1
|
Ji L, Menu I, Majbri A, Bhatia T, Trentacosta CJ, Thomason ME. Trajectories of human brain functional connectome maturation across the birth transition. PLoS Biol 2024; 22:e3002909. [PMID: 39561110 PMCID: PMC11575827 DOI: 10.1371/journal.pbio.3002909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Understanding the sequence and timing of brain functional network development at the beginning of human life is critically important from both normative and clinical perspectives. Yet, we presently lack rigorous examination of the longitudinal emergence of human brain functional networks over the birth transition. Leveraging a large, longitudinal perinatal functional magnetic resonance imaging (fMRI) data set, this study models developmental trajectories of brain functional networks spanning 25 to 55 weeks of post-conceptual gestational age (GA). The final sample includes 126 fetal scans (GA = 31.36 ± 3.83 weeks) and 58 infant scans (GA = 48.17 ± 3.73 weeks) from 140 unique subjects. In this study, we document the developmental changes of resting-state functional connectivity (RSFC) over the birth transition, evident at both network and graph levels. We observe that growth patterns are regionally specific, with some areas showing minimal RSFC changes, while others exhibit a dramatic increase at birth. Examples with birth-triggered dramatic change include RSFC within the subcortical network, within the superior frontal network, within the occipital-cerebellum joint network, as well as the cross-hemisphere RSFC between the bilateral sensorimotor networks and between the bilateral temporal network. Our graph analysis further emphasized the subcortical network as the only region of the brain exhibiting a significant increase in local efficiency around birth, while a concomitant gradual increase was found in global efficiency in sensorimotor and parietal-frontal regions throughout the fetal to neonatal period. This work unveils fundamental aspects of early brain development and lays the foundation for future work on the influence of environmental factors on this process.
Collapse
Affiliation(s)
- Lanxin Ji
- Department of Child and Adolescent Psychiatry, New York University School of Medicine, New York, New York State, United States of America
| | - Iris Menu
- Department of Child and Adolescent Psychiatry, New York University School of Medicine, New York, New York State, United States of America
| | - Amyn Majbri
- Department of Child and Adolescent Psychiatry, New York University School of Medicine, New York, New York State, United States of America
| | - Tanya Bhatia
- Department of Child and Adolescent Psychiatry, New York University School of Medicine, New York, New York State, United States of America
| | | | - Moriah E Thomason
- Department of Child and Adolescent Psychiatry, New York University School of Medicine, New York, New York State, United States of America
- Department of Population Health, New York University School of Medicine, New York, New York State, United States of America
- Neuroscience Institute, New York University School of Medicine, New York, New York State, United States of America
| |
Collapse
|
2
|
Lo JO, Shaw B, Robalino S, Ayers CK, Durbin S, Rushkin MC, Olyaei A, Kansagara D, Harrod CS. Cannabis Use in Pregnancy and Neonatal Outcomes: A Systematic Review and Meta-Analysis. Cannabis Cannabinoid Res 2024; 9:470-485. [PMID: 36730710 PMCID: PMC11262585 DOI: 10.1089/can.2022.0262] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Objective: To determine whether prenatal cannabis use alone increases the likelihood of fetal and neonatal morbidity and mortality. Study Design: We searched bibliographic databases, such as PubMed, Embase, Scopus, Cochrane reviews, PsycInfo, MEDLINE, Clinicaltrials.gov, and Google Scholar from inception through February 14, 2022. Cohort or case-control studies with prespecified fetal or neonatal outcomes in pregnancies with prenatal cannabis use. Primary outcomes were preterm birth (PTB; <37 weeks of gestation), small-for-gestational-age (SGA), birthweight (grams), and perinatal mortality. Two independent reviewers screened studies. Studies were extracted by one reviewer and confirmed by a second using a predefined template. Risk of bias assessment of studies, using the Newcastle-Ottawa Quality Assessment Scale, and Grading of Recommendations Assessment, Development, and Evaluation for evaluating the certainty of evidence for select outcomes were performed by two independent reviewers with disagreements resolved by a third. Random effects meta-analyses were conducted, using adjusted and unadjusted effect estimates, to compare groups according to prenatal exposure to cannabis use status. Results: Fifty-three studies were included. Except for birthweight, unadjusted and adjusted meta-analyses had similar results. We found very-low- to low-certainty evidence that cannabis use during pregnancy was significantly associated with greater odds of PTB (adjusted odds ratio [aOR], 1.42; 95% confidence interval [CI], 1.19 to 1.69; I2, 93%; p=0.0001), SGA (aOR, 1.76; 95% CI, 1.52 to 2.05; I2, 86%; p<0.0001), and perinatal mortality (aOR, 1.5; 95% CI, 1.39 to 1.62; I2, 0%; p<0.0001), but not significantly different for birthweight (mean difference, -40.69 g; 95% CI, -124.22 to 42.83; I2, 85%; p=0.29). Because of substantial heterogeneity, we also conducted a narrative synthesis and found comparable results to meta-analyses. Conclusion: Prenatal cannabis use was associated with greater odds of PTB, SGA, and perinatal mortality even after accounting for prenatal tobacco use. However, our confidence in these findings is limited. Limitations of most existing studies was the failure to not include timing or quantity of cannabis use. This review can help guide health care providers with counseling, management, and addressing the limited existing safety data. Protocol Registration: PROSPERO CRD42020172343.
Collapse
Affiliation(s)
- Jamie O. Lo
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon, USA
| | - Beth Shaw
- Department of Obstetrics and Gynecology, Center for Evidence-Based Policy, Oregon Health and Science University, Portland, Oregon, USA
| | - Shannon Robalino
- Department of Obstetrics and Gynecology, Center for Evidence-Based Policy, Oregon Health and Science University, Portland, Oregon, USA
| | - Chelsea K. Ayers
- Center for the Involvement of Veterans in their Care (CIVIC), VA Portland Health Care System, Portland, Oregon, USA
| | - Shauna Durbin
- Department of Obstetrics and Gynecology, Center for Evidence-Based Policy, Oregon Health and Science University, Portland, Oregon, USA
| | - Megan C. Rushkin
- Department of Obstetrics and Gynecology, Center for Evidence-Based Policy, Oregon Health and Science University, Portland, Oregon, USA
| | - Amy Olyaei
- Division of Neonatology, Department of Pediatrics, Oregon Health and Science University, Portland, Oregon, USA
| | - Devan Kansagara
- Center for the Involvement of Veterans in their Care (CIVIC), VA Portland Health Care System, Portland, Oregon, USA
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Curtis S. Harrod
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
3
|
Sorkhou M, Singla DR, Castle DJ, George TP. Birth, cognitive and behavioral effects of intrauterine cannabis exposure in infants and children: A systematic review and meta-analysis. Addiction 2024; 119:411-437. [PMID: 37968824 PMCID: PMC10872597 DOI: 10.1111/add.16370] [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: 11/21/2022] [Accepted: 09/18/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND AND AIMS Δ9-tetrahydrocannabinol (THC), the principal psychoactive component of cannabis, has been implicated in affecting fetal neurodevelopment by readily crossing the placenta. However, little is known regarding the long-term effects of intrauterine cannabis exposure. This systematic review and meta-analysis synthesized prospective and cross-sectional human studies to measure the effects of intrauterine cannabis exposure on birth, behavioral, psychological and cognitive outcomes in infancy until early childhood. METHODS Reporting according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement, cross-sectional and prospective studies published from database inception until June 2023, investigating developmental outcomes of infants, toddlers and young children with intrauterine cannabis exposure were considered. All articles were obtained from PubMed or PsycINFO databases. RESULTS The literature search resulted in 932 studies, in which 57 articles met eligibility criteria. The meta-analysis revealed that intrauterine cannabis exposure increases the risk of preterm delivery [odds ratio (OR) = 1.68, 95% confidence interval (CI) = 1.05-2.71, P = 0.03], low birth weight (OR = 2.60, CI = 1.71-3.94, P < 0.001) and requirement for neonatal intensive care unit (NICU) admission (OR = 2.51, CI = 1.46-4.31; P < 0.001). Our qualitative synthesis suggests that intrauterine cannabis exposure may be associated with poorer attention and externalizing problems in infancy and early childhood. We found no evidence for impairments in other cognitive domains or internalizing behaviors. CONCLUSIONS Prenatal cannabis use appears to be associated with lower birth weight, preterm birth and neonatal intensive care unit admission in newborns, but there is little evidence that prenatal cannabis exposure adversely impacts behavioral or cognitive outcomes in early childhood, with the exception of attention and externalizing problems.
Collapse
Affiliation(s)
- Maryam Sorkhou
- Centre for Complex Interventions, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Department of Psychiatry, University of Toronto, ON, Canada
| | - Daisy R Singla
- Centre for Complex Interventions, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Department of Psychiatry, University of Toronto, ON, Canada
| | - David J Castle
- Tasmania Centre for Mental Health Service Innovation, University of Tasmania, Hobart, Australia
| | - Tony P George
- Centre for Complex Interventions, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Department of Psychiatry, University of Toronto, ON, Canada
| |
Collapse
|
4
|
Cabral L, Calabro FJ, Foran W, Parr AC, Ojha A, Rasmussen J, Ceschin R, Panigrahy A, Luna B. Multivariate and regional age-related change in basal ganglia iron in neonates. Cereb Cortex 2024; 34:bhad456. [PMID: 38059685 PMCID: PMC11494441 DOI: 10.1093/cercor/bhad456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 12/08/2023] Open
Abstract
In the perinatal period, reward and cognitive systems begin trajectories, influencing later psychiatric risk. The basal ganglia is important for reward and cognitive processing but early development has not been fully characterized. To assess age-related development, we used a measure of basal ganglia physiology, specifically brain tissue iron, obtained from nT2* signal in resting-state functional magnetic resonance imaging (rsfMRI), associated with dopaminergic processing. We used data from the Developing Human Connectome Project (n = 464) to assess how moving from the prenatal to the postnatal environment affects rsfMRI nT2*, modeling gestational and postnatal age separately for basal ganglia subregions in linear models. We did not find associations with tissue iron and gestational age [range: 24.29-42.29] but found positive associations with postnatal age [range:0-17.14] in the pallidum and putamen, but not the caudate. We tested if there was an interaction between preterm birth and postnatal age, finding early preterm infants (GA < 35 wk) had higher iron levels and changed less over time. To assess multivariate change, we used support vector regression to predict age from voxel-wise-nT2* maps. We could predict postnatal but not gestational age when maps were residualized for the other age term. This provides evidence subregions differentially change with postnatal experience and preterm birth may disrupt trajectories.
Collapse
Affiliation(s)
- Laura Cabral
- Department of Radiology University of Pittsburgh, Pittsburgh, PA 15224, United States
| | - Finnegan J Calabro
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, United States
- Department of Bioengineering, University of Pittsburgh, 15213, United States
| | - Will Foran
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Ashley C Parr
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Amar Ojha
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, United States
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Jerod Rasmussen
- Development, Health and Disease Research Program, University of California, Irvine, CA 92697, United States
- Department of Pediatrics, University of California, Irvine, CA 92697, United States
| | - Rafael Ceschin
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15224, United States
| | - Ashok Panigrahy
- Department of Radiology University of Pittsburgh, Pittsburgh, PA 15224, United States
| | - Beatriz Luna
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, United States
| |
Collapse
|
5
|
Vishnubhotla RV, Ahmad ST, Zhao Y, Radhakrishnan R. Impact of prenatal marijuana exposure on adolescent brain structural and functional connectivity and behavioural outcomes. Brain Commun 2024; 6:fcae001. [PMID: 38444906 PMCID: PMC10914455 DOI: 10.1093/braincomms/fcae001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 11/01/2023] [Accepted: 01/05/2024] [Indexed: 03/07/2024] Open
Abstract
There has been an increase in the number of women using marijuana whilst pregnant. Previous studies have shown that children with prenatal marijuana exposure have developmental deficits in memory and decreased attentiveness. In this study, we assess whether prenatal marijuana exposure is associated with alterations in brain regional morphometry and functional and structural connectivity in adolescents. We downloaded behavioural scores and subject image files from the Adolescent Brain Cognitive DevelopmentSM Study. A total of 178 anatomical and diffusion magnetic resonance imaging files (88 prenatal marijuana exposure and 90 age- and gender-matched controls) and 152 resting-state functional magnetic resonance imaging files (76 prenatal marijuana exposure and 76 controls) were obtained. Behavioural metrics based on the parent-reported child behavioural checklist were also obtained for each subject. The associations of prenatal marijuana exposure with 17 subscales of the child behavioural checklist were calculated. We assessed differences in brain morphometry based on voxel-based and surface-based morphometry in adolescents with prenatal marijuana exposure versus controls. We also evaluated group differences in structural and functional connectivity in adolescents for region-to-region connectivity and graph theoretical metrics. Interactions of prenatal marijuana exposure and graph networks were assessed for impact on behavioural scores. Multiple comparison correction was performed as appropriate. Adolescents with prenatal marijuana exposure had greater abnormal or borderline child behavioural checklist scores in 9 out of 17 subscales. There were no significant differences in voxel- or surface-based morphometry, structural connectivity or functional connectivity between prenatal marijuana exposure and controls. However, there were significant differences in prenatal marijuana exposure-graph network interactions with respect to behavioural scores. There were three structural prenatal marijuana exposure-graph network interactions and seven functional prenatal marijuana exposure-graph network interactions that were significantly associated with behavioural scores. Whilst this study was not able to confirm anatomical or functional differences between prenatal marijuana exposure and unexposed pre-adolescent children, there were prenatal marijuana exposure-brain structural and functional graph network interactions that were significantly associated with behavioural scores. This suggests that altered brain networks may underlie behavioural outcomes in adolescents with prenatal marijuana exposure. More work needs to be conducted to better understand the prognostic value of brain structural and functional network measures in prenatal marijuana exposure.
Collapse
Affiliation(s)
- Ramana V Vishnubhotla
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sidra T Ahmad
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yi Zhao
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Authement AK, Isoherranen N. The impact of pregnancy and associated hormones on the pharmacokinetics of Δ 9-tetrahydrocannabinol. Expert Opin Drug Metab Toxicol 2024; 20:73-93. [PMID: 38258511 DOI: 10.1080/17425255.2024.2309213] [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: 10/24/2023] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
INTRODUCTION (-)-Δ9-tetrahydrocannabinol (THC) is the main psychoactive component of cannabis. Cannabis is the most widely used drug of abuse by pregnant individuals, but its maternal-fetal safety is still unclear. The changes in THC disposition during pregnancy may affect THC safety and pharmacology. AREAS COVERED This review summarizes the current literature on THC metabolism and pharmacokinetics in humans. It provides an analysis of how hormonal changes during pregnancy may alter the expression of cannabinoid metabolizing enzymes and THC and its metabolite pharmacokinetics. THC is predominately (>70%) cleared by hepatic metabolism to its psychoactive active metabolite, 11-OH-THC by cytochrome P450 (CYP) 2C9 and to other metabolites (<30%) by CYP3A4. Other physiological processes that change during pregnancy and may alter cannabinoid disposition are also reviewed. EXPERT OPINION THC and its metabolites disposition likely change during pregnancy. Hepatic CYP2C9 and CYP3A4 are induced in pregnant individuals and in vitro by pregnancy hormones. This induction of CYP2C9 and CYP3A4 is predicted to lead to altered THC and 11-OH-THC disposition and pharmacodynamic effects. More in vitro studies of THC metabolism and induction of the enzymes metabolizing cannabinoids are necessary to improve the prediction of THC pharmacokinetics in pregnant individuals.
Collapse
Affiliation(s)
- Aurora K Authement
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
7
|
Kraaijenvanger EJ, Banaschewski T, Eickhoff SB, Holz NE. A coordinate-based meta-analysis of human amygdala connectivity alterations related to early life adversities. Sci Rep 2023; 13:16541. [PMID: 37783710 PMCID: PMC10545708 DOI: 10.1038/s41598-023-43057-2] [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: 10/12/2022] [Accepted: 09/19/2023] [Indexed: 10/04/2023] Open
Abstract
By affecting core neurobiological systems early in development, early life adversities (ELAs) might confer latent vulnerability to future psychopathologies. This coordinate-based meta-analysis aims to identify significant convergent alterations in functional connectivity of the amygdala related to ELAs across resting-state and task-based fMRI-studies. Five electronic databases were systematically searched until 22 October 2020, retrieving 49 eligible studies (n = 3162 participants). Convergent alterations in functional connectivity related to ELAs between the amygdala and the anterior cingulate cortex (ACC) and left hippocampus were found. Sub-analyses based on hemisphere and direction showed that connectivity seeded in the right amygdala was affected and, moreover, revealed that connectivity with ACC was decreased. Analyses based on paradigm and age showed that amygdala-ACC coupling was altered during resting state and that amygdala-left hippocampus connectivity was mostly affected during task-based paradigms and in adult participants. While both regions showed altered connectivity during emotion processing and following adverse social postnatal experiences such as maltreatment, amygdala-ACC coupling was mainly affected when ELAs were retrospectively assessed through self-report. We show that ELAs are associated with altered functional connectivity of the amygdala with the ACC and hippocampus. As such, ELAs may embed latent vulnerability to future psychopathologies by systematically affecting important neurocognitive systems.
Collapse
Affiliation(s)
- Eline J Kraaijenvanger
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J5, 68159, Mannheim, Germany
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J5, 68159, Mannheim, Germany
| | - Simon B Eickhoff
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
| | - Nathalie E Holz
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J5, 68159, Mannheim, Germany.
- Donders Institute, Radboud University, Nijmegen, The Netherlands.
- Radboud University Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
8
|
Holz NE, Berhe O, Sacu S, Schwarz E, Tesarz J, Heim CM, Tost H. Early Social Adversity, Altered Brain Functional Connectivity, and Mental Health. Biol Psychiatry 2023; 93:430-441. [PMID: 36581495 DOI: 10.1016/j.biopsych.2022.10.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022]
Abstract
Early adverse environmental exposures during brain development are widespread risk factors for the onset of severe mental disorders and strong and consistent predictors of stress-related mental and physical illness and reduced life expectancy. Current evidence suggests that early negative experiences alter plasticity processes during developmentally sensitive time windows and affect the regular functional interaction of cortical and subcortical neural networks. This, in turn, may promote a maladapted development with negative consequences on the mental and physical health of exposed individuals. In this review, we discuss the role of functional magnetic resonance imaging-based functional connectivity phenotypes as potential biomarker candidates for the consequences of early environmental exposures-including but not limited to-childhood maltreatment. We take an expanded concept of developmentally relevant adverse experiences from infancy over childhood to adolescence as our starting point and focus our review of functional connectivity studies on a selected subset of functional magnetic resonance imaging-based phenotypes, including connectivity in the limbic and within the frontoparietal as well as default mode networks, for which we believe there is sufficient converging evidence for a more detailed discussion in a developmental context. Furthermore, we address specific methodological challenges and current knowledge gaps that complicate the interpretation of early stress effects on functional connectivity and deserve particular attention in future studies. Finally, we highlight the forthcoming prospects and challenges of this research area with regard to establishing functional connectivity measures as validated biomarkers for brain developmental processes and individual risk stratification and as target phenotypes for mechanism-based interventions.
Collapse
Affiliation(s)
- Nathalie E Holz
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands; Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig Holstein, Kiel University, Kiel, Germany; Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Oksana Berhe
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Seda Sacu
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jonas Tesarz
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Christine M Heim
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Berlin, Germany; College of Health and Human Development, The Pennsylvania State University, University Park, Pennsylvania
| | - Heike Tost
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
9
|
Grecco GG, Shahid SS, Atwood BK, Wu YC. Alterations of brain microstructures in a mouse model of prenatal opioid exposure detected by diffusion MRI. Sci Rep 2022; 12:17085. [PMID: 36224335 PMCID: PMC9556691 DOI: 10.1038/s41598-022-21416-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/27/2022] [Indexed: 01/04/2023] Open
Abstract
Growing opioid use among pregnant women is fueling a crisis of infants born with prenatal opioid exposure. A large body of research has been devoted to studying the management of opioid withdrawal during the neonatal period in these infants, but less substantive work has explored the long-term impact of prenatal opioid exposure on neurodevelopment. Using a translationally relevant mouse model of prenatal methadone exposure (PME), the aim of the study is to investigate the cerebral microstructural differences between the mice with PME and prenatal saline exposure (PSE). The brains of eight-week-old male offspring with either PME (n = 15) or PSE (n = 15) were imaged using high resolution in-vivo diffusion magnetic resonance imaging on a 9.4 Tesla small animal scanner. Brain microstructure was characterized using diffusion tensor imaging (DTI) and Bingham neurite orientation dispersion and density imaging (Bingham-NODDI). Voxel-based analysis (VBA) was performed using the calculated microstructural parametric maps. The VBA showed significant (p < 0.05) bilateral alterations in fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD), radial diffusivity (RD), orientation dispersion index (ODI) and dispersion anisotropy index (DAI) across several cortical and subcortical regions, compared to PSE. Particularly, in PME offspring, FA, MD and AD were significantly higher in the hippocampus, dorsal amygdala, thalamus, septal nuclei, dorsal striatum and nucleus accumbens. These DTI-based results suggest widespread bilateral microstructural alterations across cortical and subcortical regions in PME offspring. Consistent with the observations in DTI, Bingham-NODDI derived ODI exhibited significant reduction in PME offspring within the hippocampus, dorsal striatum and cortex. NODDI-based results further suggest reduction in dendritic arborization in PME offspring across multiple cortical and subcortical regions. To our best knowledge, this is the first study of prenatal opioid exposure to examine microstructural organization in vivo. Our findings demonstrate perturbed microstructural complexity in cortical and subcortical regions persisting into early adulthood which could interfere with critical neurodevelopmental processes in individuals with prenatal opioid exposure.
Collapse
Affiliation(s)
- Gregory G Grecco
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana University School of Medicine, Medical Scientist Training Program, Indianapolis, IN, 46202, USA
| | - Syed Salman Shahid
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, Suite 4100, Indianapolis, IN, 46202, USA
| | - Brady K Atwood
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yu-Chien Wu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, Suite 4100, Indianapolis, IN, 46202, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
10
|
Lowell AF, Morie K, Potenza MN, Crowley MJ, Mayes LC. An intergenerational lifespan perspective on the neuroscience of prenatal substance exposure. Pharmacol Biochem Behav 2022; 219:173445. [PMID: 35970340 DOI: 10.1016/j.pbb.2022.173445] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 07/26/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022]
Abstract
Prenatal substance exposure has the potential to impact a variety of domains, with neurobiological effects that last throughout the lifespan. Different substances may impact the brain in both specific and diffuse ways; however, the aberrant neural outcomes following exposure tend to coalesce in three areas: (1) sensorimotor development; (2) arousal, motivation, and reward; and (3) executive functioning, impulse control, and emotion regulation. This manuscript represents a summary and update of a previous review (Morie et al., 2019). We organize this piece by domain and summarize data from published neuroimaging studies that examine the neural correlates of prenatal exposure across developmental stages. While the published neuroimaging literature in the area of prenatal exposure has a range of sampling concerns that may limit generalizability as well as longitudinal prediction, the findings to date do point to domains of interest warranting further study. With this caveat, we synthesize the extant findings to describe ways in which prenatal substance exposure is associated with developmental psychopathology and implicated in potentially aberrant behavioral patterns beginning in infancy and persisting through childhood, adolescence, adulthood, and even parenthood. We also examine how substance abuse may impact parenting behaviors that in turn influences infant and child behavior in ways that may be additive or obscure the direct teratological effects of prenatal exposure. Given this observation, we offer an additional intergenerational lens through which prenatal substance exposure should be studied.
Collapse
Affiliation(s)
- Amanda F Lowell
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA.
| | - Kristen Morie
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Marc N Potenza
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA; Connecticut Mental Health Center, New Haven, CT, USA; Connecticut Council on Problem Gambling, Wethersfield, CT, USA
| | - Michael J Crowley
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Linda C Mayes
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
11
|
Crume TL, Powers S, Dufford AJ, Kim P. Cannabis and Pregnancy: Factors Associated with Cannabis Use Among Pregnant Women and the Consequences for Offspring Neurodevelopment and Early Postpartum Parenting Behavior. CURRENT ADDICTION REPORTS 2022. [DOI: 10.1007/s40429-022-00419-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
12
|
Liu J, Grewen K, Gao W. Evidence for the Normalization Effects of Medication for Opioid Use Disorder on Functional Connectivity in Neonates with Prenatal Opioid Exposure. J Neurosci 2022; 42:4555-4566. [PMID: 35552232 PMCID: PMC9172285 DOI: 10.1523/jneurosci.2232-21.2022] [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] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/30/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Altered functional connectivity has been reported in infants with prenatal exposure to opioids, which significantly interrupts and influences endogenous neurotransmitter/receptor signaling during fetal programming. Better birth outcomes and long-term developmental outcomes are associated with medication for opioid use disorder (MOUD) during pregnancy, but the neural mechanisms underlying these benefits are largely unknown. We aimed to characterize effects of prenatal opioid/other drug exposure (PODE) and the neural basis for the reported beneficial effects of MOUD by examining neonatal brain functional organization. A cohort of 109 human newborns, 42 PODE, 39 with prenatal exposure to drugs excluding opioids (PDE), 28 drug-free controls (males and females) underwent resting-state fMRI at 2 weeks of age. To examine neural effects of MOUD, PODE infants were separated into subgroups based on whether mothers received MOUD (n = 31) or no treatment (n = 11). A novel heatmap analysis was designed to characterize PODE-associated functional connectivity alterations and MOUD-related effects, and permutation testing identified regions of interest with significant effects. PODE neonates showed alterations beyond those associated with PDE, particularly in reward-related frontal-sensory connectivity. MOUD was associated with a significant reduction of PODE-related alterations in key regions of endogenous opioid pathways including limbic and frontal connections. However, significant residual effects in limbic and subcortical circuitry were observed. These findings confirm altered brain functional organization associated with PODE. Importantly, widespread normalization effects associated with MOUD reveal, for the first time, the potential brain basis of the beneficial effects of MOUD on the developing brain and underscore the importance of this treatment intervention for better developmental outcomes.SIGNIFICANCE STATEMENT This is the first study to reveal the potential neural mechanisms underlying the beneficial effects on the neonate brain associated with MOUD during pregnancy. We identified both normalization and residual effects of MOUD on brain functional architecture by directly comparing neonates prenatally exposed to opioids with MOUD and those exposed to opioids but without MOUD. Our findings confirm altered brain functional organization associated with prenatal opioid exposure and demonstrate that although significant residual effects remain in reward circuitry, MOUD confers significant normalization effects on functional connectivity of regions associated with socioemotional development and reward processing. Together, our results highlight the importance of MOUD intervention for better neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Janelle Liu
- Cedars-Sinai Biomedical Imaging Research Institute, Los Angeles, California 90048
- Departments of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Karen Grewen
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Wei Gao
- Cedars-Sinai Biomedical Imaging Research Institute, Los Angeles, California 90048
- Departments of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, California 90048
| |
Collapse
|
13
|
Jiang W, Merhar SL, Zeng Z, Zhu Z, Yin W, Zhou Z, Wang L, He L, Vannest J, Lin W. Neural alterations in opioid-exposed infants revealed by edge-centric brain functional networks. Brain Commun 2022; 4:fcac112. [PMID: 35602654 PMCID: PMC9117006 DOI: 10.1093/braincomms/fcac112] [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: 12/10/2021] [Revised: 03/29/2022] [Accepted: 05/03/2022] [Indexed: 12/02/2022] Open
Abstract
Prenatal opioid exposure has been linked to adverse effects spanning multiple neurodevelopmental domains, including cognition, motor development, attention, and vision. However, the neural basis of these abnormalities is largely unknown. A total of 49 infants, including 21 opioid-exposed and 28 controls, were enrolled and underwent MRI (43 ± 6 days old) after birth, including resting state functional MRI. Edge-centric functional networks based on dynamic functional connections were constructed, and machine-learning methods were employed to identify neural features distinguishing opioid-exposed infants from unexposed controls. An accuracy of 73.6% (sensitivity 76.25% and specificity 69.33%) was achieved using 10 times 10-fold cross-validation, which substantially outperformed those obtained using conventional static functional connections (accuracy 56.9%). More importantly, we identified that prenatal opioid exposure preferentially affects inter- rather than intra-network dynamic functional connections, particularly with the visual, subcortical, and default mode networks. Consistent results at the brain regional and connection levels were also observed, where the brain regions and connections associated with visual and higher order cognitive functions played pivotal roles in distinguishing opioid-exposed infants from controls. Our findings support the clinical phenotype of infants exposed to opioids in utero and may potentially explain the higher rates of visual and emotional problems observed in this population. Finally, our findings suggested that edge-centric networks could better capture the neural differences between opioid-exposed infants and controls by abstracting the intrinsic co-fluctuation along edges, which may provide a promising tool for future studies focusing on investigating the effects of prenatal opioid exposure on neurodevelopment.
Collapse
Affiliation(s)
- Weixiong Jiang
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Stephanie L. Merhar
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital and University of Cincinnati Department of Pediatrics, Cincinnati OH, United States
| | - Zhuohao Zeng
- East Chapel Hill High School, Chapel Hill, North Carolina, United States
| | - Ziliang Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Weiyan Yin
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Zhen Zhou
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Li Wang
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Lili He
- Department of Radiology, Cincinnati Children’s Hospital and University of Cincinnati, Cincinnati OH, United States
| | - Jennifer Vannest
- Department of Communication Sciences and Disorders, University of Cincinnati, Cincinnati OH, United States
| | - Weili Lin
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
14
|
Hendrix CL, Thomason ME. A survey of protocols from 54 infant and toddler neuroimaging research labs. Dev Cogn Neurosci 2022; 54:101060. [PMID: 35033971 PMCID: PMC8762357 DOI: 10.1016/j.dcn.2022.101060] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/20/2021] [Accepted: 01/09/2022] [Indexed: 01/13/2023] Open
Abstract
Infant and toddler MRI enables unprecedented insight into the developing brain. However, consensus about optimal data collection practices is lacking, which slows growth of the field and impedes replication efforts. The goal of this study was to collect systematic data across a large number of infant/toddler research laboratories to better understand preferred practices. Survey data addressed MRI acquisition strategies, scan success rates, visit preparations, scanning protocols, accommodations for families, study design, and policies regarding incidental findings. Respondents had on average 8 years' experience in early life neuroimaging and represented more than fifty research laboratories. Areas of consensus across labs included higher success rates among newborns compared to older infants or toddlers, high rates of data loss across age groups, endorsement of multiple layers of hearing protection, and age-specific scan preparation and participant accommodation. Researchers remain divided on decisions in longitudinal study design and practices regarding incidental findings. This study summarizes practices honed over years of work by a large collection of scientists, which may serve as an important resource for those new to the field. The ability to reference data about best practices facilitates future harmonization, data sharing, and reproducibility, all of which advance this important frontier in developmental science.
Collapse
Affiliation(s)
- Cassandra L Hendrix
- Department of Child and Adolescent Psychiatry, New York University Medical Center, New York, NY, USA.
| | - Moriah E Thomason
- Department of Child and Adolescent Psychiatry, New York University Medical Center, New York, NY, USA; Department of Population Health, New York University Medical Center, New York, NY, USA; Neuroscience Institute, New York University Medical Center, New York, NY, USA
| |
Collapse
|
15
|
Dufford AJ, Spann M, Scheinost D. How prenatal exposures shape the infant brain: Insights from infant neuroimaging studies. Neurosci Biobehav Rev 2021; 131:47-58. [PMID: 34536461 DOI: 10.1016/j.neubiorev.2021.09.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/30/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
Brain development during the prenatal period is rapid and unparalleled by any other time during development. Biological systems undergoing rapid development are at higher risk for disorganizing influences. Therefore, certain prenatal exposures impact brain development, increasing risk for negative neurodevelopmental outcome. While prenatal exposures have been associated with cognitive and behavioral outcomes later in life, the underlying macroscopic brain pathways remain unclear. Here, we review magnetic resonance imaging (MRI) studies investigating the association between prenatal exposures and infant brain development focusing on prenatal exposures via maternal physical health factors, maternal mental health factors, and maternal drug and medication use. Further, we discuss the need for studies to consider multiple prenatal exposures in parallel and suggest future directions for this body of research.
Collapse
Affiliation(s)
| | - Marisa Spann
- Columbia University Irving Medical Center, 622 West 168th Street, New York, NY, 10032, USA
| | - Dustin Scheinost
- Child Study Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, USA; Department of Statistics and Data Science, Yale University, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| |
Collapse
|
16
|
Wang P, Feng J, Wang Y, Zhu W, Wei S, Im H, Wang Q. Sex-specific static and dynamic functional networks of sub-divisions of striatum linking to the greed personality trait. Neuropsychologia 2021; 163:108066. [PMID: 34678357 DOI: 10.1016/j.neuropsychologia.2021.108066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 11/30/2022]
Abstract
The study of greed has been broadly investigated and discussed in the field of social sciences, including economics, political science, and psychology. However, the neural mechanisms underlying greed personality trait (GPT) have received little attention from the cognitive neuroscience field and still remain unclear. In this study, we explored the associations between GPT and static/dynamic reward circuit-specifically its sub-regions' functional networks including caudate, nucleus accumbens (NAcc), and putamen. Behavioral analyses revealed significant associations of GPT with Past-Negative and Present-Fatalistic time attitude as well as attention impulsivity. Imaging analyses revealed a significant interaction effect between sex and GPT on the static reward functional networks. In particular, GPT was positively correlated with static caudate-NAcc, caudate-cerebellum, and NAcc-parahippocampus/medial orbitofrontal cortex (PHG/mOFC) for males but negatively correlated for females. GPT was also marginally and negatively correlated with static putamen-occipital pole functional connectivities among males. Interestingly, sex difference interaction patterns were further observed in the dynamic reward functional networks. Further, dynamic reward functional networks also exhibited some specific characteristics, manifesting in more brain regions involved for greedy behaviors. These findings suggest sex-specific static and dynamic functional networks underlying human dispositional greed, and also implicate the critical contributions of reward circuit, especially for sub-circuits of reward, on greed.
Collapse
Affiliation(s)
- Pinchun Wang
- Faculty of Psychology, Tianjin Normal University, Tianjin, 300387, China
| | - Jie Feng
- Faculty of Psychology, Tianjin Normal University, Tianjin, 300387, China
| | - Yajie Wang
- Faculty of Psychology, Tianjin Normal University, Tianjin, 300387, China
| | - Wenwei Zhu
- Faculty of Psychology, Tianjin Normal University, Tianjin, 300387, China
| | - Shiyu Wei
- Faculty of Psychology, Tianjin Normal University, Tianjin, 300387, China
| | - Hohjin Im
- Department of Psychological Science, University of California, Irvine, 92697-7085, CA, USA.
| | - Qiang Wang
- Faculty of Psychology, Tianjin Normal University, Tianjin, 300387, China; Key Research Base of Humanities and Social of the Ministry of Education, Academy of Psychology and Behavior, Tianjin Normal University, Tianjin, 300387, China; Tianjin Social Science Laboratory of Students' Mental Development and Learning, Tianjin, 300387, China.
| |
Collapse
|
17
|
Merhar SL, Jiang W, Parikh NA, Yin W, Zhou Z, Tkach JA, Wang L, Kline-Fath BM, He L, Braimah A, Vannest J, Lin W. Effects of prenatal opioid exposure on functional networks in infancy. Dev Cogn Neurosci 2021; 51:100996. [PMID: 34388637 PMCID: PMC8363826 DOI: 10.1016/j.dcn.2021.100996] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 11/20/2022] Open
Abstract
Prenatal opioid exposure has been linked to altered neurodevelopment and visual problems such as strabismus and nystagmus. The neural substrate underlying these alterations is unclear. Resting-state functional connectivity MRI (rsfMRI) is an advanced and well-established technique to evaluate brain networks. Few studies have examined the effects of prenatal opioid exposure on resting-state network connectivity in infancy. In this pilot study, we characterized network connectivity in opioid-exposed infants (n = 19) and controls (n = 20) between 4–8 weeks of age using both a whole-brain connectomic approach and a seed-based approach. Prenatal opioid exposure was associated with differences in distribution of betweenness centrality and connection length, with positive connections unique to each group significantly longer than common connections. The unique connections in the opioid-exposed group were more often inter-network connections while unique connections in controls and connections common to both groups were more often intra-network. The opioid-exposed group had smaller network volumes particularly in the primary visual network, but similar network strength as controls. Network topologies as determined by dice similarity index were different between groups, particularly in visual and executive control networks. These results may provide insight into the neural basis for the developmental and visual problems associated with prenatal opioid exposure.
Collapse
Affiliation(s)
- Stephanie L Merhar
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital and University of Cincinnati, Department of Pediatrics, Cincinnati, OH, USA.
| | - Weixiong Jiang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nehal A Parikh
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital and University of Cincinnati, Department of Pediatrics, Cincinnati, OH, USA
| | - Weiyan Yin
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhen Zhou
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jean A Tkach
- Imaging Research Center, Cincinnati Children's Hospital, Cincinnati, OH, USA; Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Li Wang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Beth M Kline-Fath
- Imaging Research Center, Cincinnati Children's Hospital, Cincinnati, OH, USA; Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Lili He
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital and University of Cincinnati, Department of Pediatrics, Cincinnati, OH, USA
| | - Adebayo Braimah
- Imaging Research Center, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Jennifer Vannest
- Department of Communication Sciences and Disorders, College of Allied Health Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Weili Lin
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
18
|
Liu J, Chen Y, Stephens R, Cornea E, Goldman B, Gilmore JH, Gao W. Hippocampal functional connectivity development during the first two years indexes 4-year working memory performance. Cortex 2021; 138:165-177. [PMID: 33691225 PMCID: PMC8058274 DOI: 10.1016/j.cortex.2021.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/03/2020] [Accepted: 02/05/2021] [Indexed: 02/08/2023]
Abstract
The hippocampus is a key limbic region involved in higher-order cognitive processes including learning and memory. Although both typical and atypical functional connectivity patterns of the hippocampus have been well-studied in adults, the developmental trajectory of hippocampal connectivity during infancy and how it relates to later working memory performance remains to be elucidated. Here we used resting state fMRI (rsfMRI) during natural sleep to examine the longitudinal development of hippocampal functional connectivity using a large cohort (N = 202) of infants at 3 weeks (neonate), 1 year, and 2 years of age. Next, we used multivariate modeling to investigate the relationship between both cross-sectional and longitudinal growth in hippocampal connectivity and 4-year working memory outcome. Results showed robust local functional connectivity of the hippocampus in neonates with nearby limbic and subcortical regions, with dramatic maturation and increasing connectivity with key default mode network (DMN) regions resulting in adult-like topology of the hippocampal functional connectivity by the end of the first year. This pattern was stabilized and further consolidated by 2 years of age. Importantly, cross-sectional and longitudinal measures of hippocampal connectivity in the first year predicted subsequent behavioral measures of working memory at 4 years of age. Taken together, our findings provide insight into the development of hippocampal functional circuits underlying working memory during this early critical period.
Collapse
Affiliation(s)
- Janelle Liu
- Biomedical Imaging Research Institute, Department of Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Yuanyuan Chen
- Biomedical Imaging Research Institute, Department of Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Rebecca Stephens
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC, USA.
| | - Emil Cornea
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC, USA.
| | - Barbara Goldman
- FPG Child Development Institute and Department of Psychology & Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, USA.
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC, USA.
| | - Wei Gao
- Biomedical Imaging Research Institute, Department of Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA; David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Abstract
Prenatal alcohol exposure leads to alterations in cognition, behavior and underlying brain architecture. However, prior studies have not integrated structural and functional imaging data in children with prenatal alcohol exposure. The aim of this study was to characterize disruptions in both structural and functional brain network organization after prenatal alcohol exposure in very early life. A group of 11 neonates with prenatal alcohol exposure and 14 unexposed controls were investigated using diffusion weighted structural and resting state functional magnetic resonance imaging. Covariance networks were created using graph theoretical analyses for each data set, controlling for age and sex. Group differences in global hub arrangement and regional connectivity were determined using nonparametric permutation tests. Neonates with prenatal alcohol exposure and controls exhibited similar global structural network organization. However, global functional networks of neonates with prenatal alcohol exposure comprised of temporal and limbic hubs, while hubs were more distributed in controls representing an early default mode network. On a regional level, controls showed prominent structural and functional connectivity in parietal and occipital regions. Neonates with prenatal alcohol exposure showed regionally, predominant structural and functional connectivity in several subcortical regions and occipital regions. The findings suggest early functional disruption on a global and regional level after prenatal alcohol exposure and indicate suboptimal organization of functional networks. These differences likely underlie sensory dysregulation and behavioral difficulties in prenatal alcohol exposure.
Collapse
|
20
|
Radhakrishnan R, Elsaid NMH, Sadhasivam S, Reher TA, Hines AC, Yoder K, Saykin AJ, Wu YC. Resting state functional MRI in infants with prenatal opioid exposure-a pilot study. Neuroradiology 2021; 63. [PMID: 32978671 PMCID: PMC9162800 DOI: 10.1007/s00234-020-02552-3,] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
PURPOSE Exposure to prenatal opioids may adversely impact the developing brain networks. The aim of this pilot study was to evaluate alterations in amygdalar functional connectivity in human infants with prenatal opioid exposure. METHODS In this prospective IRB approved study, we performed resting state functional MRI (rs-fMRI) in 10 infants with prenatal opioid exposure and 12 infants without prenatal drug exposure at < 48 weeks corrected gestational age. Following standard preprocessing, we performed seed-based functional connectivity analysis with the right and left amygdala as the regions of interest after correcting for maternal depression and infant sex. We compared functional connectivity of the amygdala network between infants with and without prenatal opioid exposure. RESULTS There were significant differences in connectivity of the amygdala seed regions to the several cortical regions including the medial prefrontal cortex in infants who had prenatal opioid exposure when compared with opioid naïve infants. CONCLUSION This finding of increased amygdala functional connectivity in infants with in utero opioid exposure suggests a potential role of maternal opioid exposure on infants' altered amygdala function. This association with prenatal exposure needs to be replicated in future larger studies.
Collapse
Affiliation(s)
- Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Nahla M H Elsaid
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Thomas A Reher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Abbey C Hines
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Karmen Yoder
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Yu-Chien Wu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
21
|
Radhakrishnan R, Grecco G, Stolze K, Atwood B, Jennings SG, Lien IZ, Saykin AJ, Sadhasivam S. Neuroimaging in infants with prenatal opioid exposure: Current evidence, recent developments and targets for future research. J Neuroradiol 2021; 48:112-120. [PMID: 33065196 PMCID: PMC7979441 DOI: 10.1016/j.neurad.2020.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/29/2022]
Abstract
Prenatal opioid exposure (POE) has shown to be a risk factor for adverse long-term cognitive and behavioral outcomes in offspring. However, the neural mechanisms of these outcomes remain poorly understood. While preclinical and human studies suggest that these outcomes may be due to opioid-mediated changes in the fetal and early postnatal brain, other maternal, social, and environmental factors are also shown to play a role. Recent neuroimaging studies reveal brain alterations in children with POE. Early neuroimaging and novel methodology could provide an in vivo mechanistic understanding of opioid mediated alterations in developing brain. However, this is an area of ongoing research. In this review we explore recent imaging developments in POE, with emphasis on the neonatal and infant brain, and highlight some of the challenges of imaging the developing brain in this population. We also highlight evidence from animal models and imaging in older children and youth to understand areas where future research may be targeted in infants with POE.
Collapse
Affiliation(s)
- Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | - Gregory Grecco
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Brady Atwood
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Samuel G Jennings
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Izlin Z Lien
- Department of Pediatrics, Division of Neonatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | |
Collapse
|
22
|
Gao W, Chen Y, Cornea E, Goldman BD, Gilmore JH. Neonatal brain connectivity outliers identify over forty percent of IQ outliers at 4 years of age. Brain Behav 2020; 10:e01846. [PMID: 32945129 PMCID: PMC7749582 DOI: 10.1002/brb3.1846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Defining reliable brain markers for the prediction of abnormal behavioral outcomes remains an urgent but extremely challenging task in neuroscience research. This is particularly important for infant studies given the most dramatic brain and behavioral growth during infancy. METHODS In this study, we proposed a novel prediction scheme through abstracting individual newborn's whole-brain functional connectivity pattern to three outlier measures (Triple O) and tested the hypothesis that neonates identified as "brain outliers" based on Triple O were more likely to develop as IQ outliers at 4 years of age. Without need for training with behavioral data, Triple O represents a novel proof-of-concept approach to predict later IQ outcomes based on neonatal brain data. RESULTS Triple O correctly identified 42.1% true IQ outliers among a mixed cohort of 175 newborns with different term, twin, and maternal disorder statuses. Triple O also reached a high level of specificity (96.2%) and overall accuracy (90.3%). Further incorporating a demographic information indicator, the enhanced Triple O+ could further differentiate between high and low 4YR IQ outliers. Validation tests against seven independent reference samples revealed highly consistent results and a minimum sample size of ~50 for robust performance. CONCLUSIONS Considering that postnatal brain growth and various environmental factors likely also contribute to 4YR IQ, the fact that Triple O, based purely on neonatal functional connectivity data, could identify >40% of 4YR IQ outliers is striking. Together with the very high level of specificity, each outlier predicted by Triple O represents a meaningful risk but future efforts are needed to explore ways to identify the rest of outliers. Overall, with no need for training, a high level of robustness, and a minimal requirement on sample size, the proposed Triple O approach demonstrates great potential to predict later outlying IQ performances using neonatal functional connectivity data.
Collapse
Affiliation(s)
- Wei Gao
- Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Yuanyuan Chen
- Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emil Cornea
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Barbara D Goldman
- Department of Psychology and Neuroscience FPG Child Development Institute, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Resting state functional MRI in infants with prenatal opioid exposure-a pilot study. Neuroradiology 2020; 63:585-591. [PMID: 32978671 DOI: 10.1007/s00234-020-02552-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Exposure to prenatal opioids may adversely impact the developing brain networks. The aim of this pilot study was to evaluate alterations in amygdalar functional connectivity in human infants with prenatal opioid exposure. METHODS In this prospective IRB approved study, we performed resting state functional MRI (rs-fMRI) in 10 infants with prenatal opioid exposure and 12 infants without prenatal drug exposure at < 48 weeks corrected gestational age. Following standard preprocessing, we performed seed-based functional connectivity analysis with the right and left amygdala as the regions of interest after correcting for maternal depression and infant sex. We compared functional connectivity of the amygdala network between infants with and without prenatal opioid exposure. RESULTS There were significant differences in connectivity of the amygdala seed regions to the several cortical regions including the medial prefrontal cortex in infants who had prenatal opioid exposure when compared with opioid naïve infants. CONCLUSION This finding of increased amygdala functional connectivity in infants with in utero opioid exposure suggests a potential role of maternal opioid exposure on infants' altered amygdala function. This association with prenatal exposure needs to be replicated in future larger studies.
Collapse
|
24
|
Chen Y, Liu S, Salzwedel A, Stephens R, Cornea E, Goldman BD, Gilmore JH, Gao W. The Subgrouping Structure of Newborns with Heterogenous Brain-Behavior Relationships. Cereb Cortex 2020; 31:301-311. [PMID: 32946557 DOI: 10.1093/cercor/bhaa226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
The presence of heterogeneity/subgroups in infants and older populations against single-domain brain or behavioral measures has been previously characterized. However, few attempts have been made to explore heterogeneity at the brain-behavior relationship level. Such a hypothesis posits that different subgroups of infants may possess qualitatively different brain-behavior relationships that could ultimately contribute to divergent developmental outcomes even with relatively similar brain phenotypes. In this study, we aimed to explore such relationship-level heterogeneity and delineate the subgrouping structure of newborns with differential brain-behavior associations based on a typically developing sample of 81 infants with 3-week resting-state functional magnetic resonance imaging scans and 4-year intelligence quotient (IQ) measures. Our results not only confirmed the existence of relationship-level heterogeneity in newborns but also revealed divergent developmental outcomes associated with two subgroups showing similar brain functional connectivity but contrasting brain-behavior relationships. Importantly, further analyses unveiled an intriguing pattern that the subgroup with higher 4-year IQ outcomes possessed brain-behavior relationships that were congruent to their functional connectivity pattern in neonates while the subgroup with lower 4-year IQ not, providing potential explanations for the observed IQ differences. The characterization of heterogeneity at the brain-behavior relationship level may not only improve our understanding of the patterned intersubject variability during infancy but could also pave the way for future development of heterogeneity-inspired, personalized, subgroup-specific models for better prediction.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shuxin Liu
- Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,School of Educational Sciences, Minnan Normal University, Zhangzhou, Fujian 36300, China
| | - Andrew Salzwedel
- Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rebecca Stephens
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, USA
| | - Emil Cornea
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, USA
| | - Barbara D Goldman
- Department of Psychology, FPG Child Development Institute, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, USA
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wei Gao
- Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
25
|
Hayward DA, Pomares F, Casey KF, Ismaylova E, Levesque M, Greenlaw K, Vitaro F, Brendgen M, Rénard F, Dionne G, Boivin M, Tremblay RE, Booij L. Birth weight is associated with adolescent brain development: A multimodal imaging study in monozygotic twins. Hum Brain Mapp 2020; 41:5228-5239. [PMID: 32881198 PMCID: PMC7670633 DOI: 10.1002/hbm.25188] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 08/02/2020] [Accepted: 08/04/2020] [Indexed: 01/20/2023] Open
Abstract
Previous research has shown that the prenatal environment, commonly indexed by birth weight (BW), is a predictor of morphological brain development. We previously showed in monozygotic (MZ) twins associations between BW and brain morphology that were independent of genetics. In the present study, we employed a longitudinal MZ twin design to investigate whether variations in prenatal environment (as indexed by discordance in BW) are associated with resting‐state functional connectivity (rs‐FC) and with structural connectivity. We focused on the limbic and default mode networks (DMNs), which are key regions for emotion regulation and internally generated thoughts, respectively. One hundred and six healthy adolescent MZ twins (53 pairs; 42% male pairs) followed longitudinally from birth underwent a magnetic resonance imaging session at age 15. Graph theoretical analysis was applied to rs‐FC measures. TrackVis was used to determine track count as an indicator of structural connectivity strength. Lower BW twins had less efficient limbic network connectivity as compared to their higher BW co‐twin, driven by differences in the efficiency of the right hippocampus and right amygdala. Lower BW male twins had fewer tracks connecting the right hippocampus and right amygdala as compared to their higher BW male co‐twin. There were no associations between BW and the DMN. These findings highlight the possible role of unique prenatal environmental influences in the later development of efficient spontaneous limbic network connections within healthy individuals, irrespective of DNA sequence or shared environment.
Collapse
Affiliation(s)
- Dana A Hayward
- Sainte-Justine Hospital Research Centre, Montreal, Canada.,Department of Psychology, Concordia University, Montreal, Canada
| | - Florence Pomares
- Sainte-Justine Hospital Research Centre, Montreal, Canada.,Department of Psychology, Concordia University, Montreal, Canada
| | - Kevin F Casey
- Sainte-Justine Hospital Research Centre, Montreal, Canada.,Department of Psychology, Concordia University, Montreal, Canada
| | - Elmira Ismaylova
- Sainte-Justine Hospital Research Centre, Montreal, Canada.,Department of Psychology, Concordia University, Montreal, Canada
| | | | - Keelin Greenlaw
- Sainte-Justine Hospital Research Centre, Montreal, Canada.,Department of Psychology, Concordia University, Montreal, Canada
| | - Frank Vitaro
- Sainte-Justine Hospital Research Centre, Montreal, Canada.,School of Psychoeducation, University of Montreal, Montreal, Canada
| | - Mara Brendgen
- Department of Psychology, University of Quebec in Montreal, Montreal, Canada
| | - Felix Rénard
- Grenoble Hospital, University of Grenoble, Grenoble, France
| | - Ginette Dionne
- Department of Psychology, University Laval, Quebec, Canada
| | - Michel Boivin
- Department of Psychology, University Laval, Quebec, Canada
| | - Richard E Tremblay
- Sainte-Justine Hospital Research Centre, Montreal, Canada.,Department of Psychology and Pediatrics, University of Montreal, Montreal, Canada.,School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | - Linda Booij
- Sainte-Justine Hospital Research Centre, Montreal, Canada.,Department of Psychology, Concordia University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Psychiatry and Addiction, University of Montreal, Montreal, Canada
| |
Collapse
|
26
|
Salzwedel A, Chen G, Chen Y, Grewen K, Gao W. Functional dissection of prenatal drug effects on baby brain and behavioral development. Hum Brain Mapp 2020; 41:4789-4803. [PMID: 32779835 PMCID: PMC7643353 DOI: 10.1002/hbm.25158] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Prenatal drug exposure (PDE) is known to affect fetal brain development with documented long‐term consequences. Most studies of PDE effects on the brain are based on animal models. In this study, based on a large sample of 133 human neonates and leveraging a novel linear mixed‐effect model designed for intersubject variability analyses, we studied the effects of six prenatally exposed drugs (i.e., nicotine, alcohol, selective serotonin reuptake inhibitor, marijuana, cocaine, and opioids) on neonatal whole‐brain functional organization and compared them with five other critical nondrug variables (i.e., gestational age at birth/scan, sex, birth weight, and maternal depression). The behavioral implications were also examined. Magnitude‐wise, through summing across individual drug effects, our results highlighted ~5% of whole‐brain functional connections (FCs) affected by PDE, which was highly comparable with the combined effects of the five nond rug variables. Spatially, the detected PDE effects featured drug‐specific patterns with a common bias in higher‐order brain regions/networks. Regarding brain–behavioral relationships, the detected connections showing significant drug effects also demonstrated significant correlations with 3‐month behavioral outcomes. Further mediation analyses supported a mediation role of the detected brain FCs between PDE status and cognitive/language outcomes. Our findings of widespread, and spatially biased PDE effect patterns coupled with significant behavioral implications may hopefully stimulate more human‐based studies into effects of PDE on long‐term developmental outcomes.
Collapse
Affiliation(s)
- Andrew Salzwedel
- Department of Biomedical Sciences, Imaging, and Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gang Chen
- Scientific and Statistical Computing Core, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Yuanyuan Chen
- Department of Biomedical Sciences, Imaging, and Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Karen Grewen
- Department of Psychiatry, Neurobiology, and Psychology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wei Gao
- Department of Biomedical Sciences, Imaging, and Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
27
|
Cedillo-Pozos A, Ternovoy SK, Roldan-Valadez E. Imaging methods used in the assessment of environmental disease networks: a brief review for clinicians. Insights Imaging 2020; 11:18. [PMID: 32034587 PMCID: PMC7007482 DOI: 10.1186/s13244-019-0814-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/04/2019] [Indexed: 02/08/2023] Open
Abstract
Background Across the globe, diseases secondary to environmental exposures have been described, and it was also found that existing diseases have been modified by exposure to environmental chemicals or an environmental factor that has been found in their pathogenesis. The Institute of Medicine has shared a permanent concern related to the nations environmental health capacity since 1988. Main body Contemporary imaging methods in the last 15 years started reporting alterations in different human systems such as the central nervous system, cardiovascular system and pulmonary system among others; evidence suggests the existence of a human environmental disease network. The primary anatomic regions, affected by environmental diseases, recently assessed with imaging methods include Brain (lead exposure, cerebral stroke, pesticide neurotoxicity), uses MRI, DTI, carotid ultrasonography and MRS; Lungs (smoke inhalation, organophosphates poisoning) are mainly assessed with radiography; Gastrointestinal system (chronic inflammatory bowel disease), recent studies have reported the use of aortic ultrasound; Heart (myocardial infarction), its link to environmental diseased has been proved with carotid ultrasound; and Arteries (artery hypertension), the impairment of aortic mechanical properties has been revealed with the use of aortic and brachial ultrasound. Conclusions Environmental epidemiology has revealed that several organs and systems in the human body are targets of air pollutants. Current imaging methods that can assess the deleterious effects of pollutants includes a whole spectrum: radiography, US, CT and MRI. Future studies will help to reveal additional links among environmental disease networks.
Collapse
Affiliation(s)
- Aime Cedillo-Pozos
- Directorate of Research, Hospital General de Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico
| | - Sergey K Ternovoy
- Department of Radiology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.,A.L. Myasnikov Research Institute of Clinical Cardiology of National Medical Research Center of Cardiology of the Ministry of Health of Russia, Moscow, Russia
| | - Ernesto Roldan-Valadez
- Directorate of Research, Hospital General de Mexico "Dr. Eduardo Liceaga", Mexico City, Mexico. .,Department of Radiology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| |
Collapse
|
28
|
Functional Connectome of the Fetal Brain. J Neurosci 2019; 39:9716-9724. [PMID: 31685648 PMCID: PMC6891066 DOI: 10.1523/jneurosci.2891-18.2019] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 08/22/2019] [Accepted: 10/01/2019] [Indexed: 01/05/2023] Open
Abstract
Large-scale functional connectome formation and reorganization is apparent in the second trimester of pregnancy, making it a crucial and vulnerable time window in connectome development. Here we identified which architectural principles of functional connectome organization are initiated before birth, and contrast those with topological characteristics observed in the mature adult brain. A sample of 105 pregnant women participated in human fetal resting-state fMRI studies (fetal gestational age between 20 and 40 weeks). Connectome analysis was used to analyze weighted network characteristics of fetal macroscale brain wiring. We identified efficient network attributes, common functional modules, and high overlap between the fetal and adult brain network. Our results indicate that key features of the functional connectome are present in the second and third trimesters of pregnancy. Understanding the organizational principles of fetal connectome organization may bring opportunities to develop markers for early detection of alterations of brain function.SIGNIFICANCE STATEMENT The fetal to neonatal period is well known as a critical stage in brain development. Rapid neurodevelopmental processes establish key functional neural circuits of the human brain. Prenatal risk factors may interfere with early trajectories of connectome formation and thereby shape future health outcomes. Recent advances in MRI have made it possible to examine fetal brain functional connectivity. In this study, we evaluate the network topography of normative functional network development during connectome genesis in utero Understanding the developmental trajectory of brain connectivity provides a basis for understanding how the prenatal period shapes future brain function and disease dysfunction.
Collapse
|
29
|
de Water E, Curtin P, Zilverstand A, Sjödin A, Bonilla A, Herbstman JB, Ramirez J, Margolis AE, Bansal R, Whyatt RM, Peterson BS, Factor-Litvak P, Horton MK. A preliminary study on prenatal polybrominated diphenyl ether serum concentrations and intrinsic functional network organization and executive functioning in childhood. J Child Psychol Psychiatry 2019; 60:1010-1020. [PMID: 30882909 PMCID: PMC7105394 DOI: 10.1111/jcpp.13040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND The prenatal period is a period of vulnerability during which neurotoxic exposures exert persistent changes in brain development and behavior. Polybrominated diphenyl ethers (PBDEs), used as flame retardants in commercial products, are known to be developmental neurotoxicants. PBDEs were phased out of use in the United States a decade ago, but exposure remains widespread due to their release from existing products and biopersistence. Despite consistent animal and epidemiological evidence of developmental neurotoxicity, the neural substrates linking prenatal PBDE serum concentrations to impaired neurodevelopment are poorly understood. METHODS In the present study, we used resting state functional magnetic resonance imaging (fMRI) to examine associations between prenatal PBDE concentrations measured in maternal serum and intrinsic functional network organization (i.e., global and local efficiency; estimated using a graph-theoretical approach) in 5-year-old children (n = 34). We explored whether PBDE serum concentrations were associated with executive functioning (EF) assessed using a parent-report questionnaire (BRIEF-P) (n = 106) and whether changes in intrinsic functional network organization linked the association between prenatal PBDE serum concentrations and EF problems. RESULTS Children with higher prenatal PBDE serum concentrations showed: (a) increased global efficiency of brain areas involved in visual attention (e.g., inferior occipital gyrus) (β's = .01, FDR-corrected p's ≤ .05); (b) more reported EF problems (β's = .001, FDR-corrected p's ≤ .05). Higher global efficiency of brain areas involved in visual attention was associated with more EF problems (β's = .01, FDR-corrected p's < .05). CONCLUSIONS Intrinsic functional network organization of visual attention brain areas linked prenatal PBDE concentrations to EF problems in childhood. Visual attention may contribute to the development of higher-order cognitive functions, such as EF, which could be explored in future studies.
Collapse
Affiliation(s)
- Erik de Water
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul Curtin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Zilverstand
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| | - Andreas Sjödin
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Anny Bonilla
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie B. Herbstman
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia Center for Children’s Environmental Health, Columbia University, New York, NY, USA
| | - Judyth Ramirez
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia Center for Children’s Environmental Health, Columbia University, New York, NY, USA
| | - Amy E. Margolis
- The Division of Child and Adolescent Psychiatry in the Department of Psychiatry, The New York State Psychiatric Institute and the College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Ravi Bansal
- Institute for the Developing Mind, Children’s Hospital Los Angeles and the Department of Psychiatry at the Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robin M. Whyatt
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia Center for Children’s Environmental Health, Columbia University, New York, NY, USA
| | - Bradley S. Peterson
- Institute for the Developing Mind, Children’s Hospital Los Angeles and the Department of Psychiatry at the Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Megan K. Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
30
|
Shi F, Salzwedel AP, Lin W, Gilmore JH, Gao W. Functional Brain Parcellations of the Infant Brain and the Associated Developmental Trends. Cereb Cortex 2019; 28:1358-1368. [PMID: 28334317 DOI: 10.1093/cercor/bhx062] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Indexed: 01/17/2023] Open
Abstract
Resting-state functional connectivity studies have dramatically improved our understanding of the early human brain functional development during the past decade. However, one emerging problem that could potentially impede future progresses in the field is the definition of regions of interest (ROI), since it is well known that functional connectivity estimation can be seriously contaminated by within-ROI signal heterogeneity. In this study, based on a large-scale rsfMRI data set in human infants (230 neonates, 143 1-year olds, and 107 2-year olds), we aimed to derive a set of anatomically constrained, infant-specific functional brain parcellations using functional connectivity-based clustering. Our results revealed significantly higher levels of signal homogeneity within the newly defined functional parcellations compared with other schemes. Importantly, the global functional connectivity patterns associated with the newly defined functional subunits demonstrated significantly increasing levels of differentiation with age, confirming increasing levels of local specialization. Subsequent whole brain connectivity analysis revealed intriguing patterns of regional-level functional connectivity developments and system-level hub redistribution during infancy. Overall, the newly derived infant-specific functional brain parcellations and the associated novel developmental patterns will likely prove valuable for future early developmental studies using the functional connectivity technique.
Collapse
Affiliation(s)
- Feng Shi
- Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew P Salzwedel
- Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Weili Lin
- Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wei Gao
- Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute (BIRI), Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
31
|
Gut microbiome and brain functional connectivity in infants-a preliminary study focusing on the amygdala. Psychopharmacology (Berl) 2019; 236:1641-1651. [PMID: 30604186 PMCID: PMC6599471 DOI: 10.1007/s00213-018-5161-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/21/2018] [Indexed: 12/26/2022]
Abstract
Recently, there has been a surge of interest in the possibility that microbial communities inhabiting the human gut could affect cognitive development and increase risk for mental illness via the "microbiome-gut-brain axis." Infancy likely represents a critical period for the establishment of these relationships, as it is the most dynamic stage of postnatal brain development and a key period in the maturation of the microbiome. Indeed, recent reports indicate that characteristics of the infant gut microbiome are associated with both temperament and cognitive performance. The neural circuits underlying these relationships have not yet been delineated. To address this gap, resting-state fMRI scans were acquired from 39 1-year-old human infants who had provided fecal samples for identification and relative quantification of bacterial taxa. Measures of alpha diversity were generated and tested for associations with measures of functional connectivity. Primary analyses focused on the amygdala as manipulation of the gut microbiota in animal models alters the structure and neurochemistry of this brain region. Secondary analyses explored functional connectivity of nine canonical resting-state functional networks. Alpha diversity was significantly associated with functional connectivity between the amygdala and thalamus and between the anterior cingulate cortex and anterior insula. These regions play an important role in processing/responding to threat. Alpha diversity was also associated with functional connectivity between the supplementary motor area (SMA, representing the sensorimotor network) and the inferior parietal lobule (IPL). Importantly, SMA-IPL connectivity also related to cognitive outcomes at 2 years of age, suggesting a potential pathway linking gut microbiome diversity and cognitive outcomes during infancy. These results provide exciting new insights into the gut-brain axis during early human development and should stimulate further studies into whether microbiome-associated changes in brain circuitry influence later risk for psychopathology.
Collapse
|
32
|
Morie KP, Crowley MJ, Mayes LC, Potenza MN. Prenatal drug exposure from infancy through emerging adulthood: Results from neuroimaging. Drug Alcohol Depend 2019; 198:39-53. [PMID: 30878766 PMCID: PMC6688747 DOI: 10.1016/j.drugalcdep.2019.01.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 11/28/2022]
Abstract
Prenatal drug exposure may have important repercussions across the lifespan for cognition and behavior. While alcohol is a recognized teratogen, the influences of other substances may also be substantial. The neural underpinnings of the influences of prenatal drug exposure have been examined using longitudinal approaches and multiple imaging techniques. Here we review the existing literature on the neural correlates of prenatal drug exposure. We focused the review on studies that have employed functional neuroimaging and electroencephalography and on substances other than alcohol. We also framed the review through the lens of four developmental life stages (infancy, childhood, adolescence and emerging adulthood). We included papers that have examined any drug use, including tobacco, opiates, cocaine, marijuana, methamphetamines, or polysubstance use. Data suggest that prenatal drug exposure has long-lasting, deleterious influences on cognition and reward processing in infancy and childhood that persist into adolescence and emerging adulthood and may underlie some behavioral tendencies, such as increased externalizing and risk-taking behaviors, seen in these groups.
Collapse
Affiliation(s)
- Kristen P. Morie
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA,Corresponding author at: Department of Psychiatry, Yale University School of Medicine, 300 George St., #901, New Haven, CT, 06510, USA. (K.P. Morie)
| | - Michael J. Crowley
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Linda C. Mayes
- Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA,Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06510, USA,Department of Psychology, Yale University, New Haven, CT, 06511, USA
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA,Connecticut Mental Health Center, New Haven, CT, 06519, USA,Connecticut Council on Problem Gambling, Wethersfield, CT, 06109, USA
| |
Collapse
|
33
|
Gao W, Grewen K, Knickmeyer RC, Qiu A, Salzwedel A, Lin W, Gilmore JH. A review on neuroimaging studies of genetic and environmental influences on early brain development. Neuroimage 2019; 185:802-812. [PMID: 29673965 PMCID: PMC6191379 DOI: 10.1016/j.neuroimage.2018.04.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 12/11/2022] Open
Abstract
The past decades witnessed a surge of interest in neuroimaging study of normal and abnormal early brain development. Structural and functional studies of normal early brain development revealed massive structural maturation as well as sequential, coordinated, and hierarchical emergence of functional networks during the infancy period, providing a great foundation for the investigation of abnormal early brain development mechanisms. Indeed, studies of altered brain development associated with either genetic or environmental risks emerged and thrived. In this paper, we will review selected studies of genetic and environmental risks that have been relatively more extensively investigated-familial risks, candidate risk genes, and genome-wide association studies (GWAS) on the genetic side; maternal mood disorders and prenatal drug exposures on the environmental side. Emerging studies on environment-gene interactions will also be reviewed. Our goal was not to perform an exhaustive review of all studies in the field but to leverage some representative ones to summarize the current state, point out potential limitations, and elicit discussions on important future directions.
Collapse
Affiliation(s)
- Wei Gao
- Biomedical Imaging Research Institute (BIRI), Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, CA, United States; Department of Medicine, University of California, Los Angeles, CA, United States.
| | - Karen Grewen
- Department of Psychiatry, Neurobiology, and Psychology, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Rebecca C Knickmeyer
- Department of Psychiatry, University of North Carolina at Chapel Hill, N.C, United States
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Andrew Salzwedel
- Biomedical Imaging Research Institute (BIRI), Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, CA, United States
| | - Weili Lin
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, NC, United States
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina at Chapel Hill, N.C, United States
| |
Collapse
|
34
|
Moreno-Rius J. The Cerebellum, THC, and Cannabis Addiction: Findings from Animal and Human Studies. THE CEREBELLUM 2019; 18:593-604. [DOI: 10.1007/s12311-018-0993-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
35
|
Gilmore JH, Knickmeyer RC, Gao W. Imaging structural and functional brain development in early childhood. Nat Rev Neurosci 2018; 19:123-137. [PMID: 29449712 PMCID: PMC5987539 DOI: 10.1038/nrn.2018.1] [Citation(s) in RCA: 491] [Impact Index Per Article: 81.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In humans, the period from term birth to ∼2 years of age is characterized by rapid and dynamic brain development and plays an important role in cognitive development and risk of disorders such as autism and schizophrenia. Recent imaging studies have begun to delineate the growth trajectories of brain structure and function in the first years after birth and their relationship to cognition and risk of neuropsychiatric disorders. This Review discusses the development of grey and white matter and structural and functional networks, as well as genetic and environmental influences on early-childhood brain development. We also discuss initial evidence regarding the usefulness of early imaging biomarkers for predicting cognitive outcomes and risk of neuropsychiatric disorders.
Collapse
Affiliation(s)
- John H Gilmore
- Department of Psychiatry, CB# 7160, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Rebecca C Knickmeyer
- Department of Psychiatry, CB# 7160, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Wei Gao
- Biomedical Imaging Research Institute, Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
36
|
Arichi T, Whitehead K, Barone G, Pressler R, Padormo F, Edwards AD, Fabrizi L. Localization of spontaneous bursting neuronal activity in the preterm human brain with simultaneous EEG-fMRI. eLife 2017; 6. [PMID: 28893378 PMCID: PMC5595428 DOI: 10.7554/elife.27814] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 08/03/2017] [Indexed: 12/21/2022] Open
Abstract
Electroencephalographic recordings from the developing human brain are characterized by spontaneous neuronal bursts, the most common of which is the delta brush. Although similar events in animal models are known to occur in areas of immature cortex and drive their development, their origin in humans has not yet been identified. Here, we use simultaneous EEG-fMRI to localise the source of delta brush events in 10 preterm infants aged 32–36 postmenstrual weeks. The most frequent patterns were left and right posterior-temporal delta brushes which were associated in the left hemisphere with ipsilateral BOLD activation in the insula only; and in the right hemisphere in both the insular and temporal cortices. This direct measure of neural and hemodynamic activity shows that the insula, one of the most densely connected hubs in the developing cortex, is a major source of the transient bursting events that are critical for brain maturation.
Collapse
Affiliation(s)
- Tomoki Arichi
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, United Kingdom.,Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Kimberley Whitehead
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Giovanni Barone
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, United Kingdom.,Department of Pediatrics, Catholic University of Sacred Heart, Rome, Italy
| | - Ronit Pressler
- Clinical Neurosciences, UCL-Institute of Child Health, London, United Kingdom
| | - Francesco Padormo
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, United Kingdom.,Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - A David Edwards
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, United Kingdom.,Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Lorenzo Fabrizi
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
37
|
Cao M, Huang H, He Y. Developmental Connectomics from Infancy through Early Childhood. Trends Neurosci 2017; 40:494-506. [PMID: 28684174 PMCID: PMC5975640 DOI: 10.1016/j.tins.2017.06.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022]
Abstract
The human brain undergoes rapid growth in both structure and function from infancy through early childhood, and this significantly influences cognitive and behavioral development in later life. A newly emerging research framework, developmental connectomics, provides unprecedented opportunities for exploring the developing brain through non-invasive mapping of structural and functional connectivity patterns. Within this framework, we review recent neuroimaging and neurophysiological studies investigating connectome development from 20 postmenstrual weeks to 5 years of age. Specifically, we highlight five fundamental principles of brain network development during the critical first years of life, emphasizing strengthened segregation/integration balance, a remarkable hierarchical order from primary to higher-order regions, unparalleled structural and functional maturations, substantial individual variability, and high vulnerability to risk factors and developmental disorders.
Collapse
Affiliation(s)
- Miao Cao
- National Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Hao Huang
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yong He
- National Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
38
|
Gao W, Lin W, Grewen K, Gilmore JH. Functional Connectivity of the Infant Human Brain: Plastic and Modifiable. Neuroscientist 2016; 23:169-184. [PMID: 26929236 PMCID: PMC5145769 DOI: 10.1177/1073858416635986] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Infancy is a critical and immensely important period in human brain development. Subtle changes during this stage may be greatly amplified with the unfolding of different developmental processes, exerting far-reaching consequences. Studies of the structure and behavioral manifestations of the infant brain are fruitful. However, the specific functional brain mechanisms that enable the execution of different behaviors remained elusive until the advent of functional connectivity fMRI (fcMRI), which provides an unprecedented opportunity to probe the infant functional brain development in vivo. Since its inception, a burgeoning field of infant brain functional connectivity study has emerged and thrived during the past decade. In this review, we describe (1) findings of normal development of functional connectivity networks and their relationships to behaviors and (2) disruptions of the normative functional connectivity development due to identifiable genetic and/or environmental risk factors during the first 2 years of human life. Technical considerations of infant fcMRI are also provided. It is our hope to consolidate previous findings so that the field can move forward with a clearer picture toward the ultimate goal of fcMRI-based objective methods for early diagnosis/identification of risks and evaluation of early interventions to optimize developing functional connectivity networks in this critical developmental window.
Collapse
Affiliation(s)
- Wei Gao
- 1 Biomedical Imaging Research Institute (BIRI), Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Weili Lin
- 2 Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, NC, USA
| | - Karen Grewen
- 3 Departments of Psychiatry, Neurobiology, and Psychology, University of North Carolina at Chapel Hill, NC, USA
| | - John H Gilmore
- 4 Department of Psychiatry, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|
39
|
Erdoğan SB, Tong Y, Hocke LM, Lindsey KP, deB Frederick B. Correcting for Blood Arrival Time in Global Mean Regression Enhances Functional Connectivity Analysis of Resting State fMRI-BOLD Signals. Front Hum Neurosci 2016; 10:311. [PMID: 27445751 PMCID: PMC4923135 DOI: 10.3389/fnhum.2016.00311] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/08/2016] [Indexed: 12/12/2022] Open
Abstract
Resting state functional connectivity analysis is a widely used method for mapping intrinsic functional organization of the brain. Global signal regression (GSR) is commonly employed for removing systemic global variance from resting state BOLD-fMRI data; however, recent studies have demonstrated that GSR may introduce spurious negative correlations within and between functional networks, calling into question the meaning of anticorrelations reported between some networks. In the present study, we propose that global signal from resting state fMRI is composed primarily of systemic low frequency oscillations (sLFOs) that propagate with cerebral blood circulation throughout the brain. We introduce a novel systemic noise removal strategy for resting state fMRI data, “dynamic global signal regression” (dGSR), which applies a voxel-specific optimal time delay to the global signal prior to regression from voxel-wise time series. We test our hypothesis on two functional systems that are suggested to be intrinsically organized into anticorrelated networks: the default mode network (DMN) and task positive network (TPN). We evaluate the efficacy of dGSR and compare its performance with the conventional “static” global regression (sGSR) method in terms of (i) explaining systemic variance in the data and (ii) enhancing specificity and sensitivity of functional connectivity measures. dGSR increases the amount of BOLD signal variance being modeled and removed relative to sGSR while reducing spurious negative correlations introduced in reference regions by sGSR, and attenuating inflated positive connectivity measures. We conclude that incorporating time delay information for sLFOs into global noise removal strategies is of crucial importance for optimal noise removal from resting state functional connectivity maps.
Collapse
Affiliation(s)
- Sinem B Erdoğan
- McLean Imaging Center, McLean HospitalBelmont, MA, USA; Department of Psychiatry, Harvard Medical SchoolBoston, MA, USA
| | - Yunjie Tong
- McLean Imaging Center, McLean HospitalBelmont, MA, USA; Department of Psychiatry, Harvard Medical SchoolBoston, MA, USA
| | - Lia M Hocke
- Department of Radiology, University of Calgary Calgary, AB, Canada
| | - Kimberly P Lindsey
- McLean Imaging Center, McLean HospitalBelmont, MA, USA; Department of Psychiatry, Harvard Medical SchoolBoston, MA, USA
| | - Blaise deB Frederick
- McLean Imaging Center, McLean HospitalBelmont, MA, USA; Department of Psychiatry, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
40
|
Salzwedel AP, Grewen KM, Goldman BD, Gao W. Thalamocortical functional connectivity and behavioral disruptions in neonates with prenatal cocaine exposure. Neurotoxicol Teratol 2016; 56:16-25. [PMID: 27242332 DOI: 10.1016/j.ntt.2016.05.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/26/2016] [Accepted: 05/26/2016] [Indexed: 12/20/2022]
Abstract
Prenatal cocaine exposure (PCE) affects neurobehavioral development, however, disentangling direct drug-related mechanisms from contextual effects (e.g., socioeconomic status) has proven challenging in humans. The effects of environmental confounds are minimal immediately after birth thus we aimed to delineate neurobehavioral correlates of PCE in a large cohort of neonates (2-6weeks of age, N=152) with and without drug exposure using resting state functional magnetic resonance imaging (rsfMRI) and developmental assessments at 3months with the Bayley Scales of Infant & Toddler Development, 3rd edition. The cohort included healthy controls and neonates with similar poly-drug exposure±cocaine. We focused on the thalamus given its critical importance in early brain development and its unique positioning in the dopamine system. Our results revealed PCE-related hyper-connectivity between the thalamus and frontal regions and a drug-common hypo-connective signature between the thalamus and motor-related regions. PCE-specific neonatal thalamo-frontal connectivity was inversely related to cognitive and fine motor scores and thalamo-motor connectivity showed a positive relationship with composite (gross plus fine) motor scores. Finally, cocaine by selective-serotonin-reuptake-inhibitor (SSRI) interactions were detected, suggesting the combined use of these drugs during pregnancy could have additional consequences on fetal development. Overall, our findings provide the first delineation of PCE-related disruptions of thalamocortical functional connectivity, neurobehavioral correlations, and drug-drug interactions during infancy.
Collapse
Affiliation(s)
- Andrew P Salzwedel
- Biomedical Imaging Research Institute (BIRI), Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, USA; University of North Carolina Chapel Hill, Department of Radiology and Biomedical Research Imaging Center, Chapel Hill, NC 27599, USA
| | - Karen M Grewen
- University of North Carolina Chapel Hill, Department of Psychiatry, Neurobiology, and Psychology, Chapel Hill, NC 27599, USA.
| | - Barbara D Goldman
- University of North Carolina Chapel Hill, Department of Psychology and Neuroscience, FPG Child Development Institute, Chapel Hill, NC 27599, USA
| | - Wei Gao
- Biomedical Imaging Research Institute (BIRI), Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, USA; University of North Carolina Chapel Hill, Department of Radiology and Biomedical Research Imaging Center, Chapel Hill, NC 27599, USA.
| |
Collapse
|