1
|
Hong N, Yoon SR, Ahn JC. Photobiomodulation using an 830-nm laser alleviates hippocampal reactive gliosis and cognitive dysfunction in a mouse model of adolescent chronic alcohol exposure. Pharmacol Biochem Behav 2025; 248:173956. [PMID: 39793712 DOI: 10.1016/j.pbb.2025.173956] [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: 12/18/2023] [Revised: 08/14/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
Chronic alcoholism is known to have detrimental effects on the brain, including cognitive impairment, neurotransmitter imbalances, and brain atrophy. The hippocampus, crucial for spatial memory and cognitive functions, is particularly susceptible to alcohol-induced changes. Photobiomodulation (PBM), a non-invasive therapeutic method that utilizes red or near-infrared light, has shown promising applications in the central and peripheral nervous systems. Near-infrared (NIR) light, in particular, has been shown to prevent apoptosis, and neuroinflammation, as well as to improve cognitive functions. In this study, we aimed to investigate whether 830-nm laser irradiation could mitigate cognitive deficits in a chronic alcohol mouse model. Chronic alcoholism was induced in C57BL/6 mice through continuous ethanol gavage for 4 weeks at a dosage of 5 g/kg/day. Gavaging was performed 3 times per week for 4 weeks. Mice were transcranial irradiated by 830-nm laser, following making a chronic alcohol mouse model. Laser irradiation (50 mW/cm2) was performed 5 times per week for 3 weeks. To verify memory and cognitive defeats of a chronic alcohol mouse model, we performed animal behavior tasks such as Morris water maze, Y maze, and novel objective recognition. Our results confirmed the cognitive impairment in the chronic alcohol mouse model compared to the control group in conducted tasks. However, cognitive and spatial memory significantly improved following 830-nm laser irradiation. Additionally, we confirmed whether the behavior tasks result from histological changes. We performed immunofluorescence staining in the hippocampus region (CA3, CA1 and hilus) using astrocyte (GFAP) and microglia (Iba1) markers. As a result, reactive astrocyte was significantly increased in the chronic alcohol mouse model compared to control mice, whereas the number of GFAP-positive cells was significantly reduced by 830-nm laser irradiation. These findings indicate that chronic alcohol exposure induces spatial memory and cognitive impairment, which can be effectively rescued through near-infrared laser irradiation.
Collapse
Affiliation(s)
- Namgue Hong
- Medical Laser Research Center, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Sung-Ryeong Yoon
- Department of Medical Science, Graduate School of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Jin-Chul Ahn
- Medical Laser Research Center, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea; Department of Biomedical Science, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea.
| |
Collapse
|
2
|
Quinn P. Art therapy's engagement of brain networks for enduring recovery from addiction. Front Psychiatry 2025; 15:1458063. [PMID: 39834572 PMCID: PMC11743619 DOI: 10.3389/fpsyt.2024.1458063] [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: 07/01/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
The field of addiction in its priority to save lives has emphasized harm reduction and medication therapies that have taken precedence over counseling and psychotherapy. The extensive mental health needs, traumatic histories and cognitive challenges of this population call for more availability of all treatments, but also in-depth treatment for the causes of the addiction. The prevalence of trauma is examined with regard to the challenge it presents in treatment for substance use disorder (SUD), and other comorbidities. Two case examples are offered that exemplify how art therapy expedites key information about underlying trauma. Art therapy is proposed as a treatment approach for SUD for its apparent activation of key neural networks that are also impacted by trauma, and its usefulness in engaging those who have cognitive challenges experientially. Quantitative research is cited that suggests art therapy's activation of the reward system, which may make art therapy useful in treating the stress and inhibition coefficients of addiction that map to neural networks of addiction. The need for additional empirical research is cited that may improve the efficiency and effectiveness of art therapy and mental health treatment.
Collapse
Affiliation(s)
- Patricia Quinn
- School of Fine Arts – Graduate Program in Art Therapy, Maharashtra Institute of Technology, Pune, India
| |
Collapse
|
3
|
Prignitz M, Guldner S, Lehmler SJ, Aggensteiner PM, Nees F. An Ecological Momentary Assessment Approach of Environmental Triggers in the Role of Daily Affect, Rumination, and Movement Patterns in Early Alcohol Use Among Healthy Adolescents: Exploratory Study. JMIR Mhealth Uhealth 2024; 12:e53401. [PMID: 39657181 DOI: 10.2196/53401] [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: 10/05/2023] [Revised: 05/07/2024] [Accepted: 08/08/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Adolescence is a period characterized by an increased susceptibility to developing risky alcohol consumption habits. This susceptibility can be influenced by social and situational factors encountered in daily life, which, in conjunction with emotions and thoughts, contribute to behavioral patterns related to alcohol use even in the early stages of alcohol experimentation, when initial experiences with alcohol are formed, and regular consumption is still evolving. OBJECTIVE This study aimed to examine the association between detailed behavioral and movement patterns, along with emotional and cognitive factors, and the early onset of alcohol use in the everyday lives of adolescents. METHODS A total of 65 healthy adolescents (33 male, twenty-nine 14-year-olds, and thirty-six 16-year-olds) underwent mobile-based ecological momentary assessments on alcohol (once a day at 9 AM, assessing alcohol use the day before), positive and negative affect, craving, rumination, and social context (6 prompts/day at 9 AM, 11 AM, 2 PM, 4 PM, 6 PM and 8 PM), type of day (weekdays or weekends, with weekend including Fridays, Saturdays, and Sundays), and using geospatial measures (specifically roaming entropy and number and type of trigger points for alcohol use met) over 14 days. After adjusting for a compliance rate of at least 50%, 52 participants (26 male and twenty-four 14-year-olds) were included in the analyses. RESULTS Generalized linear multilevel models revealed that higher positive affect (b=0.685, P=.007), higher rumination (b=0.586, P=.02), and a larger movement radius (roaming entropy) (b=8.126, P=.02) were positively associated with alcohol use on the same day. However, social context (b=-0.076, P=.90), negative affect (b=-0.077, P=.80), or potential trigger points (all P>.05) did not show significant associations. Alcohol use varied depending on the type of day, with more alcohol use on weekends (b=1.082, P<.001) and age (t50=-2.910, P=.005), with 16-year-olds (mean 1.61, SD 1.66) reporting more days of alcohol consumption than 14-year-olds (mean 0.548, SD 0.72). CONCLUSIONS Our findings support previously identified factors as significant contributors to very early and low levels of alcohol consumption through fine-grained analysis of daily behaviors. These factors include positive affect, rumination, weekend days, and age. In addition, we emphasize that exploratory environmental movement behavior (roaming entropy) is also significantly associated with adolescent alcohol use, highlighting its importance as an additional factor.
Collapse
Affiliation(s)
- Maren Prignitz
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stella Guldner
- Institute of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stephan Johann Lehmler
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig Holstein, Kiel University, Kiel, Germany
| | - Pascal-M Aggensteiner
- Institute of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Frauke Nees
- Institute of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig Holstein, Kiel University, Kiel, Germany
| |
Collapse
|
4
|
Cooke ME, Stephenson M, Brislin SJ, Latvala A, Barr PB, Piirtola M, Vuoksimaa E, Rose RJ, Kaprio J, Dick DM, Salvatore JE. Association between adolescent alcohol use and cognitive function in young adulthood: A co-twin comparison study. Addiction 2024; 119:1947-1955. [PMID: 39108000 PMCID: PMC11524136 DOI: 10.1111/add.16629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/28/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND AND AIMS Studies on adolescent alcohol use and cognition are often unable to separate the potential causal effects of alcohol use on cognition from shared etiological influences, including genetic influences or other substance use comorbidities also known to be associated with cognition, such as nicotine use. The present study aimed to fill this gap and clarify the relationship between adolescent alcohol use and young adult cognition by accounting for both measured and unmeasured confounders. DESIGN A random effects model accounting for nesting in families was used to control for measured confounders. Next, co-twin comparisons were conducted within the full sample and in monozygotic twin pairs (MZ) to control for unmeasured genetic and environmental confounders shared by co-twins. PARTICIPANTS/SETTING Participants were 812 individuals (58.6% female, 361 complete pairs, 146 MZ pairs) from the longitudinal FinnTwin12 study in Finland. MEASUREMENTS Adolescent alcohol use was indexed with measures of frequency of use and intoxication averaged across ages 14 and 17. Cognitive outcomes were measured at average age 22 and included Trail Making Test, California Stroop test, Wechsler Adult Intelligence subtests (Vocabulary, Block Design, Digit Symbol), Digit Span subtest of Wechsler Memory Scale, Mental Rotation Test and Object Location Memory test. Covariates included sex, parental education, general cognitive ability, current alcohol use and nicotine use. FINDINGS Greater frequency of alcohol use and frequency of intoxication across adolescence was associated with decreased vocabulary scores in the co-twin control [freq: stnd beta = -0.12, 95% confidence interval (CI) = -0.234, -0.013] and MZ only co-twin control models (freq: stnd beta = -0.305, 95% CI = -0.523, -0.087; intox: stnd beta = -0.301, 95% CI = -0.528, -0.074). CONCLUSIONS In Finland, there appears to be little evidence that adolescent alcohol use causes cognitive deficits in young adulthood, except modest evidence for association of higher adolescent alcohol use with lower young adult vocabulary scores.
Collapse
Affiliation(s)
- Megan E. Cooke
- Department of Psychiatry, Robert Wood Johnson Medical SchoolRutgers UniversityPiscatawayNJUSA
| | - Mallory Stephenson
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVAUSA
| | - Sarah J. Brislin
- Department of Psychiatry, Robert Wood Johnson Medical SchoolRutgers UniversityPiscatawayNJUSA
| | - Antti Latvala
- Institute of Criminology and Legal PolicyUniversity of HelsinkiHelsinkiFinland
| | - Peter B. Barr
- Department of Psychiatry and Behavioral SciencesSUNY Downstate Health Sciences UniversityBrooklynNYUSA
| | - Maarit Piirtola
- Institute for Molecular Medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
- UKK Institute for Health Promotion ResearchTampereFinland
| | - Eero Vuoksimaa
- Institute for Molecular Medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
| | - Richard J. Rose
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonINUSA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
| | - Danielle M. Dick
- Department of Psychiatry, Robert Wood Johnson Medical SchoolRutgers UniversityPiscatawayNJUSA
| | - Jessica E. Salvatore
- Department of Psychiatry, Robert Wood Johnson Medical SchoolRutgers UniversityPiscatawayNJUSA
| |
Collapse
|
5
|
Yoon G, Sofuoglu M, Petrakis IL, Pittman B, Bell MD. The combination of donepezil and cognitive training for improving treatment outcomes for alcohol use disorder: Design of a randomized controlled trial. Contemp Clin Trials 2024; 145:107657. [PMID: 39111388 PMCID: PMC11423257 DOI: 10.1016/j.cct.2024.107657] [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: 11/10/2023] [Revised: 07/13/2024] [Accepted: 08/03/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND The development of alcohol use disorder (AUD) is a major concern in public health, and cognitive impairments caused by alcohol are involved in this process. Emerging neurobiological evidence suggests that donepezil, an anticholinesterase agent, may improve AUD treatment outcomes by enhancing neurocognitive functioning. Previous research has also suggested that cognitive remediation therapy (CRT) could potentially improve cognitive function and AUD treatment outcomes. We present the rationale and design of a trial to evaluate the combination of donepezil and cognitive remediation therapy (donepezil + CRT) as an intervention for AUD. METHODS We propose a 13-week, randomized, double-blind, placebo-controlled, between-subjects trial comparing 4 groups (donepezil + CRT vs. donepezil alone vs. CRT alone vs. placebos) as an intervention for AUD. The main goal of the study is to evaluate if donepezil + CRT is superior to placebo in reducing heavy drinking days and improving neurocognitive functioning. A total of 160 patients (4 groups, 40 per each group) with AUD between the ages of 18-80 years will be recruited at Yale University and the VA Connecticut Healthcare System. Primary outcome measures include 1) heavy drinking by Timeline Follow Back (TLFB) over 13 weeks and 2) global neurocognitive functioning by a global index of neurocognitive function score at 7 and 13 weeks. DISCUSSION This protocol paper describes the rationale and proposed methods for the randomized controlled trial for improving AUD treatment outcomes. This project has significant clinical potential to help patients suffering from AUD by improving their cognition and reducing alcohol consumption. TRIAL REGISTRATION NCT05042102.
Collapse
Affiliation(s)
- Gihyun Yoon
- VA Connecticut Healthcare System, West Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
| | - Mehmet Sofuoglu
- VA Connecticut Healthcare System, West Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Ismene L Petrakis
- VA Connecticut Healthcare System, West Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Brian Pittman
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Morris D Bell
- VA Connecticut Healthcare System, West Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
6
|
Lorkiewicz SA, Müller-Oehring EM, Baker FC, Elkins BV, Schulte T. A longitudinal study of the relationship between alcohol-related blackouts and attenuated structural brain development. Dev Cogn Neurosci 2024; 69:101448. [PMID: 39307082 PMCID: PMC11440320 DOI: 10.1016/j.dcn.2024.101448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
PURPOSE Alcohol-related blackouts (ARBs) are common in adolescents and emerging adults. ARBs may also be indicative of persistent, alcohol-related neurocognitive changes. This study explored ARBs as a predictor of altered structural brain development and associated cognitive correlates. METHODS Longitudinal growth curve modeling estimated trajectories of brain volume across 6 years in participants from the National Consortium on Alcohol and Neurodevelopment in Adolescence (NCANDA) study (n = 800, 213 with lifetime ARB history). While controlling for demographics and overall alcohol use, ARB history was analyzed as a predictor of brain volume growth in regions associated with alcohol-related cognitive change. Post hoc analyses examined whether ARBs moderated relationships between brain morphology and cognition. RESULTS ARBs significantly predicted attenuated development of fusiform gyrus and hippocampal volume at unique timepoints compared to overall alcohol use. Alcohol use without ARBs significantly predicted attenuated fusiform and hippocampal growth at earlier and later timepoints, respectively. Despite altered development in regions associated with memory, ARBs did not significantly moderate relationships between brain volume and cognitive performance. CONCLUSION ARBs and overall alcohol use predicted altered brain development in the fusiform gyrus and hippocampus at different timepoints, suggesting ARBs represent a unique marker of neurocognitive risk in younger drinkers.
Collapse
Affiliation(s)
- Sara A Lorkiewicz
- Palo Alto University, Clinical Psychology, Palo Alto, CA, USA; Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Eva M Müller-Oehring
- SRI International, Neuroscience Program, Menlo Park, CA, USA; Stanford University School of Medicine, Psychiatry and Behavioral Sciences, Stanford, CA, USA; Stanford University School of Medicine, Department of Neurology and Neurological Sciences, Stanford, CA, USA
| | - Fiona C Baker
- SRI International, Neuroscience Program, Menlo Park, CA, USA; Brain Function Research Group, School of Physiology, University of Witwatersrand, Johannesburg, South Africa
| | - Brionne V Elkins
- University of Texas Medical Branch, Department of Neurology, Galveston, TX, USA
| | - Tilman Schulte
- Palo Alto University, Clinical Psychology, Palo Alto, CA, USA; SRI International, Neuroscience Program, Menlo Park, CA, USA.
| |
Collapse
|
7
|
Boer OD, Franken IHA, Muetzel RL, Cousijn J, El Marroun H. Examining associations between brain morphology in late childhood and early alcohol or tobacco use initiation in adolescence: Findings from a large prospective cohort. Biol Psychol 2024; 192:108859. [PMID: 39233273 DOI: 10.1016/j.biopsycho.2024.108859] [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: 04/17/2024] [Revised: 07/17/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
A prominent challenge in understanding neural consequences of substance use involves disentangling predispositional risk factors from resulting consequences of substance use. Existing literature has identified pre-existing brain variations as vulnerability markers for substance use throughout adolescence. As early initiation of use is an important predictor for later substance use problems, we examined whether pre-existing brain variations are associated with early initiation of use. In the Generation R Study, a prospective population-based cohort, brain morphology (gray matter volume, cortical thickness and surface area) was assessed at ages 10 and 14 using neuroimaging. In the second wave, participants reported on alcohol and tobacco use initiation. From a base study population (N = 3019), we examined the longitudinal (N = 2218) and cross-sectional (N = 1817) association between brain morphology of frontolimbic regions of interest known to be associated with substance use risk, and very early (age < 13) alcohol/tobacco use initiation. Additionally, longitudinal and cross-sectional associations were examined with a brain surface-based approach. Models were adjusted for age at neuroimaging, sex and relevant sociodemographic factors. No associations were found between brain morphology (ages 10 and 14) and early alcohol/tobacco use initiation (<13 years). Sex-specific analyses suggested a cross-sectional association between smaller brain volume and early initiated tobacco use in girls. Our findings are important for interpreting studies examining neural consequences of substance use in the general population. Future longitudinal studies are needed to specify whether these findings can be extended to initiation and continuation of alcohol/tobacco use in later stages of adolescence.
Collapse
Affiliation(s)
- Olga D Boer
- Center for Substance Use and Addiction Research (CESAR), Department of Psychology, Education and Child Studies, Erasmus School of Social and Behavioral Science, Erasmus University Rotterdam, 3000 DR Rotterdam, the Netherlands; Department of Child and Adolescent Psychiatry, University Medical Center Rotterdam, Erasmus MC, Sophia Children's Hospital, 3000 CB Rotterdam, the Netherlands
| | - Ingmar H A Franken
- Center for Substance Use and Addiction Research (CESAR), Department of Psychology, Education and Child Studies, Erasmus School of Social and Behavioral Science, Erasmus University Rotterdam, 3000 DR Rotterdam, the Netherlands
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry, University Medical Center Rotterdam, Erasmus MC, Sophia Children's Hospital, 3000 CB Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, the Netherlands
| | - Janna Cousijn
- Center for Substance Use and Addiction Research (CESAR), Department of Psychology, Education and Child Studies, Erasmus School of Social and Behavioral Science, Erasmus University Rotterdam, 3000 DR Rotterdam, the Netherlands
| | - Hanan El Marroun
- Center for Substance Use and Addiction Research (CESAR), Department of Psychology, Education and Child Studies, Erasmus School of Social and Behavioral Science, Erasmus University Rotterdam, 3000 DR Rotterdam, the Netherlands; Department of Child and Adolescent Psychiatry, University Medical Center Rotterdam, Erasmus MC, Sophia Children's Hospital, 3000 CB Rotterdam, the Netherlands.
| |
Collapse
|
8
|
Ferdous N, Zúñiga ML, Courtney KE. Neurocognitive Markers during Maximum Alcohol Use in Late Adolescence as Predictors of Change in Later Drinking Behaviors. MENTAL HEALTH SCIENCE 2024; 2:e63. [PMID: 39866645 PMCID: PMC11759326 DOI: 10.1002/mhs2.63] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 04/14/2024] [Indexed: 01/28/2025]
Abstract
Background The influence of alcohol use on later neurocognitive functioning is well researched, yet few studies have investigated whether neurocognition post-drinking initiation in adolescence predicts changes in later alcohol use. Objective Investigate neurocognitive task performance during maximum alcohol use in late adolescence as predictors of drinking behaviors 3-7 years later. Methods Analyses (n=105) were conducted on a longitudinal dataset involving adolescents (12-13 years-old) who were followed for 16 years. Time 1 (T1) was defined as the individuals' maximum drinking year within the first 10 study years and Time 2 (T2) was the first available data entry 3-7 years after T1. Four hierarchical linear regression models predicting follow-up alcohol use were estimated: drinking days, average drinks per drinking day, peak drinks, and binge episodes. All models included inhibition/cognitive flexibility, visuospatial ability, verbal memory, working memory, and their interactions with sex, while covarying for age at T1, follow-up duration and controlling for T1 drinking. Results Better visuospatial ability at T1 predicted decreases in later binge episodes at T2 (β= -.19, p= .048, partial r2 = .039). While better inhibition/cognitive flexibility at T1 predicted increases in follow-up drinks per drinking day at T2 (β= .18, p= .016, partial r2= .057). Conclusion Findings suggest specific neurocognitive abilities during maximum drinking in late adolescence are useful as predictors of change in later drinking quantity per occasion and could potentially inform intervention research targeting this age group.
Collapse
Affiliation(s)
- Nafisa Ferdous
- JDP in Interdisciplinary Research on Substance Use, Division of Global Public Health, UC San Diego, and School of Social Work, San Diego State University, San Diego, CA, USA
| | | | | |
Collapse
|
9
|
Tirrell JM, Sampa M, Wootten K, Harris SK, McGrath RE, Mulavu M, Sindano N, Kasanga L, Mweemba O, Seale DM, Seale JP, Mutale W. Measuring Character Strengths and Promoting Positive Youth Development in Zambia: Initial Findings from the GROW Hopes for Life Study. CHILD & YOUTH CARE FORUM 2024; 54:303-322. [PMID: 40115186 PMCID: PMC11920339 DOI: 10.1007/s10566-024-09814-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2024] [Indexed: 03/23/2025]
Abstract
Background The Global Resilience Oral Workshops (GROW) Free and Strong programs take a strengths-based, positive youth development (PYD) approach to promoting thriving. Through both prevention (GROW Strong) and intervention (GROW Free) exercises, these programs aim to build character and emotional resilience while also lowering unhealthy alcohol use. Objective To meaningfully assess the impact of the GROW programs on health and PYD, ecologically and psychometrically valid measures of character strengths were needed, with a focus on the strengths of hope, forgiveness, spirituality, prudence, and self-control (self-regulation) promoted by GROW. Method We tested a series of exploratory and confirmatory factor analyses of these five key constructs using two samples: a school-based youth sample enrolled in GROW Strong (n = 460; M age = 15.04 years, SD age = 1.21; 53.0% female); and a community-based adult sample enrolled in GROW Free (n = 457; M age = 20.60 years, SD age = 1.88; 49.7% female); both enrolled using a waitlist-control design. Results Measures demonstrated strong invariance across specific subgroups present in the data sets, with differences emerging across ages, urban/rural locations, and baseline study conditions. Conclusions To meaningfully document PYD programs and character development in the majority world, measurement models must be theory-predicated, robust, and empirically validated for the specific context. The results provide evidence for such a measure that will be useful in future intervention studies promoting character strengths to address unhealthy alcohol use in Zambia. Supplementary information The online version contains supplementary material available at 10.1007/s10566-024-09814-8.
Collapse
Affiliation(s)
- Jonathan M Tirrell
- Institute for Applied Research in Youth Development, Tufts University, Medford, USA
| | | | | | - Sion Kim Harris
- Boston Children's Hospital, Harvard Medical School, Boston, USA
| | | | | | | | - Lameck Kasanga
- Serenity Harm Reduction Programme Zambia, Lusaka, Zambia
| | | | | | | | | |
Collapse
|
10
|
Boer OD, El Marroun H, Muetzel RL. Adolescent substance use initiation and long-term neurobiological outcomes: insights, challenges and opportunities. Mol Psychiatry 2024; 29:2211-2222. [PMID: 38409597 DOI: 10.1038/s41380-024-02471-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 02/28/2024]
Abstract
The increased frequency of risk taking behavior combined with marked neuromaturation has positioned adolescence as a focal point of research into the neural causes and consequences of substance use. However, little work has provided a summary of the links between adolescent initiated substance use and longer-term brain outcomes. Here we review studies exploring the long-term effects of adolescent-initiated substance use with structural and microstructural neuroimaging. A quarter of all studies reviewed conducted repeated neuroimaging assessments. Long-term alcohol use, as well as tobacco use were consistently associated with smaller frontal cortices and altered white matter microstructure. This association was mostly observed in the ACC, insula and subcortical regions in alcohol users, and for the OFC in tobacco users. Long-term cannabis use was mostly related to altered frontal cortices and hippocampal volumes. Interestingly, cannabis users scanned more years after use initiation tended to show smaller measures of these regions, whereas those with fewer years since initiation showed larger measures. Long-term stimulant use tended to show a similar trend as cannabis in terms of years since initiation in measures of the putamen, insula and frontal cortex. Long-term opioid use was mostly associated with smaller subcortical and insular volumes. Of note, null findings were reported in all substance use categories, most often in cannabis use studies. In the context of the large variety in study designs, substance use assessment, methods, and sample characteristics, we provide recommendations on how to interpret these findings, and considerations for future studies.
Collapse
Affiliation(s)
- Olga D Boer
- Department of Psychology, Education and Child Studies - Erasmus School of Social and Behavioral Sciences, Erasmus University Rotterdam, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Hanan El Marroun
- Department of Psychology, Education and Child Studies - Erasmus School of Social and Behavioral Sciences, Erasmus University Rotterdam, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands.
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
11
|
Owaki Y, Yoshimoto H, Saito G, Dobashi S, Kushio S, Nakamura A, Goto T, Togo Y, Mori K, Hokazono H. Effectiveness of genetic feedback on alcohol metabolism to reduce alcohol consumption in young adults: an open-label randomized controlled trial. BMC Med 2024; 22:205. [PMID: 38769537 PMCID: PMC11106878 DOI: 10.1186/s12916-024-03422-y] [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: 01/18/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND It is unclear whether brief interventions using the combined classification of alcohol-metabolizing enzymes aldehyde dehydrogenase 2 (ALDH2) and alcohol dehydrogenase 1B (ADH1B) together with behavioral changes in alcohol use can reduce excessive alcohol consumption. This study aimed to examine the effects of a brief intervention based on the screening of ALDH2 and ADH1B gene polymorphisms on alcohol consumption in Japanese young adults. METHODS In this open-label randomized controlled trial, we enrolled adults aged 20-30 years who had excessive drinking behavior (average amount of alcohol consumed: men, ≥ 4 drinks/per day and women, ≥ 2 drinks/per day; 1 drink = 10 g of pure alcohol equivalent). Participants were randomized into intervention or control group using a simple random number table. The intervention group underwent saliva-based genotyping of alcohol-metabolizing enzymes (ALDH2 and ADH1B), which were classified into five types. A 30-min in-person or online educational counseling was conducted approximately 1 month later based on genotyping test results and their own drinking records. The control group received traditional alcohol education. Average daily alcohol consumption was calculated based on the drinking diary, which was recorded at baseline and at 3 and 6 months of follow-up. The primary endpoint was average daily alcohol consumption, and the secondary endpoints were the alcohol-use disorder identification test for consumption (AUDIT-C) score and behavioral modification stages assessed using a transtheoretical model. RESULTS Participants were allocated to the intervention (n = 100) and control (n = 96) groups using simple randomization. Overall, 28 (29.2%) participants in the control group and 21 (21.0%) in the intervention group did not complete the follow-up. Average alcohol consumption decreased significantly from baseline to 3 and 6 months in the intervention group but not in the control group. The reduction from baseline alcohol consumption values and AUDIT-C score at 3 months were greater in the intervention group than in the control group (p < 0.001). In addition, the behavioral modification stages were significantly changed by the intervention (p < 0.001). CONCLUSIONS Genetic testing for alcohol-metabolizing enzymes and health guidance on type-specific excessive drinking may be useful for reducing sustained average alcohol consumption associated with behavioral modification. TRIAL REGISTRATION R000050379, UMIN000044148, Registered on June 1, 2021.
Collapse
Affiliation(s)
- Yukiko Owaki
- Department of Nursing, School of Nursing, Gunma Prefectural College of Health Sciences, Maebashi, Japan.
- Research and Development Center for Lifestyle Innovation, University of Tsukuba, Tsukuba, Japan.
| | - Hisashi Yoshimoto
- Research and Development Center for Lifestyle Innovation, University of Tsukuba, Tsukuba, Japan.
- Department of Family Medicine, General Practice and Community Health, Institute of Medicine, University of Tsukuba, Tsukuba, Japan.
| | - Go Saito
- Research and Development Center for Lifestyle Innovation, University of Tsukuba, Tsukuba, Japan
- Department of Primary Care and Medical Education, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Shohei Dobashi
- Research and Development Center for Lifestyle Innovation, University of Tsukuba, Tsukuba, Japan
| | | | | | | | - Yusuke Togo
- Sanwa Laboratory, Sanwa Shurui Company, Usa, Japan
| | | | | |
Collapse
|
12
|
Çon N, Mercan S, Küçüköner A, Çalişkan N. Adolescent intermittent ethanol use in male rats do not change cerebellar cell numbers but initiate astroglial reaction. Int J Dev Neurosci 2024; 84:177-189. [PMID: 38327108 DOI: 10.1002/jdn.10317] [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: 11/07/2023] [Revised: 12/21/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024] Open
Abstract
Alcohol consumption during adolescence causes negative structural changes in the cerebellum and can lead to cognitive and motor skill disorders. Unfortunately, the age at which individuals begin drinking alcohol has decreased in recent years, which has drawn attention to the effects of alcohol on neurological changes during preadolescence. In this study, we investigated the effects of adolescent intermittent ethanol (AIE) exposure on the cellular composition of the cerebellum in male rats, particularly when alcohol consumption begins early. The male rats received eight doses of intermittent intraperitoneal injection of 25% (v/v) ethanol (3 g/kg) or saline from postnatal days (PND) 25 to PND 38. In rats, 28-42 days old corresponds to 10-18 years old in humans. Two hours after the last injection, the cells, neurons, and non-neuronal cells in the cerebellum were immunocytochemically labeled and the total numbers of related cells were calculated using the Isotropic Fractionator method. We found that AIE exposure does not change the cell numbers of the cerebellum in the short term, but it does activate astrocytes in the white matter of the cerebellum. These findings suggest that alcohol use during adolescence impairs the innate immune system and negatively affects brain plasticity.
Collapse
Affiliation(s)
- Nurhan Çon
- Department of Medical Services and Techniques, Ondokuz Mayıs University, Samsun, Turkey
| | - Sevcan Mercan
- Department of Medical Services and Techniques, Ondokuz Mayıs University, Samsun, Turkey
| | - Asuman Küçüköner
- Department of Medical Services and Techniques, Ondokuz Mayıs University, Samsun, Turkey
| | - Nüket Çalişkan
- Department of Medical Services and Techniques, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
13
|
de Bejczy A, Addolorato G, Aubin HJ, Guiraud J, Korpi ER, John Nutt D, Witkiewitz K, Söderpalm B. AUD in perspective. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 175:1-19. [PMID: 38555113 DOI: 10.1016/bs.irn.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Alcohol is a major cause of pre-mature death and individual suffering worldwide, and the importance of diagnosing and treating AUD cannot be overstated. Given the global burden and the high attributable factor of alcohol in a vast number of diseases, the need for additional interventions and the development of new medicines is considered a priority by the World Health Organization (WHO). As of today, AUD is severely under-treated with a treatment gap nearing 90%, strikingly higher than that for other psychiatric disorders. Patients often seek treatment late in the progress of the disease and even among those who seek treatment only a minority receive medication, mirroring the still-prevailing stigma of the disease, and a lack of access to effective treatments, as well as a reluctance to total abstinence. To increase adherence, treatment goals should focus not only on maintaining abstinence, but also on harm reduction and psychosocial functioning. A personalised approach to AUD treatment, with a holistic view, and tailored therapy has the potential to improve AUD treatment outcomes by targeting the heterogeneity in genetics and pathophysiology, as well as reason for, and reaction to drinking. Also, the psychiatric co-morbidity rates are high in AUD and dual diagnosis can worsen symptoms and influence treatment response and should be considered in the treatment strategies.
Collapse
Affiliation(s)
- Andrea de Bejczy
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Giovanni Addolorato
- Department of Medical and Surgical Sciences, Università Cattolica di Roma, Rome, Italy; Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Henri-Jean Aubin
- Université Paris-Saclay, Inserm, CESP, Villejuif, France; AP-HP, Université Paris Saclay, Villejuif, France
| | - Julien Guiraud
- Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, The Netherlands; Vergio, Clichy, France
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - David John Nutt
- Imperial College London and GABA Labs, London, United Kingdom
| | - Katie Witkiewitz
- Department of Psychology and Center on Alcohol, Substance Use, and Addictions, University of New Mexico, Albuquerque, New Mexico, United States
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
14
|
Hu X, Fang Z, Wang F, Mei Z, Huang X, Lin Y, Lin Z. A causal relationship between gut microbiota and subcortical brain structures contributes to the microbiota-gut-brain axis: a Mendelian randomization study. Cereb Cortex 2024; 34:bhae056. [PMID: 38415993 DOI: 10.1093/cercor/bhae056] [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: 12/06/2023] [Revised: 01/27/2024] [Accepted: 01/28/2024] [Indexed: 02/29/2024] Open
Abstract
A correlation between gut microbiota and brain structure, referring to as a component of the gut-brain axis, has been observed in observational studies. However, the causality of this relationship and its specific bacterial taxa remains uncertain. To reveal the causal effects of gut microbiota on subcortical brain volume, we applied Mendelian randomization (MR) studies in this study. Genome-wide association study data were obtained from the MiBioGen Consortium (n = 18,340) and the Enhancing Neuro Imaging Genetics through Meta-Analysis Consortium (n = 13,170). The primary estimate was obtained utilizing the inverse-variance weighted, while heterogeneity and pleiotropy were assessed using the Cochrane Q statistic, MR Pleiotropy RESidual Sum and Outlier, and MR-Egger intercept. Our findings provide strong evidence that a higher abundance of the genus Parasutterella is causally correlated with a decrease in intracranial volume (β = -30,921.33, 95% CI -46,671.78 to -15,170.88, P = 1.19 × 10-4), and the genus FamilyXIIIUCG001 is associated with a decrease in thalamus volume (β = -141.96, 95% CI: -214.81 to -69.12, P = 1.0× 10-4). This MR study offers novel perspectives on the intricate interplay between the gut microbiota and subcortical brain volume, thereby lending some support to the existence of the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Xuequn Hu
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Changle District, Fuzhou 350209, Fujian Province, China
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Zhiyong Fang
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Changle District, Fuzhou 350209, Fujian Province, China
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Feng Wang
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Changle District, Fuzhou 350209, Fujian Province, China
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Zhen Mei
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Changle District, Fuzhou 350209, Fujian Province, China
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Xiaofen Huang
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Changle District, Fuzhou 350209, Fujian Province, China
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Yuanxiang Lin
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Changle District, Fuzhou 350209, Fujian Province, China
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Zhangya Lin
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Changle District, Fuzhou 350209, Fujian Province, China
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| |
Collapse
|
15
|
Lynch S, Becker T, Shanker P, Martin D, Staudenmaier P, Leong A, Rice T. Predictors and Correlates of Positive Urine Drug Screening in a Retrospective Cohort Analysis of Child and Adolescent Psychiatry Inpatients Throughout the COVID-19 Pandemic. SUBSTANCE USE & ADDICTION JOURNAL 2024; 45:33-43. [PMID: 38258854 PMCID: PMC11741903 DOI: 10.1177/29767342231210711] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
BACKGROUND AND OBJECTIVES Youth substance use is associated with significant psychological, neurological, and medical complications. Risk factors for substance use among children and adolescents in the general population include peer and/or parental substance use, certain psychiatric illnesses (eg, Attention-Deficit/Hyperactivity Disorder, depression), and history of maltreatment. Co-occurring substance use and psychiatric illness have been associated with increased suicidality, but few prior studies have characterized substance use among child/adolescent inpatients. As such, it remains unclear how substance use contributing to acute psychiatric presentations has changed since the start of the COVID-19 pandemic. METHODS This is a retrospective cohort study of 816 unique child/adolescent psychiatry inpatients with urine drug screening (UDS) results from a diverse urban setting. Charts of patients hospitalized between June 1, 2018 and November 30, 2021 were reviewed for sociodemographic characteristics, indication for admission, psychiatric history, hospital course, treatment plan, and discharge diagnosis. Differences in sociodemographic and clinical characteristics, such as age, race, and diagnoses, between patients with and without positive UDS were explored throughout various periods of the COVID-19 pandemic. Descriptive and comparative statistics were performed, as well as a logistic regression model to identify the predictors of positive UDS. RESULTS Of the study sample, 18% had a positive UDS. Older age, diagnosis of impulsive or behavioral disorder, and a history of violence were found to be predictors of positive UDS. Asian/South Asian or Hispanic/LatinX race and history of a developmental or intellectual disability were found to be negative predictors. The frequency of positive UDS in this population did not change based on COVID-19. DISCUSSION AND CONCLUSIONS Multiple factors may predispose children and adolescents to substance use. Though no impact of COVID-19 was found in this sample, longer-term studies are needed. SCIENTIFIC SIGNIFICANCE This study identifies independent predictors of active substance use in the child and adolescent psychiatric inpatient population.
Collapse
Affiliation(s)
- Sean Lynch
- Icahn School of Medicine at Mount Sinai, Mount Sinai Beth Israel, Department of Psychiatry, New York NY
| | - Timothy Becker
- Icahn School of Medicine at Mount Sinai, Mount Sinai Hospital, Department of Psychiatry, New York NY
| | - Parul Shanker
- Icahn School of Medicine at Mount Sinai, Mount Sinai Beth Israel, Department of Psychiatry, New York NY
| | - Dalton Martin
- Icahn School of Medicine at Mount Sinai, Mount Sinai Morningside, Department of Psychiatry, New York NY
| | - Paige Staudenmaier
- Icahn School of Medicine at Mount Sinai, Mount Sinai Beth Israel, Department of Psychiatry, New York NY
| | - Alicia Leong
- Icahn School of Medicine at Mount Sinai, New York NY
| | - Timothy Rice
- Icahn School of Medicine at Mount Sinai, Mount Sinai Morningside, Department of Psychiatry, New York NY
| |
Collapse
|
16
|
Kirse HA, Bahrami M, Lyday RG, Simpson SL, Peterson-Sockwell H, Burdette JH, Laurienti PJ. Differences in Brain Network Topology Based on Alcohol Use History in Adolescents. Brain Sci 2023; 13:1676. [PMID: 38137124 PMCID: PMC10741456 DOI: 10.3390/brainsci13121676] [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: 09/21/2023] [Revised: 11/10/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Approximately 6 million youth aged 12 to 20 consume alcohol monthly in the United States. The effect of alcohol consumption in adolescence on behavior and cognition is heavily researched; however, little is known about how alcohol consumption in adolescence may alter brain function, leading to long-term developmental detriments. In order to investigate differences in brain connectivity associated with alcohol use in adolescents, brain networks were constructed using resting-state functional magnetic resonance imaging data collected by the National Consortium on Alcohol and NeuroDevelopment in Adolescence (NCANDA) from 698 youth (12-21 years; 117 hazardous drinkers and 581 no/low drinkers). Analyses assessed differences in brain network topology based on alcohol consumption in eight predefined brain networks, as well as in whole-brain connectivity. Within the central executive network (CEN), basal ganglia network (BGN), and sensorimotor network (SMN), no/low drinkers demonstrated stronger and more frequent connections between highly globally efficient nodes, with fewer and weaker connections between highly clustered nodes. Inverse results were observed within the dorsal attention network (DAN), visual network (VN), and frontotemporal network (FTN), with no/low drinkers demonstrating weaker connections between nodes with high efficiency and increased frequency of clustered nodes compared to hazardous drinkers. Cross-sectional results from this study show clear organizational differences between adolescents with no/low or hazardous alcohol use, suggesting that aberrant connectivity in these brain networks is associated with risky drinking behaviors.
Collapse
Affiliation(s)
- Haley A. Kirse
- Laboratory for Complex Brain Networks, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (H.A.K.); (M.B.); (R.G.L.); (S.L.S.); (H.P.-S.); (J.H.B.)
- Graduate Program, Wake Forest Graduate School of Arts and Sciences, Integrative Physiology and Pharmacology, Winston-Salem, NC 27101, USA
| | - Mohsen Bahrami
- Laboratory for Complex Brain Networks, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (H.A.K.); (M.B.); (R.G.L.); (S.L.S.); (H.P.-S.); (J.H.B.)
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Robert G. Lyday
- Laboratory for Complex Brain Networks, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (H.A.K.); (M.B.); (R.G.L.); (S.L.S.); (H.P.-S.); (J.H.B.)
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Sean L. Simpson
- Laboratory for Complex Brain Networks, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (H.A.K.); (M.B.); (R.G.L.); (S.L.S.); (H.P.-S.); (J.H.B.)
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Hope Peterson-Sockwell
- Laboratory for Complex Brain Networks, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (H.A.K.); (M.B.); (R.G.L.); (S.L.S.); (H.P.-S.); (J.H.B.)
| | - Jonathan H. Burdette
- Laboratory for Complex Brain Networks, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (H.A.K.); (M.B.); (R.G.L.); (S.L.S.); (H.P.-S.); (J.H.B.)
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Paul J. Laurienti
- Laboratory for Complex Brain Networks, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (H.A.K.); (M.B.); (R.G.L.); (S.L.S.); (H.P.-S.); (J.H.B.)
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
17
|
Mewton L, Davies S, Sunderland M, Champion K, Hoy N, Newton N, Teesson M, Squeglia LM. Longitudinal relationships between lifestyle risk factors and neurodevelopment in early adolescence. Health Psychol 2023; 42:904-912. [PMID: 37616102 PMCID: PMC10840638 DOI: 10.1037/hea0001248] [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] [Indexed: 08/25/2023]
Abstract
OBJECTIVE The goal of this study is to investigate the cross-sectional and longitudinal relationships between clustered lifestyle risk factors (sleep, physical activity, body mass index [BMI], and screen time) and neurodevelopment over the early adolescent period. METHOD Data from the ABCD Study Data Release 3.0 consisted of 11,878 participants (aged 9-10 years) at baseline and 6,571 participants (aged 11-12 years) at 2-year follow-up. The interrelationships between lifestyle risk factors and brain structure were analyzed using bivariate multiple indicator latent change score models. Using confirmatory factor analysis, a single lifestyle risk factor domain (measured by sleep, physical activity, BMI, and screen time) was shown to fit the data well. Using exploratory and confirmatory factor analysis, seven brain structure domains were extracted and labeled as temporal-parietal, frontotemporal, occipital, orbitofrontal, temporal, cingulate, parietal, and cuneus domains. All bivariate latent change score models accounted for age, sex at birth, race/ethnicity, parental education, and marital status. RESULTS Higher lifestyle risk was associated with smaller brain volume at baseline. Higher baseline lifestyle risk was also associated with a greater rate of change (i.e., greater decreases) in brain volume for the temporal-parietal, frontotemporal, orbitofrontal, parietal, and cuneus domains. Effects were not reciprocal; baseline brain volume did not predict changes in lifestyle behaviors over time. CONCLUSION These findings are important for understanding the biological mechanisms underpinning health risk factors and can be used to target interventions and improve brain health during this critical developmental phase. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
- Louise Mewton
- Centre for Healthy Brain Ageing, School of Psychiatry and Mental Health, University of New South Wales, Sydney, Australia
| | - Sarah Davies
- Centre for Healthy Brain Ageing, School of Psychiatry and Mental Health, University of New South Wales, Sydney, Australia
| | - Matthew Sunderland
- The Matilda Centre for Mental Health and Substance Use, University of Sydney, Sydney, Australia
| | - Katrina Champion
- The Matilda Centre for Mental Health and Substance Use, University of Sydney, Sydney, Australia
| | - Nicholas Hoy
- Centre for Healthy Brain Ageing, School of Psychiatry and Mental Health, University of New South Wales, Sydney, Australia
| | - Nicola Newton
- The Matilda Centre for Mental Health and Substance Use, University of Sydney, Sydney, Australia
| | - Maree Teesson
- The Matilda Centre for Mental Health and Substance Use, University of Sydney, Sydney, Australia
| | - Lindsay M. Squeglia
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
18
|
Etami Y, Lildharrie C, Manza P, Wang GJ, Volkow ND. Neuroimaging in Adolescents: Post-Traumatic Stress Disorder and Risk for Substance Use Disorders. Genes (Basel) 2023; 14:2113. [PMID: 38136935 PMCID: PMC10743116 DOI: 10.3390/genes14122113] [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: 10/01/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Trauma in childhood and adolescence has long-term negative consequences in brain development and behavior and increases the risk for psychiatric disorders. Among them, post-traumatic stress disorder (PTSD) during adolescence illustrates the connection between trauma and substance misuse, as adolescents may utilize substances to cope with PTSD. Drug misuse may in turn lead to neuroadaptations in learning processes that facilitate the consolidation of traumatic memories that perpetuate PTSD. This reflects, apart from common genetic and epigenetic modifications, overlapping neurocircuitry engagement triggered by stress and drug misuse that includes structural and functional changes in limbic brain regions and the salience, default-mode, and frontoparietal networks. Effective strategies to prevent PTSD are needed to limit the negative consequences associated with the later development of a substance use disorder (SUD). In this review, we will examine the link between PTSD and SUDs, along with the resulting effects on memory, focusing on the connection between the development of an SUD in individuals who struggled with PTSD in adolescence. Neuroimaging has emerged as a powerful tool to provide insight into the brain mechanisms underlying the connection of PTSD in adolescence and the development of SUDs.
Collapse
Affiliation(s)
| | | | | | - Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (Y.E.); (C.L.); (P.M.); (N.D.V.)
| | | |
Collapse
|
19
|
Steinfeld MR, Torregrossa MM. Consequences of adolescent drug use. Transl Psychiatry 2023; 13:313. [PMID: 37802983 PMCID: PMC10558564 DOI: 10.1038/s41398-023-02590-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/22/2023] [Accepted: 08/23/2023] [Indexed: 10/08/2023] Open
Abstract
Substance use in adolescence is a known risk factor for the development of neuropsychiatric and substance use disorders in adulthood. This is in part due to the fact that critical aspects of brain development occur during adolescence, which can be altered by drug use. Despite concerted efforts to educate youth about the potential negative consequences of substance use, initiation remains common amongst adolescents world-wide. Additionally, though there has been substantial research on the topic, many questions remain about the predictors and the consequences of adolescent drug use. In the following review, we will highlight some of the most recent literature on the neurobiological and behavioral effects of adolescent drug use in rodents, non-human primates, and humans, with a specific focus on alcohol, cannabis, nicotine, and the interactions between these substances. Overall, consumption of these substances during adolescence can produce long-lasting changes across a variety of structures and networks which can have enduring effects on behavior, emotion, and cognition.
Collapse
Affiliation(s)
- Michael R Steinfeld
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA.
| | - Mary M Torregrossa
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA
| |
Collapse
|
20
|
Kiss O, Goldstone A, de Zambotti M, Yüksel D, Hasler BP, Franzen PL, Brown SA, De Bellis MD, Nagel BJ, Nooner KB, Tapert SF, Colrain IM, Clark DB, Baker FC. Effects of emerging alcohol use on developmental trajectories of functional sleep measures in adolescents. Sleep 2023; 46:zsad113. [PMID: 37058610 PMCID: PMC10848227 DOI: 10.1093/sleep/zsad113] [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: 09/01/2022] [Revised: 02/17/2023] [Indexed: 04/16/2023] Open
Abstract
STUDY OBJECTIVES Adolescence is characterized by significant brain development, accompanied by changes in sleep timing and architecture. It also is a period of profound psychosocial changes, including the initiation of alcohol use; however, it is unknown how alcohol use affects sleep architecture in the context of adolescent development. We tracked developmental changes in polysomnographic (PSG) and electroencephalographic (EEG) sleep measures and their relationship with emergent alcohol use in adolescents considering confounding effects (e.g. cannabis use). METHODS Adolescents (n = 94, 43% female, age: 12-21 years) in the National Consortium on Alcohol and Neurodevelopment in Adolescence (NCANDA) study had annual laboratory PSG recordings across 4-years. Participants were no/low drinkers at baseline. RESULTS Linear mixed effect models showed developmental changes in sleep macrostructure and EEG, including a decrease in slow wave sleep and slow wave (delta) EEG activity with advancing age. Emergent moderate/heavy alcohol use across three follow-up years was associated with a decline in percentage rapid eye movement (REM) sleep over time, a longer sleep onset latency (SOL) and shorter total sleep time (TST) in older adolescents, and lower non-REM delta and theta power in males. CONCLUSIONS These longitudinal data show substantial developmental changes in sleep architecture. Emergent alcohol use during this period was associated with altered sleep continuity, architecture, and EEG measures, with some effects dependent on age and sex. These effects, in part, could be attributed to the effects of alcohol on underlying brain maturation processes involved in sleep-wake regulation.
Collapse
Affiliation(s)
- Orsolya Kiss
- Center for Health Sciences, Bioscience Division, SRI International, Menlo Park, CA, USA
| | - Aimée Goldstone
- Center for Health Sciences, Bioscience Division, SRI International, Menlo Park, CA, USA
| | | | - Dilara Yüksel
- Center for Health Sciences, Bioscience Division, SRI International, Menlo Park, CA, USA
| | - Brant P Hasler
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter L Franzen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sandra A Brown
- Department of Psychiatry, University of California, San Diego, San Diego, CA, USA
| | - Michael D De Bellis
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Bonnie J Nagel
- School of Medicine, Division of Clinical Psychology, Oregon Health and Sciences University, Portland, OR, USA
| | - Kate B Nooner
- Psychology Department, University of North Carolina Wilmington, Wilmington, NC, USA
| | - Susan F Tapert
- Department of Psychiatry, University of California, San Diego, San Diego, CA, USA
| | - Ian M Colrain
- Center for Health Sciences, Bioscience Division, SRI International, Menlo Park, CA, USA
| | - Duncan B Clark
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fiona C Baker
- Center for Health Sciences, Bioscience Division, SRI International, Menlo Park, CA, USA
- Brain Function Research Group, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
21
|
Baranger DAA, Paul SE, Hatoum AS, Bogdan R. Alcohol use and grey matter structure: Disentangling predispositional and causal contributions in human studies. Addict Biol 2023; 28:e13327. [PMID: 37644894 PMCID: PMC10502907 DOI: 10.1111/adb.13327] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/23/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023]
Abstract
Alcohol use is a growing global health concern and economic burden. Alcohol involvement (i.e., initiation, use, problematic use, alcohol use disorder) has been reliably associated with broad spectrum grey matter differences in cross-sectional studies. These findings have been largely interpreted as reflecting alcohol-induced atrophy. However, emerging data suggest that brain structure differences also represent pre-existing vulnerability factors for alcohol involvement. Here, we review evidence from human studies with designs (i.e., family-based, genomic, longitudinal) that allow them to assess the plausibility that these correlates reflect predispositional risk factors and/or causal consequences of alcohol involvement. These studies provide convergent evidence that grey matter correlates of alcohol involvement largely reflect predisposing risk factors, with some evidence for potential alcohol-induced atrophy. These conclusions highlight the importance of study designs that can provide causal clues to cross-sectional observations. An integrative model may best account for these data, in which predisposition to alcohol use affects brain development, effects which may then be compounded by the neurotoxic consequences of heavy alcohol use.
Collapse
Affiliation(s)
- David A A Baranger
- Department of Psychiatry, Washington University St. Louis Medical School, St. Louis, Missouri, USA
| | - Sarah E Paul
- Department of Psychological & Brain Sciences, Washington University St. Louis, St. Louis, Missouri, USA
| | - Alexander S Hatoum
- Department of Psychological & Brain Sciences, Washington University St. Louis, St. Louis, Missouri, USA
- Artificial Intelligence and the Internet of Things in Medicine Institute, Washington University St. Louis Medical School, St. Louis, Missouri, USA
| | - Ryan Bogdan
- Department of Psychological & Brain Sciences, Washington University St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
22
|
Lee SH, Shnitko TA, Hsu LM, Broadwater MA, Sardinas M, Wang TWW, Robinson DL, Vetreno RP, Crews FT, Shih YYI. Acute alcohol induces greater dose-dependent increase in the lateral cortical network functional connectivity in adult than adolescent rats. ADDICTION NEUROSCIENCE 2023; 7:100105. [PMID: 37576436 PMCID: PMC10421607 DOI: 10.1016/j.addicn.2023.100105] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Alcohol misuse and, particularly adolescent drinking, is a major public health concern. While evidence suggests that adolescent alcohol use affects frontal brain regions that are important for cognitive control over behavior little is known about how acute alcohol exposure alters large-scale brain networks and how sex and age may moderate such effects. Here, we employ a recently developed functional magnetic resonance imaging (fMRI) protocol to acquire rat brain functional connectivity data and use an established analytical pipeline to examine the effect of sex, age, and alcohol dose on connectivity within and between three major rodent brain networks: defaul mode, salience, and lateral cortical network. We identify the intra- and inter-network connectivity differences and establish moderation models to reveal significant influences of age on acute alcohol-induced lateral cortical network connectivity. Through this work, we make brain-wide isotropic fMRI data with acute alcohol challenge publicly available, with the hope to facilitate future discovery of brain regions/circuits that are causally relevant to the impact of acute alcohol use.
Collapse
Affiliation(s)
- Sung-Ho Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| | - Tatiana A. Shnitko
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Li-Ming Hsu
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Margaret A. Broadwater
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| | - Mabelle Sardinas
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Tzu-Wen Winnie Wang
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Donita L. Robinson
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Fulton T. Crews
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Tolstrup JS, Kruckow S, Becker U, Andersen O, Sawyer SM, Katikireddi SV, Møller SP. Socioeconomic inequalities in alcohol-related harm in adolescents: a prospective cohort study of 68,299 Danish 15-19-year-olds. EClinicalMedicine 2023; 62:102129. [PMID: 37576460 PMCID: PMC10415833 DOI: 10.1016/j.eclinm.2023.102129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/27/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Background Evidence shows that similar levels of alcohol consumption lead to greater harm in adults with low socioeconomic position (SEP) compared to high SEP. We investigated if SEP is associated with alcohol-related hospital contacts in adolescents, and whether differences in risk can be explained by differences in levels of alcohol consumption, drinking pattern, and substance use. Methods This is a prospective cohort study of 68,299 participants aged 15-19 years old from the Danish National Youth Cohort 2014. SEP was operationalised as parent educational level, family income and perceived financial strain in the family. Data were linked to national registers and participants were followed up for five years from 2014 to 2019. Outcomes were hospital contacts due to alcohol. Multilevel Poisson regression was used to estimate incidence rates (IR) and incidence rate ratios (IRR). Findings During 280,010 person years of follow-up, 872 participants had an alcohol-attributable hospital contact; intoxications (n = 778, 89%) were the most common diagnosis. Low as compared to high SEP was associated with higher IRR of alcohol-attributable hospital contacts for all three SEP measures. The adjusted IRR of harm was 1.73 (95% CI: 1.29-2.33) for elementary school as the highest parent education compared to longer parent education and 1.57 (95% CI: 1.30-1.89) for family financial strain compared to those without financial strain. Adjustment for weekly alcohol intake, drinking pattern and substance use did not substantially change results. Cubic spline analysis of the association between family income and alcohol-attributable hospital contacts revealed a dose-response relationship with decreasing risk of alcohol-related harm with higher income. Interpretation Our findings suggested that alcohol-related harm is more common in socioeconomically disadvantaged adolescents despite similar levels of alcohol consumption, regardless of differences in drinking pattern or substance use. Future preventive strategies should prioritise young adolescents, including those who are most disadvantaged. Funding Tryg Foundation (ID: 153539).
Collapse
Affiliation(s)
- Janne S. Tolstrup
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Sofie Kruckow
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Ulrik Becker
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Susan M. Sawyer
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Centre for Adolescent Health, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Srinivasa Vittal Katikireddi
- Medical Research Council (MRC)/Chief Scientist Office (CSO) Social & Public Health Sciences Unit, University of Glasgow, UK
| | - Sanne Pagh Møller
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| |
Collapse
|
24
|
Juntunen A, Määttä S, Könönen M, Kallioniemi E, Niskanen E, Kaarre O, Kivimäki P, Vanninen R, Tolmunen T, Ferreri F, Kekkonen V. Cortical thickness is inversely associated with transcranial magnetic stimulation-evoked N45 potential among young adults whose heavy drinking began in adolescence. Alcohol Clin Exp Res 2023; 47:1341-1351. [PMID: 37526579 DOI: 10.1111/acer.15119] [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: 02/03/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND Adolescence is a particularly vulnerable stage of development in terms of the deleterious effects of alcohol. Both lower gray matter (GM) volume and greater GABAergic activity have been associated with chronic alcohol consumption during adolescence. However, the association between these measures has not been investigated. METHODS In this exploratory study, we compared 26 young adults with a 10year history of heavy alcohol consumption with 21 controls who used little or no alcohol. Simultaneous transcranial magnetic stimulation and electroencephalography were used to assess transcranial magnetic stimulation-evoked N45 potentials, reflecting a balance between GABAergic inhibition and N-methyl-D-aspartate (NMDA) receptor-mediated glutaminergic excitation in the brain. GM thickness was measured from magnetic resonance images and GM and N45 potentials were then correlated. RESULTS Cortical thickness was significantly lower in several brain regions in the heavy-drinking group than the light-drinking group. The N45 amplitude was significantly larger frontally in the heavy-drinking group. Among heavy drinkers, there were several statistically significant correlations between thinner GM and larger frontal N45 amplitudes that were not detectable in the light-drinking group. The strongest correlations were detected in the frontal and parietal lobes, especially in the left superior frontal gyrus and the left supramarginal gyrus, and in both hemispheres in the superior parietal lobes. CONCLUSIONS These findings show that a thinner cortex and greater inhibitory neurotransmission are correlated in certain brain regions among young, long-term heavy alcohol users. Studies are needed to explore the possible causal mechanisms underlying these effects.
Collapse
Affiliation(s)
- Anna Juntunen
- School of Medicine, Faculty of Health, University of Eastern Finland, Kuopio, Finland
| | - Sara Määttä
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland
| | - Mervi Könönen
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Elisa Kallioniemi
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Eini Niskanen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Outi Kaarre
- School of Medicine, Faculty of Health, University of Eastern Finland, Kuopio, Finland
- Forensic Psychiatry Clinic of the University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Petri Kivimäki
- School of Medicine, Faculty of Health, University of Eastern Finland, Kuopio, Finland
- Vuosaari Psychiatric Outpatient Clinic, Vuosaari Health and Well-being Centre, City of Helsinki, Finland
| | - Ritva Vanninen
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Tommi Tolmunen
- School of Medicine, Faculty of Health, University of Eastern Finland, Kuopio, Finland
- Department of Adolescent Psychiatry, Kuopio University Hospital, Kuopio, Finland
| | - Florinda Ferreri
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland
- Unit of Neurology, Unit of Clinical Neurophysiology and Study Center of Neurodegeneration (CESNE), Department of Neuroscience, University of Padua, Padua, Italy
| | - Virve Kekkonen
- School of Medicine, Faculty of Health, University of Eastern Finland, Kuopio, Finland
- Department of Adolescent Psychiatry, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
25
|
Fan S, Goldfarb EV, Lacadie C, Fogelman N, Seo D, Sinha R. Binge drinking is associated with higher cortisol and lower hippocampal and prefrontal gray matter volume: Prospective association with future alcohol intake. Neurobiol Stress 2023; 25:100540. [PMID: 37323647 PMCID: PMC10265520 DOI: 10.1016/j.ynstr.2023.100540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/07/2023] [Accepted: 04/20/2023] [Indexed: 06/17/2023] Open
Abstract
Background Cortisol is a significant driver of the biological stress response that is potently activated by acute alcohol intake and increased with binge drinking. Binge drinking is associated with negative social and health consequences and risk of developing alcohol use disorder (AUD). Both cortisol levels and AUD are also associated with changes in hippocampal and prefrontal regions. However, no previous research has assessed structural gray matter volume (GMV) and cortisol concurrently to examine BD effects on hippocampal and prefrontal GMV and cortisol, and their prospective relationship to future alcohol intake. Methods Individuals who reported binge drinking (BD: N = 55) and demographically matched non-binge moderate drinkers (MD: N = 58) were enrolled and scanned using high-resolution structural MRI. Whole brain voxel-based morphometry was used to quantify regional GMV. In a second phase, 65% of the sample volunteered to participate in prospective daily assessment of alcohol intake for 30 days post-scanning. Results Relative to MD, BD showed significantly higher cortisol and smaller GMV in regions including hippocampus, dorsal lateral prefrontal cortex (dlPFC), prefrontal and supplementary motor, primary sensory and posterior parietal cortex (FWE, p < 0.05). GMV in bilateral dlPFC and motor cortices were negatively associated with cortisol levels, and smaller GMV in multiple PFC regions was associated with more subsequent drinking days in BD. Conclusion These findings indicate neuroendocrine and structural dysregulation associated with BD relative to MD. Notably, BD-associated lower GMV regions were those involved in stress, memory and cognitive control, with lower GMV in cognitive control and motor regions also predicting higher levels of future alcohol intake in BD.
Collapse
Affiliation(s)
- Siyan Fan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Cheryl Lacadie
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Nia Fogelman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Dongju Seo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Rajita Sinha
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
26
|
Joo Y, Lee S, Hwang J, Kim J, Cheon YH, Lee H, Kim S, Yurgelun-Todd DA, Renshaw PF, Yoon S, Lyoo IK. Differential alterations in brain structural network organization during addiction between adolescents and adults. Psychol Med 2023; 53:3805-3816. [PMID: 35440353 PMCID: PMC10317813 DOI: 10.1017/s0033291722000423] [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: 07/19/2021] [Revised: 01/06/2022] [Accepted: 02/04/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The adolescent brain may be susceptible to the influences of illicit drug use. While compensatory network reorganization is a unique developmental characteristic that may restore several brain disorders, its association with methamphetamine (MA) use-induced damage during adolescence is unclear. METHODS Using independent component (IC) analysis on structural magnetic resonance imaging data, spatially ICs described as morphometric networks were extracted to examine the effects of MA use on gray matter (GM) volumes and network module connectivity in adolescents (51 MA users v. 60 controls) and adults (54 MA users v. 60 controls). RESULTS MA use was related to significant GM volume reductions in the default mode, cognitive control, salience, limbic, sensory and visual network modules in adolescents. GM volumes were also reduced in the limbic and visual network modules of the adult MA group as compared to the adult control group. Differential patterns of structural connectivity between the basal ganglia (BG) and network modules were found between the adolescent and adult MA groups. Specifically, adult MA users exhibited significantly reduced connectivity of the BG with the default network modules compared to control adults, while adolescent MA users, despite the greater extent of network GM volume reductions, did not show alterations in network connectivity relative to control adolescents. CONCLUSIONS Our findings suggest the potential of compensatory network reorganization in adolescent brains in response to MA use. The developmental characteristic to compensate for MA-induced brain damage can be considered as an age-specific therapeutic target for adolescent MA users.
Collapse
Affiliation(s)
- Yoonji Joo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Suji Lee
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
| | - Jaeuk Hwang
- Department of Psychiatry, Soonchunhyang University College of Medicine, Seoul, South Korea
| | - Jungyoon Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Young-Hoon Cheon
- Department of Psychiatry, Incheon Chamsarang Hospital, Incheon, South Korea
| | - Hyangwon Lee
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Shinhye Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Deborah A. Yurgelun-Todd
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
- Diagnostic Neuroimaging, University of Utah, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, VA VISN 19 Mental Illness Research, Education and Clinical Center (MIRECC), Salt Lake City, UT, USA
| | - Perry F. Renshaw
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
- Diagnostic Neuroimaging, University of Utah, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, VA VISN 19 Mental Illness Research, Education and Clinical Center (MIRECC), Salt Lake City, UT, USA
| | - Sujung Yoon
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - In Kyoon Lyoo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
27
|
Zhao Y, Yang X, Cheng S, Li C, He D, Cai Q, Wei W, Qin X, Zhang N, Shi S, Chu X, Meng P, Zhang F. Assessing the effect of interaction between lifestyle and longitudinal changes in brain structure on sleep phenotypes. Cereb Cortex 2023:7030864. [PMID: 36750265 DOI: 10.1093/cercor/bhac526] [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: 10/11/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 02/09/2023] Open
Abstract
Longitudinal changes in brain structure and lifestyle can affect sleep phenotypes. However, the influence of the interaction between longitudinal changes in brain structure and lifestyle on sleep phenotypes remains unclear. Genome-wide association study dataset of longitudinal changes in brain structure was obtained from published study. Phenotypic data of lifestyles and sleep phenotypes were obtained from UK Biobank cohort. Using genotype data from UK Biobank, we calculated polygenetic risk scores of longitudinal changes in brain structure phenotypes. Linear/logistic regression analysis was conducted to evaluate interactions between longitudinal changes in brain structure and lifestyles on sleep duration, chronotype, insomnia, snoring and daytime dozing. Multiple lifestyle × longitudinal changes in brain structure interactions were detected for 5 sleep phenotypes, such as physical activity×caudate_age2 for daytime dozing (OR = 1.0389, P = 8.84 × 10-3) in total samples, coffee intake×cerebellar white matter volume_age2 for daytime dozing (OR = 0.9652, P = 1.13 × 10-4) in females. Besides, we found 4 overlapping interactions in different sleep phenotypes. We conducted sex stratification analysis and identified one overlapping interaction between female and male. Our results support the moderate effects of interaction between lifestyle and longitudinal changes in brain structure on sleep phenotypes, and deepen our understanding of the pathogenesis of sleep disorders.
Collapse
Affiliation(s)
- Yijing Zhao
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Chun'e Li
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Dan He
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Qingqing Cai
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wenming Wei
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyue Qin
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Na Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Sirong Shi
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoge Chu
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Peilin Meng
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
28
|
Sun D, Adduru VR, Phillips RD, Bouchard HC, Sotiras A, Michael AM, Baker FC, Tapert SF, Brown SA, Clark DB, Goldston D, Nooner KB, Nagel BJ, Thompson WK, De Bellis MD, Morey RA. Adolescent alcohol use is linked to disruptions in age-appropriate cortical thinning: an unsupervised machine learning approach. Neuropsychopharmacology 2023; 48:317-326. [PMID: 36209230 PMCID: PMC9750971 DOI: 10.1038/s41386-022-01457-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/16/2022] [Accepted: 09/06/2022] [Indexed: 12/26/2022]
Abstract
Cortical thickness changes dramatically during development and is associated with adolescent drinking. However, previous findings have been inconsistent and limited by region-of-interest approaches that are underpowered because they do not conform to the underlying spatially heterogeneous effects of alcohol. In this study, adolescents (n = 657; 12-22 years at baseline) from the National Consortium on Alcohol and Neurodevelopment in Adolescence (NCANDA) study who endorsed little to no alcohol use at baseline were assessed with structural magnetic resonance imaging and followed longitudinally at four yearly intervals. Seven unique spatial patterns of covarying cortical thickness were obtained from the baseline scans by applying an unsupervised machine learning method called non-negative matrix factorization (NMF). The cortical thickness maps of all participants' longitudinal scans were projected onto vertex-level cortical patterns to obtain participant-specific coefficients for each pattern. Linear mixed-effects models were fit to each pattern to investigate longitudinal effects of alcohol consumption on cortical thickness. We found in six NMF-derived cortical thickness patterns, the longitudinal rate of decline in no/low drinkers was similar for all age cohorts. Among moderate drinkers the decline was faster in the younger adolescent cohort and slower in the older cohort. Among heavy drinkers the decline was fastest in the younger cohort and slowest in the older cohort. The findings suggested that unsupervised machine learning successfully delineated spatially coordinated patterns of vertex-level cortical thickness variation that are unconstrained by neuroanatomical features. Age-appropriate cortical thinning is more rapid in younger adolescent drinkers and slower in older adolescent drinkers, an effect that is strongest among heavy drinkers.
Collapse
Affiliation(s)
- Delin Sun
- Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
- VA Mid-Atlantic MIRECC, Durham VA Medical Center, Durham VA, Durham, NC, USA
- Department of Psychology, The Education University of Hong Kong, Hong Kong, China
| | - Viraj R Adduru
- Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
- VA Mid-Atlantic MIRECC, Durham VA Medical Center, Durham VA, Durham, NC, USA
- Chester F. Carlson Center for Imaging Sciences, Rochester Institute of Technology, Rochester, NY, USA
| | - Rachel D Phillips
- Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
- VA Mid-Atlantic MIRECC, Durham VA Medical Center, Durham VA, Durham, NC, USA
| | - Heather C Bouchard
- Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
- VA Mid-Atlantic MIRECC, Durham VA Medical Center, Durham VA, Durham, NC, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute for Informatics, University of Washington, St Louis, MO, USA
| | - Andrew M Michael
- Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
- Chester F. Carlson Center for Imaging Sciences, Rochester Institute of Technology, Rochester, NY, USA
| | - Fiona C Baker
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | - Susan F Tapert
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Sandra A Brown
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Duncan B Clark
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - David Goldston
- Department of Psychology, University of North Carolina Wilmington, Wilmington, NC, USA
| | - Kate B Nooner
- Department of Psychology, University of North Carolina Wilmington, Wilmington, NC, USA
| | - Bonnie J Nagel
- Departments of Psychiatry and Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Wesley K Thompson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Michael D De Bellis
- Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Rajendra A Morey
- Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham, NC, USA.
- VA Mid-Atlantic MIRECC, Durham VA Medical Center, Durham VA, Durham, NC, USA.
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.
- Center for Cognitive Neuroscience, Duke University, Durham, NC, USA.
| |
Collapse
|
29
|
Deschamps C, Debris M, Vilpoux C, Naassila M, Pierrefiche O. [Binge drinking in the young population: Lost memory after initial ethanol exposure via neuroinflammation and epigenetic]. Med Sci (Paris) 2023; 39:31-37. [PMID: 36692315 DOI: 10.1051/medsci/2022191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Binge drinking (BD) in young adults/adolescents can lead to cognitive deficits in the adult probably through neuroinflammation and epigenetic. However, the mode of action of alcohol during the initial exposure is less known while it may be the origin of the deficits seen in adults. Recent studies in adolescent rat hippocampus revealed that loss of memory occurred since the very first exposure to BD with similar mechanisms than those highlighted for longer alcohol exposure. Thus, initiation to BD in the young is responsible for cognitive deficits that will be probably entertained by repeated BD behavior. These kind of data may serve to reinforce the prevention campaigns towards the young population who practice BD.
Collapse
Affiliation(s)
- Chloé Deschamps
- Université de Picardie Jules Verne, Inserm UMR1247 GRAP, Amiens, France
| | - Margot Debris
- Université de Picardie Jules Verne, Inserm UMR1247 GRAP, Amiens, France
| | - Catherine Vilpoux
- Université de Picardie Jules Verne, Inserm UMR1247 GRAP, Amiens, France
| | - Mickael Naassila
- Université de Picardie Jules Verne, Inserm UMR1247 GRAP, Amiens, France
| | | |
Collapse
|
30
|
Davis CN, Dash GF, Miller MB, Slutske WS. Past year high-intensity drinking moderates the association between simultaneous alcohol and marijuana use and blackout frequency among college students. JOURNAL OF AMERICAN COLLEGE HEALTH : J OF ACH 2023; 71:140-146. [PMID: 33577429 PMCID: PMC8357845 DOI: 10.1080/07448481.2021.1880415] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/15/2020] [Accepted: 01/15/2021] [Indexed: 06/12/2023]
Abstract
Objective: The role of simultaneous alcohol and marijuana (SAM) use in the experience of blackouts among college students is unclear. To clarify discrepancies, the current study evaluated whether the association between SAM user status and blackouts was moderated by high-intensity drinking (HID). Participants and Methods: College students (N = 1,224; 63.7% female) reported on their past year experiences of blackout, marijuana use, SAM use, and HID (i.e., drinking at least twice the binge threshold). Results: SAM users had more past year blackouts than non-SAM users, but this effect was only significant among SAM users who had engaged in HID in the past year (nonbinge: F(5,37) = 0.50, p = 0.49; binge: F(5,138) = 0.23, p = 0.63; HID: F(5,328) = 4.52, p = 0.03). Conclusions: Effects of SAM user status on the experience of alcohol-related blackouts may be limited to individuals who engage in HID.
Collapse
Affiliation(s)
- Christal N. Davis
- Department of Psychological Sciences, University of Missouri, 210 McAlester Hall, Columbia, MO 65211, USA
| | - Genevieve F. Dash
- Department of Psychological Sciences, University of Missouri, 210 McAlester Hall, Columbia, MO 65211, USA
| | - Mary Beth Miller
- Department of Psychiatry, University of Missouri, 1 Hospital Drive DC067.00, Columbia 65212, USA
| | - Wendy S. Slutske
- Department of Psychological Sciences, University of Missouri, 210 McAlester Hall, Columbia, MO 65211, USA
| |
Collapse
|
31
|
Martelli C, Artiges E, Miranda R, Romeo B, Petillion A, Aubin HJ, Amirouche A, Chanraud S, Benyamina A, Martinot JL. Caudate gray matter volumes and risk of relapse in Type A alcohol-dependent patients: A 7-year MRI follow-up study. Front Psychiatry 2023; 14:1067326. [PMID: 36873223 PMCID: PMC9975333 DOI: 10.3389/fpsyt.2023.1067326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/20/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Whether alteration in regional brain volumes can be detected in Type A alcoholics both at baseline and after a long follow-up remains to be confirmed. Therefore, we examined volume alterations at baseline, and longitudinal changes in a small follow-up subsample. METHODS In total of 26 patients and 24 healthy controls were assessed at baseline using magnetic resonance imaging and voxel-based morphometry, among which 17 patients and 6 controls were re-evaluated 7 years later. At baseline, regional cerebral volumes of patients were compared to controls. At follow-up, three groups were compared: abstainers (n = 11, more than 2 years of abstinence), relapsers (n = 6, <2 years of abstinence), and controls (n = 6). RESULTS The cross-sectional analyses detected, at both times, higher caudate nuclei volumes bilaterally in relapsers compared to abstainers. In abstainers, the longitudinal analysis indicated recovery of normal gray matter volumes in the middle and inferior frontal gyrus, and in the middle cingulate, while white matter volumes recovery was detected in the corpus callosum and in anterior and superior white matter specific regions. CONCLUSIONS Overall, the present investigation revealed larger caudate nuclei in the relapser AUD patient group both at baseline and at follow-up in the cross-sectional analyses. This finding suggest that a higher caudate volume could be a candidate risk factor of relapse. In patients with specific type A alcohol-dependence, we showed that long-term recovery in fronto-striato-limbic GM and WM volumes occurs during long-term abstinence. These results support the crucial role of frontal circuitry in AUD.
Collapse
Affiliation(s)
- Catherine Martelli
- Institut National de la Santé et de la Recherche Médicale (INSERM) Research Unit 1299 "Trajectoires développementales en psychiatrie", École Normale Supérieure Paris-Saclay, Université Paris-Saclay, Centre National de la Recherche Scientifique (CNRS) 9010, Centre Borelli, Gif-sur-Yvette, France.,Department of Psychiatry and Addictology, Assistance Publique - Hôpitaux de Paris, Paul-Brousse Hospital, Villejuif, France.,Psychiatry-Comorbidities-Addictions Research Unit (PSYCOMADD), Paris-Saclay University, Gif-sur-Yvette, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale (INSERM) Research Unit 1299 "Trajectoires développementales en psychiatrie", École Normale Supérieure Paris-Saclay, Université Paris-Saclay, Centre National de la Recherche Scientifique (CNRS) 9010, Centre Borelli, Gif-sur-Yvette, France.,Department of Psychiatry, Établissement Public de Santé (EPS) Barthélemy Durand, Etampes, France
| | - Rubén Miranda
- Institut National de la Santé et de la Recherche Médicale (INSERM) Research Unit 1299 "Trajectoires développementales en psychiatrie", École Normale Supérieure Paris-Saclay, Université Paris-Saclay, Centre National de la Recherche Scientifique (CNRS) 9010, Centre Borelli, Gif-sur-Yvette, France.,Department of Psychiatry and Addictology, Assistance Publique - Hôpitaux de Paris, Paul-Brousse Hospital, Villejuif, France.,Psychiatry-Comorbidities-Addictions Research Unit (PSYCOMADD), Paris-Saclay University, Gif-sur-Yvette, France
| | - Bruno Romeo
- Department of Psychiatry and Addictology, Assistance Publique - Hôpitaux de Paris, Paul-Brousse Hospital, Villejuif, France.,Psychiatry-Comorbidities-Addictions Research Unit (PSYCOMADD), Paris-Saclay University, Gif-sur-Yvette, France
| | - Amélie Petillion
- Department of Psychiatry and Addictology, Assistance Publique - Hôpitaux de Paris, Paul-Brousse Hospital, Villejuif, France.,Psychiatry-Comorbidities-Addictions Research Unit (PSYCOMADD), Paris-Saclay University, Gif-sur-Yvette, France
| | - Henri-Jean Aubin
- Department of Psychiatry and Addictology, Assistance Publique - Hôpitaux de Paris, Paul-Brousse Hospital, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale Research Unit 1018, Centre de Recherche en Epidémiologie et Santé des Populations (CESP), Paris, France
| | - Ammar Amirouche
- Department of Psychiatry and Addictology, Assistance Publique - Hôpitaux de Paris, Paul-Brousse Hospital, Villejuif, France.,Psychiatry-Comorbidities-Addictions Research Unit (PSYCOMADD), Paris-Saclay University, Gif-sur-Yvette, France
| | - Sandra Chanraud
- Paris Sciences & Lettres (PSL) Research University-École Pratique des Hautes Études (EPHE), Paris, France.,Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 5287, University of Bordeaux, Bordeaux, France
| | - Amine Benyamina
- Department of Psychiatry and Addictology, Assistance Publique - Hôpitaux de Paris, Paul-Brousse Hospital, Villejuif, France.,Psychiatry-Comorbidities-Addictions Research Unit (PSYCOMADD), Paris-Saclay University, Gif-sur-Yvette, France
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale (INSERM) Research Unit 1299 "Trajectoires développementales en psychiatrie", École Normale Supérieure Paris-Saclay, Université Paris-Saclay, Centre National de la Recherche Scientifique (CNRS) 9010, Centre Borelli, Gif-sur-Yvette, France
| |
Collapse
|
32
|
Vulser H, Lemaître HS, Guldner S, Bezivin-Frère P, Löffler M, Sarvasmaa AS, Massicotte-Marquez J, Artiges E, Paillère Martinot ML, Filippi I, Miranda R, Stringaris A, van Noort BM, Penttilä J, Grimmer Y, Becker A, Banaschewski T, Bokde ALW, Desrivières S, Fröhner JH, Garavan H, Grigis A, Gowland PA, Heinz A, Papadopoulos Orfanos D, Poustka L, Smolka MN, Spechler PA, Walter H, Whelan R, Schumann G, Flor H, Martinot JL, Nees F. Chronotype, Longitudinal Volumetric Brain Variations Throughout Adolescence, and Depressive Symptom Development. J Am Acad Child Adolesc Psychiatry 2023; 62:48-58. [PMID: 35714839 DOI: 10.1016/j.jaac.2022.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/04/2022] [Accepted: 06/03/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Adolescence is a critical period for circadian rhythm, with a strong shift toward eveningness around age 14. Also, eveningness in adolescence has been found to predict later onset of depressive symptoms. However, no previous study has investigated structural variations associated with chronotype in early adolescence and how this adds to the development of depressive symptoms. METHOD Assessment of 128 community-based adolescents (51% girls) at age 14 and 19 years was performed. Using whole-brain voxel-based morphometry, baseline (at age 14) regional gray matter volumes (GMVs), follow-up (at age 19) regional GMVs, and longitudinal changes (between 14 and 19) associated with Morningness/Eveningness Scale in Children score and sleep habits at baseline were measured. The association of GMV with depressive symptoms at 19 years was studied, and the role of potential clinical and genetic factors as mediators and moderators was assessed. RESULTS Higher eveningness was associated with larger GMV in the right medial prefrontal cortex at ages 14 and 19 in the whole sample. GMV in this region related to depressive symptoms at age 19 in catechol-O-methyltransferase (COMT) Val/Val, but not in Met COMT, carriers. Larger GMV also was observed in the right fusiform gyrus at age 14, which was explained by later wake-up time during weekends. CONCLUSION In adolescence, eveningness and its related sleep habits correlated with distinct developmental patterns. Eveningness was specifically associated with GMV changes in the medial prefrontal cortex; this could serve as a brain vulnerability factor for later self-reported depressive symptoms in COMT Val/Val carriers.
Collapse
Affiliation(s)
- Hélène Vulser
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Germany; AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, France.
| | - Hervé S Lemaître
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, Bordeaux, France
| | - Stella Guldner
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Germany
| | - Pauline Bezivin-Frère
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299 A10 "Trajectoires développementales en psychiatrie," Ecole Normale supérieure Paris-Saclay, Université Paris-Saclay, Université de Paris, CNRS UMR9010, Centre Borelli, Gif-sur-Yvette, France
| | - Martin Löffler
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Germany
| | - Anna S Sarvasmaa
- National Institute for Health and Welfare, Mental Health Unit, Helsinki, Finland, and the University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jessica Massicotte-Marquez
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299 A10 "Trajectoires développementales en psychiatrie," Ecole Normale supérieure Paris-Saclay, Université Paris-Saclay, Université de Paris, CNRS UMR9010, Centre Borelli, Gif-sur-Yvette, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299 A10 "Trajectoires développementales en psychiatrie," Ecole Normale supérieure Paris-Saclay, Université Paris-Saclay, Université de Paris, CNRS UMR9010, Centre Borelli, Gif-sur-Yvette, France; EPS Barthélémy Durand, Etampes, France
| | - Marie-Laure Paillère Martinot
- AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, France; Institut National de la Santé et de la Recherche Médicale, INSERM U1299 A10 "Trajectoires développementales en psychiatrie," Ecole Normale supérieure Paris-Saclay, Université Paris-Saclay, Université de Paris, CNRS UMR9010, Centre Borelli, Gif-sur-Yvette, France
| | - Irina Filippi
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299 A10 "Trajectoires développementales en psychiatrie," Ecole Normale supérieure Paris-Saclay, Université Paris-Saclay, Université de Paris, CNRS UMR9010, Centre Borelli, Gif-sur-Yvette, France
| | - Ruben Miranda
- AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, France
| | | | | | - Jani Penttilä
- Psychosocial Services Adolescent Outpatient Clinic, Lahti, Finland
| | - Yvonne Grimmer
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Tobias Banaschewski
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Arun L W Bokde
- School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, London, United Kingdom
| | | | | | - Antoine Grigis
- NeuroSpin, Commissariat à l'Energie Atomique, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Penny A Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Andreas Heinz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Luise Poustka
- University Medical Centre Göttingen, Göttingen, Germany
| | | | | | - Henrik Walter
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Ireland
| | - Gunter Schumann
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, London, United Kingdom, the PONS Research Group, Campus Charite Mitte, Humboldt University, Berlin, Leibniz Institute for Neurobiology, Magdeburg, Germany and the Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, P.R. China
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Germany; School of Social Sciences, University of Mannheim, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299 A10 "Trajectoires développementales en psychiatrie," Ecole Normale supérieure Paris-Saclay, Université Paris-Saclay, Université de Paris, CNRS UMR9010, Centre Borelli, Gif-sur-Yvette, France
| | - Frauke Nees
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Germany; Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig Holstein, Kiel University, Kiel, Germany, and the Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Germany
| |
Collapse
|
33
|
Obray JD, Landin JD, Vaughan DT, Scofield MD, Chandler LJ. Adolescent alcohol exposure reduces dopamine 1 receptor modulation of prelimbic neurons projecting to the nucleus accumbens and basolateral amygdala. ADDICTION NEUROSCIENCE 2022; 4:100044. [PMID: 36643604 PMCID: PMC9836047 DOI: 10.1016/j.addicn.2022.100044] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Binge drinking during adolescence is highly prevalent despite increasing evidence of its long-term impact on behaviors associated with modulation of behavioral flexibility by the medial prefrontal cortex (mPFC). In the present study, male and female rats underwent adolescent intermittent ethanol (AIE) exposure by vapor inhalation. After aging to adulthood, retrograde bead labelling and viral tagging were used to identify populations of neurons in the prelimbic region (PrL) of the mPFC that project to specific subcortical targets. Electrophysiological recording from bead-labelled neurons in PrL slices revealed that AIE did not alter the intrinsic excitability of PrL neurons that projected to either the NAc or the BLA. Similarly, recordings of spontaneous inhibitory and excitatory post-synaptic currents revealed no AIE-induced changes in synaptic drive onto either population of projection neurons. In contrast, AIE exposure was associated with a loss of dopamine receptor 1 (D1), but no change in dopamine receptor 2 (D2), modulation of evoked firing of both populations of projection neurons. Lastly, confocal imaging of proximal and apical dendritic tufts of viral-labelled PrL neurons that projected to the nucleus accumbens (NAc) revealed AIE did not alter the density of dendritic spines. Together, these observations provide evidence that AIE exposure results in disruption of D1 receptor modulation of PrL inputs to at least two major subcortical target regions that have been implicated in AIE-induced long-term changes in behavioral control.
Collapse
Affiliation(s)
- J. Daniel Obray
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA
| | - Justine D. Landin
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA
| | - Dylan T. Vaughan
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA
| | - Michael D. Scofield
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA,Department of Anesthesiology, Medical University of South Carolina, Charleston SC, USA
| | - L. Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA,Corresponding author. (L.J. Chandler)
| |
Collapse
|
34
|
Healey KL, Bell A, Scofield MD, Swartzwelder H. Adolescent intermittent ethanol exposure reduces astrocyte-synaptic proximity in the adult medial prefrontal cortex in rats: Reversal by gabapentin. ADDICTION NEUROSCIENCE 2022; 4:100047. [PMID: 36643603 PMCID: PMC9836051 DOI: 10.1016/j.addicn.2022.100047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Alcohol consumption in adolescence causes multiple acute negative changes in neural and behavioral function that persist well into adulthood and possibly throughout life. The medial prefrontal cortex (mPFC) and dorsal hippocampus are critical for executive function and memory and are especially vulnerable to adolescent ethanol exposure. We have reported that astrocytes, particularly in the mPFC, change both in morphology and synaptic proximity during adolescence. Moreover, adolescent intermittent ethanol (AIE) exposure produces enduring effects on both astrocyte function and synaptic proximity in the adult hippocampal formation, and the latter effect was reversed by the clinically used agent gabapentin (Neurontin), an anticonvulsant and analgesic that is an inhibitor of the VGCC α2δ1 subunit. These findings underscore the importance of investigating AIE effects on astrocytes in the mPFC, a region that undergoes marked changes in structure and connectivity during adolescence. Using astrocyte-specific viral labeling and immunohistochemistry, mPFC astrocytic morphology and colocalization with AMPA-(α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) glutamate receptor 1 (GluA1), an AMPA receptor subunit and established neuronal marker of excitatory synapses, were assessed to quantify the proximity of astrocyte processes with glutamatergic synaptic puncta. AIE exposure significantly reduced astrocyte-synaptic proximity in adulthood, an effect that was reversed by sub-chronic gabapentin treatment in adulthood. There was no effect of AIE on astrocytic glutamate homeostasis machinery or neuronal synaptic proteins in the mPFC. These findings indicate a possible glial-neuronal mechanism underlying the effects of AIE on frontal lobe-mediated behaviors and suggest a specific therapeutic approach for the amelioration of those effects.
Collapse
Affiliation(s)
- Kati L. Healey
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, N.C. 27710, United States of America
| | - Amelia Bell
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, N.C. 27710, United States of America
| | - Michael D. Scofield
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, S.C. 29425, United States of America
| | - H.S. Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, N.C. 27710, United States of America
| |
Collapse
|
35
|
Davis CN, Gizer IR, Colodro-Conde L, Statham DJ, Martin NG, Slutske WS. Educational Attainment Polygenic Scores: Examining Evidence for Gene-Environment Interplay with Adolescent Alcohol, Tobacco and Cannabis Use. Twin Res Hum Genet 2022; 25:187-195. [PMID: 36189823 DOI: 10.1017/thg.2022.33] [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] [Indexed: 11/07/2022]
Abstract
Genes associated with educational attainment may be related to or interact with adolescent alcohol, tobacco and cannabis use. Potential gene-environment interplay between educational attainment polygenic scores (EA-PGS) and adolescent alcohol, tobacco, and cannabis use was evaluated with a series of regression models fitted to data from a sample of 1871 adult Australian twins. All models controlled for age, age2, cohort, sex and genetic ancestry as fixed effects, and a genetic relatedness matrix was included as a random effect. Although there was no evidence that adolescent alcohol, tobacco or cannabis use interacted with EA-PGS to influence educational attainment, there was a significant, positive gene-environment correlation with adolescent alcohol use at all PGS thresholds (ps <.02). Higher EA-PGS were associated with an increased likelihood of using alcohol as an adolescent (ΔR2 ranged from 0.5% to 1.1%). The positive gene-environment correlation suggests a complex relationship between educational attainment and alcohol use that is due to common genetic factors.
Collapse
Affiliation(s)
- Christal N Davis
- Department of Psychological Sciences, University of Missouri, Columbia, MO65211, USA
| | - Ian R Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO65211, USA
| | - Lucía Colodro-Conde
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland4006, Australia
| | | | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland4006, Australia
| | - Wendy S Slutske
- Center for Tobacco Research and Intervention and Department of Family Medicine and Community Health, University of Wisconsin School of Medicine and Public Health, Madison, WI53711, USA
| |
Collapse
|
36
|
Quijano Cardé NA, Shaw J, Carter C, Kim S, Stitzel JA, Venkatesh SK, Ramchandani VA, De Biasi M. Mutation of the α5 nicotinic acetylcholine receptor subunit increases ethanol and nicotine consumption in adolescence and impacts adult drug consumption. Neuropharmacology 2022; 216:109170. [PMID: 35752273 PMCID: PMC9308728 DOI: 10.1016/j.neuropharm.2022.109170] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/16/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
Alcohol and nicotine are commonly used during adolescence, establishing long-lasting neuroplastic alterations that influence subsequent drug use and abuse. Drinking- and smoking-related traits have been extensively associated with variation in CHRNA5 - the gene that encodes the α5 subunit of neuronal nicotinic acetylcholine receptors (nAChRs). The single nucleotide polymorphism (SNP) rs16969968 in CHRNA5 encodes an amino acid substitution (D398N) that alters the function and pharmacokinetics of α5-containing nAChR. When expressed in rodents, this variant results in increased ethanol and nicotine operant self-administration. How disruption of α5-containing nAChRs influences adolescent ethanol and nicotine intake, and how it modulates interactions between these drugs has not been previously explored. In the present study, we examined volitional ethanol and nicotine consumption in adolescent mice (post-natal day 30-43) of both sexes with mutated (SNP) or lacking (KO) the α5 nAChR subunit. The effect of adolescent alcohol or nicotine exposure on home cage consumption of the opposite drug in adulthood and its modulation by Chrna5 mutation and sex were examined. During adolescence, we found that α5 nAChR disruption increases nicotine intake in mice of both sexes, but the effect on alcohol intake was only observed in females. The sex-specific increase in alcohol consumption in α5 SNP and KO was replicated in adulthood. The effect of adolescent alcohol or nicotine exposure on subsequent intake of the opposite drug in adulthood is modulated by sex and Chrna5 mutation. These observations suggest sex differences in the genetic architecture of alcohol dependence, and modulators of alcohol and nicotine interactions.
Collapse
Affiliation(s)
- Natalia A Quijano Cardé
- Pharmacology Graduate Group, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jessica Shaw
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Christina Carter
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Seung Kim
- Neuroscience Program, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Shyamala K Venkatesh
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Laboratory of Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Vijay A Ramchandani
- Laboratory of Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Mariella De Biasi
- Pharmacology Graduate Group, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Dunn ME, Schreiner AM, Flori JN, Crisafulli MJ, Willis EA, Lynch GT, Leary AV, Dvorak RD. Effective prevention programming for reducing alcohol-related harms experienced by first year college students: Evaluation of the expectancy challenge alcohol literacy curriculum (ECALC). Addict Behav 2022; 131:107338. [PMID: 35472696 PMCID: PMC10349386 DOI: 10.1016/j.addbeh.2022.107338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/01/2022]
Abstract
Prevention programs may have contributed to modest declines in alcohol use among college students in recent years, but negative consequences continue to be pervasive. First year college students (FYCS) are particularly vulnerable, and there is clearly a need for more effective methods to reduce risk. Meta-analyses focused on expectancy challenge (EC) have found this approach to be effective, but "experiential" EC that includes a drinking exercise is not suitable for most FYCS, many of whom are underage. A non-experiential alternative, the Expectancy Challenge Alcohol Literacy Curriculum (ECALC), is practical for widespread implementation. ECALC has been effective with mandated students and members of fraternities, and in the present study, we focused on evaluating effects with FYCS. In a group randomized trial, 48 class sections of a course designed for FYCS received either ECALC or an attention-matched control presentation. ECALC was associated with significant changes on six expectancy subscales of the Comprehensive Effects of Alcohol Scale (CEOA). Structural equation modeling was used to examine the mediated effects of the intervention on alcohol-related harms via alcohol expectancies. There were significant indirect effects from condition to alcohol use (IND = -0.04, p <.001) and alcohol harms (IND = -0.07, p <.001). This model accounted for 54% of the variance in alcohol use and 46% of the variance in alcohol-related harms. These findings suggest ECALC is an effective, single session group-delivered program that can be incorporated into classroom curricula.
Collapse
|
38
|
Family Structure, Unstructured Socializing, and Heavy Substance Use among Adolescents. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19148818. [PMID: 35886673 PMCID: PMC9317110 DOI: 10.3390/ijerph19148818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 12/10/2022]
Abstract
Background: Psychoactive substance use is a transient behavior among many adolescents and diminishes as they mature, but some engage in heavy forms of substance use, which increases their risk of health and behavioral challenges. A consistent predictor of substance use among youth is family structure, with adolescents living in single-parent, stepparent, or no-parent families at higher risk than others of several forms of substance use. The objective of this research was to investigate whether unstructured socializing mediated the association between family structure and heavy alcohol or substance use. Methods: Data from 30 nations (n = 65,737) were used to test the hypothesis using a generalized structural equation model and tests of mediation. Results: The analysis furnished clear support for a mediation effect among adolescents living with a single parent but less support among those living with a stepparent or neither parent. Conclusion: The association between living in a single-parent household and heavy alcohol or other substance use was mediated largely by time spent outside the home with friends in unsupervised activities. Additional research that uses longitudinal data and more nuanced measures of family structure is needed to validate this finding.
Collapse
|
39
|
Pérez-García JM, Cadaveira F, Canales-Rodríguez EJ, Suárez-Suárez S, Rodríguez Holguín S, Corral M, Blanco-Ramos J, Doallo S. Effects of Persistent Binge Drinking on Brain Structure in Emerging Adults: A Longitudinal Study. Front Psychiatry 2022; 13:935043. [PMID: 35815019 PMCID: PMC9260041 DOI: 10.3389/fpsyt.2022.935043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
Previous cross-sectional research has largely associated binge drinking (BD) with changes in volume and thickness during adolescence and early adulthood. Nevertheless, the long-term alcohol-related effects on gray matter features in youths who had maintained a BD pattern over time have not yet been sufficiently explored. The present study aimed to assess group differences both cross-sectionally and longitudinally [using symmetric percent change (SPC)] on several structural measures (i.e., thickness, surface area, volume). For this purpose, magnetic resonance imaging was recorded twice within a 2-year interval; at baseline (18-19 years) and a follow-up (20-21 years). The sample included 44 university students who were classified as 16 stable binge drinkers (8 females) and 28 stable controls (13 females). Whole-brain analysis showed larger insular surface area in binge drinkers relative to controls at follow-up (cluster-wise p = 0.045). On the other hand, region of interest (ROI) analyses on thickness also revealed a group by sex interaction at follow-up (p = 0.005), indicating that BD males had smaller right rostral middle frontal gyrus thickness than both control males (p = 0.011) and BD females (p = 0.029). Similarly, ROI-based analysis on longitudinal data showed a group by sex interaction in the right nucleus accumbens (p = 0.009) which revealed a decreased volume across time in BD males than in control males (p = 0.007). Overall, continued BD pattern during emerging adulthood appears to lead to gray matter abnormalities in regions intimately involved in reward processing, emotional regulation and executive functions. Notably, some anomalies varied significantly depending on sex, suggesting a sex-specific impact of BD on typical neurodevelopment processes.
Collapse
Affiliation(s)
- Jose Manuel Pérez-García
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Fernando Cadaveira
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Erick J. Canales-Rodríguez
- Signal Processing Laboratory 5 (LTS5), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- FIDMAG Germanes Hospitalàries Research Foundation, Sant Boi de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Samuel Suárez-Suárez
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Socorro Rodríguez Holguín
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Montserrat Corral
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Javier Blanco-Ramos
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Sonia Doallo
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| |
Collapse
|
40
|
Pérez-García JM, Suárez-Suárez S, Doallo S, Cadaveira F. Effects of binge drinking during adolescence and emerging adulthood on the brain: A systematic review of neuroimaging studies. Neurosci Biobehav Rev 2022; 137:104637. [PMID: 35339481 DOI: 10.1016/j.neubiorev.2022.104637] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 01/19/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
Binge drinking (BD) is a common pattern of alcohol consumption which is generating great concern because of its deleterious consequences. We selected 33 neuroimaging studies of healthy young binge drinkers (BDs) by following PRISMA guidelines. This review provides a comprehensive overview of the relationship between BD and neurocognitive anomalies reported across magnetic resonance studies. Moreover, this work is the first in which results of relatively new imaging techniques, such as resting-state functional connectivity (RS-FC) and neurite orientation dispersion and density imaging (NODDI), have been reviewed using a systematic procedure. We established strict inclusion criteria in order to isolate the various potential effects of BD on the adolescent brain. Two authors independently evaluated the methodological quality, assessing different aspects related to sample size, and statistical correction methods, which are of particular importance in neuroimaging studies. BD is associated with structural and functional anomalies in several cortical and subcortical brain regions intimately involved in the control and regulation of impulsive or risky behaviours, as well as in the processing of reinforcing stimuli.
Collapse
Affiliation(s)
- Jose Manuel Pérez-García
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain.
| | - Samuel Suárez-Suárez
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain.
| | - Sonia Doallo
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain.
| | - Fernando Cadaveira
- Department of Clinical Psychology and Psychobiology, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain.
| |
Collapse
|
41
|
Rane RP, de Man EF, Kim J, Görgen K, Tschorn M, Rapp MA, Banaschewski T, Bokde ALW, Desrivieres S, Flor H, Grigis A, Garavan H, Gowland PA, Brühl R, Martinot JL, Martinot MLP, Artiges E, Nees F, Papadopoulos Orfanos D, Lemaitre H, Paus T, Poustka L, Fröhner J, Robinson L, Smolka MN, Winterer J, Whelan R, Schumann G, Walter H, Heinz A, Ritter K. Structural differences in adolescent brains can predict alcohol misuse. eLife 2022; 11:e77545. [PMID: 35616520 PMCID: PMC9255959 DOI: 10.7554/elife.77545] [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: 02/02/2022] [Accepted: 05/25/2022] [Indexed: 12/02/2022] Open
Abstract
Alcohol misuse during adolescence (AAM) has been associated with disruptive development of adolescent brains. In this longitudinal machine learning (ML) study, we could predict AAM significantly from brain structure (T1-weighted imaging and DTI) with accuracies of 73 -78% in the IMAGEN dataset (n∼1182). Our results not only show that structural differences in brain can predict AAM, but also suggests that such differences might precede AAM behavior in the data. We predicted 10 phenotypes of AAM at age 22 using brain MRI features at ages 14, 19, and 22. Binge drinking was found to be the most predictable phenotype. The most informative brain features were located in the ventricular CSF, and in white matter tracts of the corpus callosum, internal capsule, and brain stem. In the cortex, they were spread across the occipital, frontal, and temporal lobes and in the cingulate cortex. We also experimented with four different ML models and several confound control techniques. Support Vector Machine (SVM) with rbf kernel and Gradient Boosting consistently performed better than the linear models, linear SVM and Logistic Regression. Our study also demonstrates how the choice of the predicted phenotype, ML model, and confound correction technique are all crucial decisions in an explorative ML study analyzing psychiatric disorders with small effect sizes such as AAM.
Collapse
Affiliation(s)
- Roshan Prakash Rane
- Charité – Universitätsmedizin Berlin (corporate member of Freie Universiät at Berlin, Humboldt-Universiät at zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Psychotherapy, Bernstein Center for Computational NeuroscienceBerlinGermany
| | - Evert Ferdinand de Man
- Faculty IV – Electrical Engineering and Computer Science, Technische Universität BerlinBerlinGermany
| | - JiHoon Kim
- Department of Education and Psychology, Freie Universität BerlinBerlinGermany
| | - Kai Görgen
- Charité – Universitätsmedizin Berlin (corporate member of Freie Universiät at Berlin, Humboldt-Universiät at zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Psychotherapy, Bernstein Center for Computational NeuroscienceBerlinGermany
- Science of Intelligence, Research Cluster of ExcellenceBerlinGermany
| | - Mira Tschorn
- Social and Preventive Medicine, Department of Sports and Health Sciences, Intra-faculty unit “Cognitive Sciences”, Faculty of Human Science, and Faculty of Health Sciences Brandenburg, Research Area Services Research and e-Health, University of PotsdamPotsdamGermany
| | - Michael A Rapp
- Social and Preventive Medicine, Department of Sports and Health Sciences, Intra-faculty unit “Cognitive Sciences”, Faculty of Human Science, and Faculty of Health Sciences Brandenburg, Research Area Services Research and e-Health, University of PotsdamPotsdamGermany
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg UniversityMannheimGermany
| | - Arun LW Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College DublinDublinIreland
| | - Sylvane Desrivieres
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology Neuroscience SGDP Centre, King’s College LondonLondonUnited Kingdom
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg UniversityHeidelbergGermany
- Department of Psychology, School of Social Sciences, University of MannheimMannheimGermany
| | | | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of VermontBurlingtonUnited States
| | - Penny A Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of NottinghamNottinghamUnited Kingdom
| | | | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 ”Trajectoires développementales en psychiatrie” Universite Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, Centre BorelliGif-sur-YvetteFrance
| | - Marie-Laure Paillere Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 ”Trajectoires développementales en psychiatrie” Universite Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, Centre BorelliGif-sur-YvetteFrance
- AP-HP Sorbonne Université, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière HospitalParisFrance
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 ”Trajectoires développementales en psychiatrie” Universite Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, Centre BorelliGif-sur-YvetteFrance
- Psychiatry Department, EPS Barthélémy DurandEtampesFrance
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg UniversityMannheimGermany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg UniversityHeidelbergGermany
- PONS Research Group, Dept of Psychiatry and Psychotherapy, Campus Charite Mitte, Humboldt UniversityBerlinGermany
| | | | - Herve Lemaitre
- NeuroSpin, CEA, Université Paris-SaclayParisFrance
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, University of BordeauxBordeauxFrance
| | - Tomas Paus
- Department of Psychiatry, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of MontrealMontrealCanada
- Departments of Psychiatry and Psychology, University of TorontoTorontoCanada
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre GöttingenGöttingenGermany
| | - Juliane Fröhner
- Department of Psychiatry and Neuroimaging Center, Technische Universität DresdenDresdenGermany
| | - Lauren Robinson
- Department of Psychological Medicine, Section for Eating Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College LondonLondonUnited Kingdom
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität DresdenDresdenGermany
| | - Jeanne Winterer
- Charité – Universitätsmedizin Berlin (corporate member of Freie Universiät at Berlin, Humboldt-Universiät at zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Psychotherapy, Bernstein Center for Computational NeuroscienceBerlinGermany
- Department of Education and Psychology, Freie Universität BerlinBerlinGermany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College DublinDublinIreland
| | - Gunter Schumann
- PONS Research Group, Dept of Psychiatry and Psychotherapy, Campus Charite Mitte, Humboldt UniversityBerlinGermany
| | - Henrik Walter
- Charité – Universitätsmedizin Berlin (corporate member of Freie Universiät at Berlin, Humboldt-Universiät at zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Psychotherapy, Bernstein Center for Computational NeuroscienceBerlinGermany
| | - Andreas Heinz
- Charité – Universitätsmedizin Berlin (corporate member of Freie Universiät at Berlin, Humboldt-Universiät at zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Psychotherapy, Bernstein Center for Computational NeuroscienceBerlinGermany
| | - Kerstin Ritter
- Charité – Universitätsmedizin Berlin (corporate member of Freie Universiät at Berlin, Humboldt-Universiät at zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Psychotherapy, Bernstein Center for Computational NeuroscienceBerlinGermany
| | | |
Collapse
|
42
|
Machado do Vale TC, da Silva Chagas L, de Souza Pereira H, Giestal-de-Araujo E, Arévalo A, Oliveira-Silva Bomfim P. Neuroscience Outside the Box: From the Laboratory to Discussing Drug Abuse at Schools. Front Hum Neurosci 2022; 16:782205. [PMID: 35634202 PMCID: PMC9133440 DOI: 10.3389/fnhum.2022.782205] [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: 09/24/2021] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
One of the effects of the current COVID-19 pandemic is that low-income countries were pushed further into extreme poverty, exacerbating social inequalities and increasing susceptibility to drug use/abuse in people of all ages. The risks of drug abuse may not be fully understood by all members of society, partly because of the taboo nature of the subject, and partly because of the considerable gap between scientific production/understanding and communication of such knowledge to the public at large. Drug use is a major challenge to social development and a leading cause of school dropout rates worldwide. Some public policies adopted in several countries in recent decades failed to prevent drug use, especially because they focused on imposing combative or coercive measures, investing little or nothing in education and prevention. Here we highlight the role of neuroscience education as a valid approach in drug use education and prevention. We propose building a bridge between schools and scientists by promoting information, student engagement and honest dialogue, and show evidence that public policy regulators should be persuaded to support such science-based education programs in their efforts to effect important positive changes in society.
Collapse
Affiliation(s)
- Thereza Cristina Machado do Vale
- NuPEDEN, Nucleus for Research, Education, Dissemination and Neurosciences Popularization, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, Brazil
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, Brazil
| | - Luana da Silva Chagas
- NuPEDEN, Nucleus for Research, Education, Dissemination and Neurosciences Popularization, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, Brazil
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, Brazil
| | - Helena de Souza Pereira
- Department of Molecular and Cell Biology, Institute of Biology, Federal Fluminense University, Niterói, Brazil
| | - Elizabeth Giestal-de-Araujo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, Brazil
| | - Analía Arévalo
- Department of Experimental Surgery, University of São Paulo Medical School, São Paulo, Brazil
| | - Priscilla Oliveira-Silva Bomfim
- NuPEDEN, Nucleus for Research, Education, Dissemination and Neurosciences Popularization, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, Brazil
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, Brazil
- *Correspondence: Priscilla Oliveira-Silva Bomfim,
| |
Collapse
|
43
|
Abstract
This article is part of a Festschrift commemorating the 50th anniversary of the National Institute on Alcohol Abuse and Alcoholism (NIAAA). Established in 1970, first as part of the National Institute of Mental Health and later as an independent institute of the National Institutes of Health, NIAAA today is the world's largest funding agency for alcohol research. In addition to its own intramural research program, NIAAA supports the entire spectrum of innovative basic, translational, and clinical research to advance the diagnosis, prevention, and treatment of alcohol use disorder and alcohol-related problems. To celebrate the anniversary, NIAAA hosted a 2-day symposium, "Alcohol Across the Lifespan: 50 Years of Evidence-Based Diagnosis, Prevention, and Treatment Research," devoted to key topics within the field of alcohol research. This article is based on Dr. Tapert's presentation at the event. NIAAA Director George F. Koob, Ph.D., serves as editor of the Festschrift.
Collapse
Affiliation(s)
- Susan F Tapert
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | | |
Collapse
|
44
|
Lodha J, Brocato E, Wolstenholme JT. Areas of Convergence and Divergence in Adolescent Social Isolation and Binge Drinking: A Review. Front Behav Neurosci 2022; 16:859239. [PMID: 35431830 PMCID: PMC9009335 DOI: 10.3389/fnbeh.2022.859239] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Adolescence is a critical developmental period characterized by enhanced social interactions, ongoing development of the frontal cortex and maturation of synaptic connections throughout the brain. Adolescents spend more time interacting with peers than any other age group and display heightened reward sensitivity, impulsivity and diminished inhibitory self-control, which contribute to increased risky behaviors, including the initiation and progression of alcohol use. Compared to adults, adolescents are less susceptible to the negative effects of ethanol, but are more susceptible to the negative effects of stress, particularly social stress. Juvenile exposure to social isolation or binge ethanol disrupts synaptic connections, dendritic spine morphology, and myelin remodeling in the frontal cortex. These structural effects may underlie the behavioral and cognitive deficits seen later in life, including social and memory deficits, increased anxiety-like behavior and risk for alcohol use disorders (AUD). Although the alcohol and social stress fields are actively investigating the mechanisms through which these effects occur, significant gaps in our understanding exist, particularly in the intersection of the two fields. This review will highlight the areas of convergence and divergence in the fields of adolescent social stress and ethanol exposure. We will focus on how ethanol exposure or social isolation stress can impact the development of the frontal cortex and lead to lasting behavioral changes in adulthood. We call attention to the need for more mechanistic studies and the inclusion of the evaluation of sex differences in these molecular, structural, and behavioral responses.
Collapse
Affiliation(s)
- Jyoti Lodha
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Emily Brocato
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T. Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
45
|
Cao R, Gao T, Ren H, Hu Y, Qin Z, Liang L, Li C, Mei S. Unique and cumulative effects of lifestyle-related behaviors on depressive symptoms among Chinese adolescents. Int J Soc Psychiatry 2022; 68:354-364. [PMID: 33622084 DOI: 10.1177/0020764021996739] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND It is well known that some lifestyle-related behaviors are related to depressive symptoms, but the unique and cumulative effects of lifestyle-related behaviors on depressive symptoms among Chinese adolescents are still controversial. AIMS The aims of this study were to examine the unique and cumulative effects of lifestyle-related behaviors on depressive symptoms among Chinese adolescents, and explored the potential influences of gender difference on these associations. METHODS We conducted a cross-sectional study among 3967 Chinese adolescents aged 11 to 19 from Jilin, China during September and October of 2018. Students reported their lifestyle factors including sleep duration, time spent on computer, time spent on television, time spent on homework, eating breakfast, smoking, drinking, physical activity, and outdoor activity. Depressive symptoms were measured using the Center for Epidemiologic Studies Depression Scale (CES-D). RESULTS The prevalence of depressive symptoms was 28.2% among Chinese adolescents. Multivariate logistic regression analysis revealed that sleep duration <8 hour/day, time spent on homework ⩾3 hour/day, skipping breakfast, alcohol use, physical activity <3 days/week, and outdoor activity <2 hour/day were positively associated with depressive symptoms in both girls and boys. Time spent on computer ⩾2 hour/day was an independent risk predictor for depressive symptoms in males, while smoking only showed higher risk of depressive symptoms in females. There was an additive relationship between the lifestyle risk index scores and the risk of depressive symptoms for both genders, the relationship being strongest among females. CONCLUSION The important role of lifestyle factors should be taking into consideration when create intervention programs to prevent and reduce depressive symptoms among adolescents. In addition, preventive interventions may need to focus on gender-informed approaches when targeting multiple lifestyle factors.
Collapse
Affiliation(s)
- Ruilin Cao
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, China
| | - Tingting Gao
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, China
| | - Hui Ren
- Department of Gastrointestinal Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yueyang Hu
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, China
| | - Zeying Qin
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, China
| | - Leilei Liang
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, China
| | - Chuanen Li
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, China
| | - Songli Mei
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
46
|
Schwarz EB, Chatterton B, Fix M, Tebb K, Rodriguez F, Tancredi DJ, Muriki M, Satterfield J. Remotely Educating Young Women About Alcohol: A Randomized Trial of the PartyWise Intervention. J Womens Health (Larchmt) 2022; 31:1179-1187. [DOI: 10.1089/jwh.2021.0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Eleanor Bimla Schwarz
- Division of General Internal Medicine at San Francisco General Hospital, University of California, San Francisco, San Francisco, California, USA
| | - Brittany Chatterton
- Division of General Internal Medicine, University of California, Davis, Sacramento, California, USA
| | - Margaret Fix
- Center for Healthcare Policy and Research, University of California, Davis, Sacramento, California, USA
| | - Kathleen Tebb
- Department of Pediatrics, Division of Adolescent and Young Adult Medicine, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, California, USA
| | - Felicia Rodriguez
- Department of Pediatrics, Division of Adolescent and Young Adult Medicine, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, California, USA
| | - Daniel J. Tancredi
- Center for Healthcare Policy and Research, University of California, Davis, Sacramento, California, USA
| | - Maneesha Muriki
- Center for Healthcare Policy and Research, University of California, Davis, Sacramento, California, USA
| | - Jason Satterfield
- Division of General Internal Medicine at Mt Zion, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
47
|
Kirsch DE, Tretyak V, Le V, Huffman A, Fromme K, Strakowski SM, Lippard ET. Alcohol Use and Prefrontal Cortex Volume Trajectories in Young Adults with Mood Disorders and Associated Clinical Outcomes. Behav Sci (Basel) 2022; 12:57. [PMID: 35323376 PMCID: PMC8945008 DOI: 10.3390/bs12030057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/05/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Alcohol use in the course of mood disorders is associated with worse clinical outcomes. The mechanisms by which alcohol use alters the course of illness are unclear but may relate to prefrontal cortical (PFC) sensitivity to alcohol. We investigated associations between alcohol use and PFC structural trajectories in young adults with a mood disorder compared to typically developing peers. (2) Methods: 41 young adults (24 with a mood disorder, agemean = 21 ± 2 years) completed clinical evaluations, assessment of alcohol use, and two structural MRI scans approximately one year apart. Freesurfer was used to segment PFC regions of interest (ROIs) (anterior cingulate, orbitofrontal cortex, and frontal pole). Effects of group, alcohol use, time, and interactions among these variables on PFC ROIs at baseline and follow-up were modeled. Associations were examined between alcohol use and longitudinal changes in PFC ROIs with prospective mood. (3) Results: Greater alcohol use was prospectively associated with decreased frontal pole volume in participants with a mood disorder, but not typically developing comparison participants (time-by-group-by-alcohol interaction; p = 0.007); however, this interaction became a statistical trend in a sensitivity analysis excluding one outlier in terms of alcohol use. Greater alcohol use and a decrease in frontal pole volume related to longer duration of major depression during follow-up (p’s < 0.05). (4) Conclusion: Preliminary findings support more research on alcohol use, PFC trajectories, and depression recurrence in young adults with a mood disorder including individuals with heavier drinking patterns.
Collapse
Affiliation(s)
- Dylan E. Kirsch
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, University of Texas, Austin, TX 78712, USA; (V.T.); (V.L.); (A.H.); (S.M.S.)
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX 78712, USA;
- Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | - Valeria Tretyak
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, University of Texas, Austin, TX 78712, USA; (V.T.); (V.L.); (A.H.); (S.M.S.)
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX 78712, USA;
- Department of Psychology, University of Texas, Austin, TX 78712, USA
| | - Vanessa Le
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, University of Texas, Austin, TX 78712, USA; (V.T.); (V.L.); (A.H.); (S.M.S.)
| | - Ansley Huffman
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, University of Texas, Austin, TX 78712, USA; (V.T.); (V.L.); (A.H.); (S.M.S.)
| | - Kim Fromme
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX 78712, USA;
- Department of Psychology, University of Texas, Austin, TX 78712, USA
| | - Stephen M. Strakowski
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, University of Texas, Austin, TX 78712, USA; (V.T.); (V.L.); (A.H.); (S.M.S.)
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX 78712, USA;
- Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
- Department of Psychology, University of Texas, Austin, TX 78712, USA
| | - Elizabeth T.C. Lippard
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, University of Texas, Austin, TX 78712, USA; (V.T.); (V.L.); (A.H.); (S.M.S.)
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX 78712, USA;
- Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
- Department of Psychology, University of Texas, Austin, TX 78712, USA
- Institute of Early Life Adversity Research, University of Texas, Austin, TX 78712, USA
| |
Collapse
|
48
|
Van Hees L, Didone V, Charlet‐Briart M, Van Ingelgom T, Alexandre A, Quertemont E, Nguyen L, Laguesse S. Voluntary alcohol binge-drinking in adolescent C57Bl6 mice induces delayed appearance of behavioural defects in both males and females. Addict Biol 2022; 27:e13102. [PMID: 34611982 PMCID: PMC9285796 DOI: 10.1111/adb.13102] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/27/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022]
Abstract
Adolescence is a developmental period characterized by significant changes in brain architecture and behaviour. The immaturity of the adolescent brain is associated with heightened vulnerability to exogenous agents, including alcohol. Alcohol is the most consumed drug among teenagers, and binge‐drinking during adolescence is a major public health concern. Studies have suggested that adolescent alcohol exposure may interfere with the maturation of frontal brain regions and lead to long‐lasting behavioural consequences. In this study, by using a slightly modified version of the Drinking in the Dark paradigm, adolescent C57Bl6 mice reach high blood alcohol concentration after voluntary binge‐drinking. In order to assess short‐ and long‐term consequences of adolescent alcohol exposure (AAE), a battery of behavioural tests was performed during late adolescence and during adulthood. We showed that AAE had no short‐term effect on young mice behaviour but rather increased anxiety‐ and depressive‐like behaviours, as well as alcohol consumption during adulthood. Moreover, alcohol binge‐drinking during adolescence dramatically decreased recognition memory performances and behavioural flexibility in both adult males and females. Furthermore, we showed that voluntary consumption of alcohol during adolescence did not trigger any major activation of the innate immune system in the prefrontal cortex. Together, our data suggest that voluntary alcohol binge‐drinking in adolescent mice induces a delayed appearance of behavioural impairments in adulthood.
Collapse
Affiliation(s)
- Laura Van Hees
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| | - Vincent Didone
- Psychology and Neuroscience of Cognition Research Unit (PsyNCog), Animal models of cognition University of Liège Liège Belgium
| | - Manon Charlet‐Briart
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| | - Théo Van Ingelgom
- Psychology and Neuroscience of Cognition Research Unit (PsyNCog), Animal models of cognition University of Liège Liège Belgium
| | - Alysson Alexandre
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| | - Etienne Quertemont
- Psychology and Neuroscience of Cognition Research Unit (PsyNCog), Animal models of cognition University of Liège Liège Belgium
| | - Laurent Nguyen
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| | - Sophie Laguesse
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| |
Collapse
|
49
|
Lorkiewicz SA, Baker FC, Müller-Oehring EM, Haas A, Wickham R, Sassoon SA, Clark DB, Nooner KB, Tapert SF, Brown SA, Schulte T. A Longitudinal Examination of Alcohol-Related Blackouts as a Predictor of Changes in Learning, Memory, and Executive Function in Adolescents. Front Psychiatry 2022; 13:866051. [PMID: 35599753 PMCID: PMC9120418 DOI: 10.3389/fpsyt.2022.866051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/23/2022] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION In adolescents, the relationship between alcohol-related blackouts (ARBs) and distinct cognitive changes lasting beyond intoxication is unclear. We examined ARBs as a predictor of persistent changes in the development of learning, memory, and executive function in participants from the National Consortium on Alcohol and Neurodevelopment in Adolescence (NCANDA) study. METHODS Descriptive analyses of the NCANDA sample (N = 831, 50.9% female, 12-21 years at baseline) identified ARB patterns within participants with an ARB history (n = 106). Latent growth curve modeling evaluated ARB-related performance changes on four neuropsychological measures across five years, excluding baseline data to reduce the magnitude of practice effects over time (n = 790). Measures included the Penn Conditional Exclusion Test (PCET), Penn Letter N-back Test (PLBT), Penn Facial Memory Test immediate (PFMTi), and delayed (PFMTd) recognition trials, and the Rey Complex Figure Test copy (RCFTc), immediate recall (RCFTi), and delayed recall (RCFTd) trials. Multivariate models were fit for raw accuracy scores from each measure, with ARB history (i.e., presence of past-year ARBs) as the main independent variable. Age, sex, race, socioeconomic status, assessment site, and alcohol use (i.e., past-year frequency) were included as covariates. Interaction effects between ARB history and alcohol use frequency were tested. RESULTS By year five, 16% of participants had experienced at least one ARB (59% of whom reported > 1 ARB and 57% of whom had an ARB lasting > 1 h). After controlling for demographics and alcohol use, ARB history predicted attenuated PFMTd performance growth at year one. Interaction effects between ARB history and alcohol use frequency predicted attenuated PFMTd performance growth at years one and two. ARB history predicted attenuated RCFTi and RCFTd performance growth by year four, but not PCET or PLBT performance over time. By contrast, greater past-year alcohol use predicted attenuated PFMTi and PFMTd performance growth between years two and four in adolescents without an ARB history. CONCLUSION We found that ARBs predict distinct, lasting changes in learning and memory for visual information, with results suggesting that the developing brain is vulnerable to ARBs during adolescence and emerging adulthood.
Collapse
Affiliation(s)
- Sara A Lorkiewicz
- Clinical Psychology, Palo Alto University, Palo Alto, CA, United States
| | - Fiona C Baker
- SRI International, Neuroscience Program, Menlo Park, CA, United States.,Brain Function Research Group, School of Physiology, University of Witwatersrand, Johannesburg, South Africa
| | - Eva M Müller-Oehring
- SRI International, Neuroscience Program, Menlo Park, CA, United States.,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States.,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Amie Haas
- Clinical Psychology, Palo Alto University, Palo Alto, CA, United States
| | - Robert Wickham
- Department of Psychological Sciences, Northern Arizona University, Flagstaff, AZ, United States
| | | | - Duncan B Clark
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kate B Nooner
- Department of Psychology, University of North Carolina, Wilmington, Wilmington, NC, United States
| | - Susan F Tapert
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States
| | - Sandra A Brown
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States.,Department of Psychology, University of California, San Diego, La Jolla, CA, United States
| | - Tilman Schulte
- Clinical Psychology, Palo Alto University, Palo Alto, CA, United States.,SRI International, Neuroscience Program, Menlo Park, CA, United States
| |
Collapse
|
50
|
Infante MA, Eberson SC, Zhang Y, Brumback T, Brown SA, Colrain IM, Baker FC, Clark DB, De Bellis MD, Goldston D, Nagel BJ, Nooner KB, Zhao Q, Pohl KM, Sullivan EV, Pfefferbaum A, Tapert SF, Thompson WK. Adolescent Binge Drinking Is Associated With Accelerated Decline of Gray Matter Volume. Cereb Cortex 2021; 32:2611-2620. [PMID: 34729592 DOI: 10.1093/cercor/bhab368] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 11/12/2022] Open
Abstract
The age- and time-dependent effects of binge drinking on adolescent brain development have not been well characterized even though binge drinking is a health crisis among adolescents. The impact of binge drinking on gray matter volume (GMV) development was examined using 5 waves of longitudinal data from the National Consortium on Alcohol and NeuroDevelopment in Adolescence study. Binge drinkers (n = 166) were compared with non-binge drinkers (n = 82 after matching on potential confounders). Number of binge drinking episodes in the past year was linked to decreased GMVs in bilateral Desikan-Killiany cortical parcellations (26 of 34 with P < 0.05/34) with the strongest effects observed in frontal regions. Interactions of binge drinking episodes and baseline age demonstrated stronger effects in younger participants. Statistical models sensitive to number of binge episodes and their temporal proximity to brain volumes provided the best fits. Consistent with prior research, results of this study highlight the negative effects of binge drinking on the developing brain. Our results present novel findings that cortical GMV decreases were greater in closer proximity to binge drinking episodes in a dose-response manner. This relation suggests a causal effect and raises the possibility that normal growth trajectories may be reinstated with alcohol abstinence.
Collapse
Affiliation(s)
- M A Infante
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - S C Eberson
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Y Zhang
- Division of Biostatistics, Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, USA.,Population Neuroscience and Genetics Lab, University of California, San Diego, USA
| | - T Brumback
- Department of Psychological Science, Northern Kentucky University, Kentucky, USA
| | - S A Brown
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.,Department of Psychology, University of California, San Diego, La Jolla, CA, USA
| | - I M Colrain
- Center for Health Sciences, SRI International, Menlo Park, CA, USA
| | - F C Baker
- Center for Health Sciences, SRI International, Menlo Park, CA, USA
| | - D B Clark
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - M D De Bellis
- Department of Psychiatry & Behavioral Sciences, Duke University, Durham, NC, USA
| | - D Goldston
- Department of Psychiatry & Behavioral Sciences, Duke University, Durham, NC, USA
| | - B J Nagel
- Departments of Psychiatry and Behavioral Neuroscience, Oregon Health & Sciences University, Portland, OR, USA
| | - K B Nooner
- Department of Psychology, University of North Carolina Wilmington, Wilmington, NC, USA
| | - Q Zhao
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - K M Pohl
- Center for Health Sciences, SRI International, Menlo Park, CA, USA.,Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - E V Sullivan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - A Pfefferbaum
- Center for Health Sciences, SRI International, Menlo Park, CA, USA.,Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - S F Tapert
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - W K Thompson
- Division of Biostatistics, Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, USA.,Population Neuroscience and Genetics Lab, University of California, San Diego, USA.,Department of Radiology, University of California, San Diego, USA
| |
Collapse
|