involvement of the central nervous system in endometriosis.
Collapse
Affiliation(s)
- Edina Szabo
- Pain and Affective Neuroscience Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, Boston, MA, USA; Biobehavioral Pain Innovations Lab, Department of Psychiatry and Behavioral Sciences, Boston Children's Hospital, Boston, MA, USA; Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Inge Timmers
- Department of Rehabilitation Medicine, Maastricht University, Maastricht, the Netherlands; Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - David Borsook
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA; Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Department of Anesthesiology, Harvard Medical School, Boston, MA, USA
| | - Laura E Simons
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Christine B Sieberg
- Pain and Affective Neuroscience Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, Boston, MA, USA; Biobehavioral Pain Innovations Lab, Department of Psychiatry and Behavioral Sciences, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Lin X, Zhu X, Zhou W, Zhang Z, Li P, Dong G, Meng S, Deng J, Lu L. Connectome-based predictive modelling of smoking severity in smokers. Addict Biol 2022; 27:e13242. [PMID: 36301219 DOI: 10.1111/adb.13242] [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: 06/06/2022] [Revised: 08/11/2022] [Accepted: 09/27/2022] [Indexed: 01/24/2023]
Abstract
The functional connectivity within and between networks could provide a framework to characterize the neurobiological mechanism of nicotine addiction. This study examined the brain regions that were functionally connected in response to smoking cues and established the brain-behaviour relationships in smokers. Sixty-seven male smokers were enrolled and scanned while performing the cue-reactivity and Stroop task. A whole-brain analysis approach, connectome-based predictive modelling (CPM), was conducted on the data from the cue-reactivity task to identify the networks that could predict the smoking severity with the Shen atlas as templates. Then, the brain-behaviour relationships were verified in a different brain state (Stroop task). CPM identified the smoking severity-related network, as indicated by a significant correlation between predicted and actual smoking severity scores (r = 0.31, p = 0.02). Identified networks mainly involved the canonical networks implicated in the reward process (motor/sensory network and salience network) and executive control (frontoparietal network). Network strength in the Stroop task marginally significantly predicted smoking severity scores (r = 0.23, p = 0.06), partially replicating the brain-behaviour relationship. The CPM results identified the whole-brain neural network related to smoking severity, which was cross-validated by the AAL and Shen atlas. These findings contribute to more profound insights into neural substrates underlying the smoking severity.
Collapse
Affiliation(s)
- Xiao Lin
- 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
| | - Ximei Zhu
- 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
| | - Weiran Zhou
- Centre for Cognition and Brain Disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Zhibo Zhang
- 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
| | - Guangheng Dong
- Centre for Cognition and Brain Disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 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
| | - Lin Lu
- 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.,National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| |
Collapse
|
44
|
Sepe-Forrest L, Kim DJ, Quinn PD, Bolbecker AR, Wisner KM, Hetrick WP, O'Donnell BF. Evidence of familial confounding of the association between cannabis use and cerebellar-cortical functional connectivity using a twin study. Neuroimage Clin 2022; 36:103237. [PMID: 36451348 PMCID: PMC9668648 DOI: 10.1016/j.nicl.2022.103237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/26/2022] [Accepted: 10/16/2022] [Indexed: 11/11/2022]
Abstract
Cerebellar-cortical resting-state functional connectivity (rsFC) has been reported to be altered in cannabis users. However, this association may be due to genetic and environmental confounding rather than a causal relationship between cannabis use and changes in rsFC. In this co-twin control study, linear mixed models were used to assess relationships between the number of lifetime cannabis uses (NLCU) and age of cannabis onset (ACO) with cerebellar-cortical rsFC. The rsFC with seven functional networks was evaluated in 147 monozygotic and 82 dizygotic twin pairs. Importantly, the use of genetically informed models in this twin sample facilitated examining whether shared genetic or environmental effects underlie crude associations between cannabis measures and connectivity. Individual-level phenotypic analyses (i.e., accounting for twin-pair non-independence) showed that individuals in the full sample with earlier ACO and higher NLCU had lower cerebellar rsFC within the VA, DA, and FP networks. Yet, there were no significant differences in cerebellar-cortical rsFC between monozygotic twins who were discordant for cannabis measures. These findings suggest shared genetic or environmental confounds contribute to associations between cannabis use and altered cerebellar-cortical rsFC, rather than unique causal impacts of cannabis use on cerebellar-cortical rsFC.
Collapse
Affiliation(s)
- Linnea Sepe-Forrest
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, United States,Program in Neuroscience, Indiana University, Bloomington, IN, United States,Corresponding author-at: Indiana University Bloomington, Department of Psychology, Room A208A, United States.
| | - Dae-Jin Kim
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Patrick D. Quinn
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, United States,Program in Neuroscience, Indiana University, Bloomington, IN, United States,Department of Applied Health Science, School of Public Health, Indiana University, Bloomington, IN, United States
| | - Amanda R. Bolbecker
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Krista M. Wisner
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, United States,Program in Neuroscience, Indiana University, Bloomington, IN, United States
| | - William P. Hetrick
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, United States,Program in Neuroscience, Indiana University, Bloomington, IN, United States,Department of Psychiatry, Indiana University, Indianapolis, IN, United States
| | - Brian F. O'Donnell
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, United States,Program in Neuroscience, Indiana University, Bloomington, IN, United States,Department of Psychiatry, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
45
|
Baselmans B, Hammerschlag AR, Noordijk S, Ip H, van der Zee M, de Geus E, Abdellaoui A, Treur JL, van ’t Ent D. The Genetic and Neural Substrates of Externalizing Behavior. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 2:389-399. [PMID: 36324656 PMCID: PMC9616240 DOI: 10.1016/j.bpsgos.2021.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
Background To gain more insight into the biological factors that mediate vulnerability to display externalizing behaviors, we leveraged genome-wide association study summary statistics on 13 externalizing phenotypes. Methods After data classification based on genetic resemblance, we performed multivariate genome-wide association meta-analyses and conducted extensive bioinformatic analyses, including genetic correlation assessment with other traits, Mendelian randomization, and gene set and gene expression analyses. Results The genetic data could be categorized into disruptive behavior (DB) and risk-taking behavior (RTB) factors, and subsequent genome-wide association meta-analyses provided association statistics for DB and RTB (N eff = 523,150 and 1,506,537, respectively), yielding 50 and 257 independent genetic signals. The statistics of DB, much more than RTB, signaled genetic predisposition to adverse cognitive, mental health, and personality outcomes. We found evidence for bidirectional causal influences between DB and substance use behaviors. Gene set analyses implicated contributions of neuronal cell development (DB/RTB) and synapse formation and transcription (RTB) mechanisms. Gene-brain mapping confirmed involvement of the amygdala and hypothalamus and highlighted other candidate regions (cerebellar dentate, cuneiform nucleus, claustrum, paracentral cortex). At the cell-type level, we noted enrichment of glutamatergic neurons for DB and RTB. Conclusions This bottom-up, data-driven study provides new insights into the genetic signals of externalizing behaviors and indicates that commonalities in genetic architecture contribute to the frequent co-occurrence of different DBs and different RTBs, respectively. Bioinformatic analyses supported the DB versus RTB categorization and indicated relevant biological mechanisms. Generally similar gene-brain mappings indicate that neuroanatomical differences, if any, escaped the resolution of our methods.
Collapse
Affiliation(s)
- Bart Baselmans
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Anke R. Hammerschlag
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Stephany Noordijk
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Hill Ip
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Matthijs van der Zee
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Eco de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Abdel Abdellaoui
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jorien L. Treur
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Dennis van ’t Ent
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
- Address correspondence to Dennis van ’t Ent, Ph.D.
| |
Collapse
|
46
|
Jackson TB, Bernard JA. Cerebellar and basal ganglia motor network predicts trait depression and hyperactivity. Front Behav Neurosci 2022; 16:953303. [PMID: 36187378 PMCID: PMC9523104 DOI: 10.3389/fnbeh.2022.953303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
In the human brain, the cerebellum (CB) and basal ganglia (BG) are implicated in cognition-, emotion-, and motor-related cortical processes and are highly interconnected, both to cortical regions via separate, trans-thalamic pathways and to each other via subcortical disynaptic pathways. We previously demonstrated a distinction between cognitive and motor CB-BG networks (CCBN, MCBN, respectively) as it relates to cortical network integration in healthy young adults, suggesting the subcortical networks separately support cortical networks. The CB and BG are also implicated in the pathophysiology of schizophrenia, Parkinson's, and compulsive behavior; thus, integration within subcortical CB-BG networks may be related to transdiagnostic symptomology. Here, we asked whether CCBN or MCBN integration predicted Achenbach Self-Report scores for anxiety, depression, intrusive thoughts, hyperactivity and inactivity, and cognitive performance in a community sample of young adults. We computed global efficiency for each CB-BG network and 7 canonical resting-state networks for all right-handed participants in the Human Connectome Project 1200 release with a complete set of preprocessed resting-state functional MRI data (N = 783). We used multivariate regression to control for substance abuse and age, and permutation testing with exchangeability blocks to control for family relationships. MCBN integration negatively predicted depression and hyperactivity, and positively predicted cortical network integration. CCBN integration predicted cortical network integration (except for the emotional network) and marginally predicted a positive relationship with hyperactivity, indicating a potential dichotomy between cognitive and motor CB-BG networks and hyperactivity. These results highlight the importance of CB-BG interactions as they relate to motivation and symptoms of depression.
Collapse
Affiliation(s)
- T. Bryan Jackson
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
- *Correspondence: T. Bryan Jackson
| | - Jessica A. Bernard
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
47
|
Moussa-Tooks AB, Rogers BP, Huang AS, Sheffield JM, Heckers S, Woodward ND. Cerebellar Structure and Cognitive Ability in Psychosis. Biol Psychiatry 2022; 92:385-395. [PMID: 35680432 PMCID: PMC9378489 DOI: 10.1016/j.biopsych.2022.03.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Dysconnectivity theories, combined with advances in fundamental cognitive neuroscience, have led to increased interest in characterizing cerebellar abnormalities in psychosis. Smaller cerebellar gray matter volume has been found in schizophrenia spectrum disorders. However, the course of these deficits across illness stage, specificity to schizophrenia (vs. psychosis more broadly), and relationship to clinical phenotypes, primarily cognitive impairment, remain unclear. METHODS The Spatially Unbiased Infratentorial toolbox, a gold standard for analyzing human neuroimaging data of the cerebellum, was used to quantify cerebellar volumes and conduct voxel-based morphometry on structural magnetic resonance images obtained from 574 individuals (249 schizophrenia spectrum, 108 bipolar with psychotic features, 217 nonpsychiatric control). Analyses examining diagnosis (schizophrenia spectrum, bipolar disorder), illness stage (early, chronic), and cognitive effects on cerebellum structure in psychosis were performed. RESULTS Cerebellar structure in psychosis did not differ significantly from healthy participants, regardless of diagnosis and illness stage (effect size = 0.01-0.14). In contrast, low premorbid cognitive functioning was associated with smaller whole and regional cerebellum volumes, including cognitive (lobules VI and VII, Crus I, frontoparietal and attention networks) and motor (lobules I-IV, V, and X; somatomotor network) regions in psychosis (effect size = 0.36-0.60). These effects were not present in psychosis cohorts with average estimated premorbid cognition. CONCLUSIONS Cerebellar structural abnormalities in psychosis are related to lower premorbid cognitive functioning implicating early antecedents, atypical neurodevelopment, or both in cerebellar dysfunction. Future research focused on identifying the impact of early-life risk factors for psychosis on the development of the cerebellum and cognition is warranted.
Collapse
Affiliation(s)
- Alexandra B Moussa-Tooks
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.
| | - Baxter P Rogers
- Vanderbilt University Institute of Imaging Science, Nashville, Tennessee
| | - Anna S Huang
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Julia M Sheffield
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Stephan Heckers
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Neil D Woodward
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
48
|
Xu X, Pu J, Shaw A, Jackson T. Neural responsiveness to Chinese versus Western food images: An functional magnetic resonance imaging study of Chinese young adults. Front Nutr 2022; 9:948039. [PMID: 36034899 PMCID: PMC9411937 DOI: 10.3389/fnut.2022.948039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
Cross-cultural studies suggest that people typically prefer to eat familiar foods from their own culture rather than foreign foods from other cultures. On this basis, it is plausible that neural responsiveness elicited by palatable food images from one’s own culture differ from those elicited by food depictions from other cultures. Toward clarifying this issue, we examined neural activation and self-report responses to indigenous (Chinese) versus Western food images among young Chinese adults. Participants (33 women, 33 men) viewed Chinese food, Western food and furniture control images during a functional magnetic resonance imaging (fMRI) scan and then rated the images on “liking,” “wanting,” and “difficult resisting.” Analyses indicated there were no significant differences in self-report ratings of Chinese versus Western food images. However, Chinese food images elicited stronger activation in regions linked to cravings, taste perception, attention, reward, and visual processing (i.e., cerebellum crus, superior temporal gyrus, supramarginal gyrus, middle temporal gyrus, inferior parietal lobule, posterior insula, middle occipital gyrus; inferior occipital gyrus). Conversely, Western food images elicited stronger activation in areas involved in visual object recognition and visual processing (inferior temporal gyrus, middle occipital gyrus, calcarine). These findings underscored culture as a potentially important influence on neural responses to visual food cues and raised concerns about the ecological validity of using “standard” Western food images in neuroimaging studies of non-Western samples. Results also provide foundations for designing culturally informed research and intervention approaches in non-Westerns contexts guided by the use of external food cues that are most salient to the cultural group under study.
Collapse
Affiliation(s)
- Xi Xu
- School of Psychology, Southwest University, Chongqing, China
| | - Jiajia Pu
- School of Psychology, Southwest University, Chongqing, China
| | - Amy Shaw
- Department of Psychology, University of Macau, Macao, Macao SAR, China
| | - Todd Jackson
- Department of Psychology, University of Macau, Macao, Macao SAR, China
| |
Collapse
|
49
|
Guarque-Chabrera J, Sanchez-Hernandez A, Ibáñez-Marín P, Melchor-Eixea I, Miquel M. Role of Perineuronal nets in the cerebellar cortex in cocaine-induced conditioned preference, extinction, and reinstatement. Neuropharmacology 2022; 218:109210. [PMID: 35985392 DOI: 10.1016/j.neuropharm.2022.109210] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 10/31/2022]
Abstract
Perineuronal nets (PNNs) are cartilage-like structures of extracellular matrix molecules that enwrap in a net-like manner the cell-body and proximal dendrites of special subsets of neurons. PNNs stabilize their incoming connections and restrict plasticity. Consequently, they have been proposed as a candidate mechanism for drug-induced learning and memory. In the cerebellum, PNNs surround Golgi inhibitory interneurons and both inhibitory and excitatory neurons in the deep cerebellar nuclei (DCN). Previous studies from the lab showed that cocaine-induced conditioned memory increased PNN expression in the granule cell layer of the posterior vermis. The present research aimed to investigate the role of cerebellar PNNs in cocaine-induced conditioned preference. For this purpose, we use the enzyme chondroitinase ABC (ChABC) to digest PNNs at different time points of the learning process to ascertain whether their removal can affect drug-induced memory. Our results show that PNN digestion using ChABC in the posterior vermis (Lobule VIII) did not affect the acquisition of cocaine-induced conditioned preference. However, the removal of PNNs in Lobule VIII -but not in the DCN- disrupted short-term memory of conditioned preference. Moreover, although PNN digestion facilitated the formation of extinction, reinstatement of cocaine-induced conditioned preference was encouraged under PNN digestion. The present findings suggests that PNNs around Golgi interneurons are needed to maintain cocaine-induced Pavlovian memory but also to stabilize extinction memory. Conversely, PNN degradation within the DCN did not affect stability of cocaine-induced memories. Therefore, degradation of PNNs in the vermis might be used as a promising tool to manipulate drug-induced memory.
Collapse
Affiliation(s)
- Julian Guarque-Chabrera
- Área de Psicobiología, Universitat Jaume I, Facultat de Ciencies de la Salut, Avenida Vicente Sos Baynat sn, 12071, Castellón de la Plana, Spain
| | - Aitor Sanchez-Hernandez
- Área de Psicobiología, Universitat Jaume I, Facultat de Ciencies de la Salut, Avenida Vicente Sos Baynat sn, 12071, Castellón de la Plana, Spain
| | - Patricia Ibáñez-Marín
- Área de Psicobiología, Universitat Jaume I, Facultat de Ciencies de la Salut, Avenida Vicente Sos Baynat sn, 12071, Castellón de la Plana, Spain
| | - Ignasi Melchor-Eixea
- Área de Psicobiología, Universitat Jaume I, Facultat de Ciencies de la Salut, Avenida Vicente Sos Baynat sn, 12071, Castellón de la Plana, Spain
| | - Marta Miquel
- Área de Psicobiología, Universitat Jaume I, Facultat de Ciencies de la Salut, Avenida Vicente Sos Baynat sn, 12071, Castellón de la Plana, Spain.
| |
Collapse
|
50
|
F Amil A, Rubio Ballester B, Maier M, F M J Verschure P. Chronic use of cannabis might impair sensory error processing in the cerebellum through endocannabinoid dysregulation. Addict Behav 2022; 131:107297. [PMID: 35417840 DOI: 10.1016/j.addbeh.2022.107297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 09/08/2021] [Accepted: 02/25/2022] [Indexed: 11/20/2022]
Abstract
Chronic use of cannabis leads to both motor deficits and the downregulation of CB1 receptors (CB1R) in the cerebellum. In turn, cerebellar damage is often related to impairments in motor learning and control. Further, a recent motor learning task that measures cerebellar-dependent adaptation has been shown to distinguish well between healthy subjects and chronic cannabis users. Thus, the deteriorating effects of chronic cannabis use in motor performance point to cerebellar adaptation as a key process to explain such deficits. We review the literature relating chronic cannabis use, the endocannabinoid system in the cerebellum, and different forms of cerebellar-dependent motor learning, to suggest that CB1R downregulation leads to a generalized underestimation and misprocessing of the sensory errors driving synaptic updates in the cerebellar cortex. Further, we test our hypothesis with a computational model performing a motor adaptation task and reproduce the behavioral effect of decreased implicit adaptation that appears to be a sign of chronic cannabis use. Finally, we discuss the potential of our hypothesis to explain similar phenomena related to motor impairments following chronic alcohol dependency.
Collapse
Affiliation(s)
- Adrián F Amil
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| | | | - Martina Maier
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.
| | - Paul F M J Verschure
- Donders Center for Neuroscience (DCN) - Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|