1
|
Dickinson A, McDonald N, Dapretto M, Campos E, Senturk D, Jeste S. Accelerated Infant Brain Rhythm Maturation in Autism. Dev Sci 2025; 28:e13593. [PMID: 39704490 DOI: 10.1111/desc.13593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 12/21/2024]
Abstract
Electroencephalography (EEG) captures characteristic oscillatory shifts in infant brain rhythms over the first year of life, offering unique insights into early functional brain development and potential markers for detecting neural differences associated with autism. This study used functional principal component analysis (FPCA) to derive dynamic markers of spectral maturation from task-free EEG recordings collected at 3, 6, 9, and 12 months from 87 infants, 51 of whom were at higher likelihood of developing autism due to an older sibling diagnosed with the condition. FPCA revealed three principal components explaining over 96% of the variance in infant power spectra, with power increases between 6 and 9 Hz (FPC1) representing the most significant age-related trend, accounting for more than 71% of the variance. Notably, this oscillatory change occurred at a faster rate in infants later diagnosed with autism, indicated by a steeper trajectory of FPC1 scores between 3 and 12 months (p < 0.001). Age-related spectral changes were consistent regardless of familial likelihood status, suggesting that differences in oscillatory timing are associated with autism outcomes rather than genetic predisposition. These findings indicate that while the typical sequence of oscillatory maturation is preserved in autism, the timing of these changes is altered, underscoring the critical role of timing in autism pathophysiology and the development of potential screening tools.
Collapse
Affiliation(s)
- Abigail Dickinson
- Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Nicole McDonald
- Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Mirella Dapretto
- Ahmanson-Lovelace Brain Mapping Center, University of California, Los Angeles, California, USA
| | - Emilie Campos
- UCLA Department of Biostatistics, Center for Health Sciences, University of California, Los Angeles, California, USA
| | - Damla Senturk
- UCLA Department of Biostatistics, Center for Health Sciences, University of California, Los Angeles, California, USA
| | - Shafali Jeste
- Division of Neurology and Neurological Institute, The Children's Hospital of Los Angeles, Los Angeles, California, USA
| |
Collapse
|
2
|
Quesada JA, Sánchez-Ferrer F, López-Pineda A. Autism Spectrum Disorder and Associated Factors in Children in Spain, 2017: Population-Based Cross-Sectional Study. Matern Child Health J 2024; 28:2051-2059. [PMID: 39424761 DOI: 10.1007/s10995-024-04007-0] [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] [Accepted: 09/27/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVES Autism spectrum disorder (ASD) is a neurodevelopmental disorder with biological, multicausal and polygenic origins. The true prevalence of ASD has not been clearly established. The aim of this study was to estimate the prevalence of ASD in children aged 3 to 14 years in Spain and to analyze the factors associated with it. METHODS A cross-sectional observational study using data from the 2017 National Survey of Health in Minors in Spain. Primary outcome was the diagnosis of ASD, and sociodemographic, behavioral, health-related, the use of health services, household and medication use variables was analyzed. Multivariable logistic regression model with a correction for modelling rare events was fitted. Complex sampling was undertaken, using the survey elevation factor in the analysis. RESULTS A total of 4409 children were included, and there were 26 children with ASD, for a prevalence of 0.59%, representing 29,143 children with ASD. Factors significantly associated were male sex, having visited a psychologist, and/or a speech therapist in the past year, presenting probable problems with peers, antisocial behavior, taking antibiotics and taking other medications. CONCLUSION The findings of this study may be useful to inform health policies and develop strategic plans to identify and address the needs of children with ASD.
Collapse
Affiliation(s)
- José Antonio Quesada
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Alicante, Spain
- Atenea Research Group, Foundation for the Promotion of Health and Biomedical Research, Alicante, Spain
| | - Francisco Sánchez-Ferrer
- GRINCAVA Research Group. Clinical Medicine Department, University Miguel Hernández de Elche, Ctra. Nnal. 332, s/n, Alicante, Elche, 03202, Spain.
- Pediatrics Department, University Hospital San Juan de Alicante, Alicante, Spain.
| | - Adriana López-Pineda
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Alicante, Spain
- Atenea Research Group, Foundation for the Promotion of Health and Biomedical Research, Alicante, Spain
- GRINCAVA Research Group. Clinical Medicine Department, University Miguel Hernández de Elche, Ctra. Nnal. 332, s/n, Alicante, Elche, 03202, Spain
| |
Collapse
|
3
|
Wang W, Liu Z, Peng D, Lin GN, Wang Z. Genomic insights into genes expressed specifically during infancy highlight their dominant influence on the neuronal system. BMC Genomics 2024; 25:1012. [PMID: 39472790 PMCID: PMC11520499 DOI: 10.1186/s12864-024-10911-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Elucidating the dynamics of gene expression across developmental stages, including the genomic characteristics of brain expression during infancy, is pivotal in deciphering human psychiatric and neurological disorders and providing insights into developmental disorders. RESULTS Leveraging comprehensive human GWAS associations with temporal and spatial brain expression data, we discovered a distinctive co-expression cluster comprising 897 genes highly expressed specifically during infancy, enriched in functions related to the neuronal system. This gene cluster notably harbors the highest ratio of genes linked to psychiatric and neurological disorders. Through computational analysis, MYT1L emerged as a potential central transcription factor governing these genes. Remarkably, the infancy-specific expressed genes, including SYT1, exhibit prominent colocalization within human accelerated regions. Additionally, chromatin state analysis unveiled prevalent epigenetic markers associated with enhancer-specific modifications. In addition, this cluster of genes has demonstrated to be specifically highly expressed in cell-types including excitatory neurons, medial ganglionic eminence and caudal ganglionic eminence. CONCLUSIONS This study comprehensively characterizes the genomics and epigenomics of genes specifically expressed during infancy, identifying crucial hub genes and transcription factors. These findings offer valuable insights into early detection strategies and interventions for psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Weidi Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zhe Liu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Daihui Peng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Guan Ning Lin
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Zhen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
- Shanghai Intelligent Psychological Evaluation and Intervention Engineering Technology Research Center, Shanghai, 200030, China.
| |
Collapse
|
4
|
Xiong W, Li X, Huang X, Xu J, Qu Z, Su Y, Li Y, Han Y, Cui T, Zhang X. Impaired motor and social skill development in infancy predict high autistic traits in toddlerhood. Neuroscience 2024; 558:114-121. [PMID: 39168171 DOI: 10.1016/j.neuroscience.2024.08.024] [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: 02/01/2024] [Revised: 07/26/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder. Early diagnosis in the critical period is important for ASD children. Recent studies of neurodevelopmental behavioral features and joint attention in high-risk infants showed there are some special cues which can distinguish ASD from typical development infant. But the findings of high-risk population may not be applicable to the general population. It is necessary to "analogically" study the potential warning traits of ASD in infancy in the general population. We did a nested case-control study from June 2019 to November 2022 in Tianjin, China, including 76 general infants whom completed the neurodevelopmental evaluation, the Checklist for Autism in Toddlers-23 (CHAT-23) screening, and eye tracking task. Social behavior quotient in infancy was negatively correlated to CHAT-23 total scores in toddlerhood. Social behavior quotient in infancy was positively correlated to initiating joint attention in toddlerhood. Regression model showed that high fine motor scale and social behaviour scale quotient in infancy were associated with an decreased risk of the total score of CHAT-23 ≥ 2 in toddlerhood. The Receiver operating characteristic curve showed the social behaviour in infancy alone and the combination of fine motor and social behaviour in infancy contributed to auxiliary diagnosis of higher level of autistic traits in toddlerhood. These findings suggest that Impaired development of fine motor and social behavior in infancy are potential warning features of high autistic traits in general population.
Collapse
Affiliation(s)
- Wenjuan Xiong
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xinyu Li
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaoqing Huang
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jinghan Xu
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Zhiyi Qu
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yuanyuan Su
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yin Li
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yu Han
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Tingkai Cui
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xin Zhang
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
5
|
Megari K, Frantzezou CK, Polyzopoulou ZA, Tzouni SK. Neurocognitive features in childhood & adulthood in autism spectrum disorder: A neurodiversity approach. Int J Dev Neurosci 2024; 84:471-499. [PMID: 38953464 DOI: 10.1002/jdn.10356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/30/2024] [Accepted: 06/10/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVES Autism spectrum disorder (ASD) is a neurodevelopmental disorder with a diverse profile of cognitive functions. Heterogeneity is observed among both baseline and comorbid features concerning the diversity of neuropathology in autism. Symptoms vary depending on the developmental stage, level of severity, or comorbidity with other medical or psychiatric diagnoses such as intellectual disability, epilepsy, and anxiety disorders. METHOD The neurodiversity movement does not face variations in neurological and cognitive development in ASD as deficits but as normal non-pathological human variations. Thus, ASD is not identified as a neurocognitive pathological disorder that deviates from the typical, but as a neuro-individuality, a normal manifestation of a neurobiological variation within the population. RESULTS In this light, neurodiversity is described as equivalent to any other human variation, such as ethnicity, gender, or sexual orientation. This review will provide insights about the neurodiversity approach in children and adults with ASD. Using a neurodiversity approach can be helpful when working with children who have autism spectrum disorder (ASD). DISCUSSION This method acknowledges and values the various ways that people with ASD interact with one another and experience the world in order to embrace the neurodiversity approach when working with children with ASD.
Collapse
Affiliation(s)
- Kalliopi Megari
- Department of Psychology, CITY College, University of York, Europe Campus, Thessaloniki, Greece
| | | | - Zoi A Polyzopoulou
- Department of Psychology, University of Western Macedonia, Florina, Greece
| | - Stella K Tzouni
- Department of Psychology, University of Western Macedonia, Florina, Greece
| |
Collapse
|
6
|
Al-Beltagi M, Saeed NK, Bediwy AS, Bediwy EA, Elbeltagi R. Decoding the genetic landscape of autism: A comprehensive review. World J Clin Pediatr 2024; 13:98468. [PMID: 39350903 PMCID: PMC11438927 DOI: 10.5409/wjcp.v13.i3.98468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/30/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by heterogeneous symptoms and genetic underpinnings. Recent advancements in genetic and epigenetic research have provided insights into the intricate mechanisms contributing to ASD, influencing both diagnosis and therapeutic strategies. AIM To explore the genetic architecture of ASD, elucidate mechanistic insights into genetic mutations, and examine gene-environment interactions. METHODS A comprehensive systematic review was conducted, integrating findings from studies on genetic variations, epigenetic mechanisms (such as DNA methylation and histone modifications), and emerging technologies [including Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 and single-cell RNA sequencing]. Relevant articles were identified through systematic searches of databases such as PubMed and Google Scholar. RESULTS Genetic studies have identified numerous risk genes and mutations associated with ASD, yet many cases remain unexplained by known factors, suggesting undiscovered genetic components. Mechanistic insights into how these genetic mutations impact neural development and brain connectivity are still evolving. Epigenetic modifications, particularly DNA methylation and non-coding RNAs, also play significant roles in ASD pathogenesis. Emerging technologies like CRISPR-Cas9 and advanced bioinformatics are advancing our understanding by enabling precise genetic editing and analysis of complex genomic data. CONCLUSION Continued research into the genetic and epigenetic underpinnings of ASD is crucial for developing personalized and effective treatments. Collaborative efforts integrating multidisciplinary expertise and international collaborations are essential to address the complexity of ASD and translate genetic discoveries into clinical practice. Addressing unresolved questions and ethical considerations surrounding genetic research will pave the way for improved diagnostic tools and targeted therapies, ultimately enhancing outcomes for individuals affected by ASD.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Alghrabia, Tanta 31511, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 12, Bahrain
- Medical Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Muharraq, Busaiteen 15503, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonology, Faculty of Medicine, Tanta University, Alghrabia, Tanta 31527, Egypt
- Department of Pulmonology, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| | - Eman A Bediwy
- Internal Medicine, Faculty of Medicine, Tanta University, Algharbia, Tanta 31527, Egypt
| | - Reem Elbeltagi
- Department of Medicine, The Royal College of Surgeons in Ireland-Bahrain, Muharraq, Busiateen 15503, Bahrain
| |
Collapse
|
7
|
Narvekar N, Carter Leno V, Pasco G, Begum Ali J, Johnson MH, Charman T, Jones EJH. The roles of sensory hyperreactivity and hyporeactivity in understanding infant fearfulness and emerging autistic traits. J Child Psychol Psychiatry 2024; 65:1022-1036. [PMID: 38172076 DOI: 10.1111/jcpp.13941] [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] [Accepted: 10/31/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Existing evidence indicates that atypical sensory reactivity is a core characteristic of autism, and has been linked to both anxiety (and its putative infant precursor of fearfulness) and repetitive behaviours. However, most work has used cross-sectional designs and not considered the differential roles of hyperreactivity and hyporeactivity to sensory inputs, and is thus limited in specificity. METHODS 161 infants with and without an elevated likelihood of developing autism and attention-deficit hyperactivity disorder (ADHD) were followed from 10 to 36 months of age. Parents rated an infant precursor of later anxiety (fearfulness) using the Infant Behaviour Questionnaire at 10 and 14 months, and the Early Childhood Behavioural Questionnaire at 24 months, and sensory hyperreactivity and hyporeactivity at 10, 14 and 24 months using the Infant Toddler Sensory Profile. Domains of autistic traits (restrictive and repetitive behaviours; RRB, and social communication interaction, SCI) were assessed using the parent-rated Social Responsiveness Scale at 36 months. Cross-lagged models tested (a) paths between fearfulness and hyperreactivity at 10-24 months, and from fearfulness and hyperreactivity to later autism traits, (b) the specificity of hyperreactivity effects by including hyporeactivity as a correlated predictor. RESULTS Hyperreactivity at 14 months was positively associated with fearfulness at 24 months, and hyperreactivity at 24 months was positively associated with SCI and RRB at 36 months. When hyporeactivity was included in the model, paths between hyperreactivity and fearfulness remained, but paths between hyperreactivity and autistic traits became nonsignificant. CONCLUSIONS Our findings indicate that alterations in early sensory reactivity may increase the likelihood of showing fearfulness in infancy, and relate to later social interactions and repetitive behaviours, particularly in individuals with a family history of autism or ADHD.
Collapse
Affiliation(s)
- Nisha Narvekar
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Virginia Carter Leno
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Greg Pasco
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | | | - Mark H Johnson
- Centre for Brain and Cognitive Development, Birkbeck, London, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Tony Charman
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Emily J H Jones
- Centre for Brain and Cognitive Development, Birkbeck, London, UK
| |
Collapse
|
8
|
Soumier A, Lio G, Demily C. Current and future applications of light-sheet imaging for identifying molecular and developmental processes in autism spectrum disorders. Mol Psychiatry 2024; 29:2274-2284. [PMID: 38443634 DOI: 10.1038/s41380-024-02487-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/07/2024]
Abstract
Autism spectrum disorder (ASD) is identified by a set of neurodevelopmental divergences that typically affect the social communication domain. ASD is also characterized by heterogeneous cognitive impairments and is associated with cooccurring physical and medical conditions. As behaviors emerge as the brain matures, it is particularly essential to identify any gaps in neurodevelopmental trajectories during early perinatal life. Here, we introduce the potential of light-sheet imaging for studying developmental biology and cross-scale interactions among genetic, cellular, molecular and macroscale levels of circuitry and connectivity. We first report the core principles of light-sheet imaging and the recent progress in studying brain development in preclinical animal models and human organoids. We also present studies using light-sheet imaging to understand the development and function of other organs, such as the skin and gastrointestinal tract. We also provide information on the potential of light-sheet imaging in preclinical drug development. Finally, we speculate on the translational benefits of light-sheet imaging for studying individual brain-body interactions in advancing ASD research and creating personalized interventions.
Collapse
Affiliation(s)
- Amelie Soumier
- Le Vinatier Hospital Center, 95 boulevard Pinel, 69675, Bron cedex, France.
- iMIND, Center of Excellence for Autism, 95 boulevard Pinel, 69675, Bron cedex, France.
- Institute of Cognitive Science Marc Jeannerod, CNRS, UMR 5229, 67 boulevard Pinel, 69675, Bron cedex, France.
- University Claude Bernard Lyon 1, 43 boulevard du 11 Novembre 1918, 69622, Villeurbanne cedex, France.
| | - Guillaume Lio
- Le Vinatier Hospital Center, 95 boulevard Pinel, 69675, Bron cedex, France
- iMIND, Center of Excellence for Autism, 95 boulevard Pinel, 69675, Bron cedex, France
- Institute of Cognitive Science Marc Jeannerod, CNRS, UMR 5229, 67 boulevard Pinel, 69675, Bron cedex, France
| | - Caroline Demily
- Le Vinatier Hospital Center, 95 boulevard Pinel, 69675, Bron cedex, France
- iMIND, Center of Excellence for Autism, 95 boulevard Pinel, 69675, Bron cedex, France
- Institute of Cognitive Science Marc Jeannerod, CNRS, UMR 5229, 67 boulevard Pinel, 69675, Bron cedex, France
- University Claude Bernard Lyon 1, 43 boulevard du 11 Novembre 1918, 69622, Villeurbanne cedex, France
| |
Collapse
|
9
|
Ford A, Walum H, Brice B, Patel H, Kunnikuru S, Jones W, Berman GJ, Shultz S. Caregiver greeting to infants under 6 months already reflects emerging differences in those later diagnosed with autism. Proc Biol Sci 2024; 291:20232494. [PMID: 38872278 DOI: 10.1098/rspb.2023.2494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/15/2024] [Indexed: 06/15/2024] Open
Abstract
As infants develop, caregivers adjust their behaviour to scaffold their infant's emerging skills, such that changes in infants' social abilities are expected to elicit changes in caregiver behaviour. We examined whether changes in the probability of infant-directed caregiving behaviour-specifically, greeting, a ubiquitous signal used by caregivers to initiate reciprocal interactions-differ between infant-caregiver dyads with an infant later diagnosed with autism and dyads with a neurotypically developing infant during infants' first 6 months. Using longitudinal data from 163 dyads, we found that caregivers in autism dyads (n = 40) used greeting less and at later infant ages than caregivers with a neurotypically developing infant (neurotypical dyads, n = 83). Caregivers in dyads with infants at elevated familial genetic likelihood for autism who did not receive an autism diagnosis (EL-non-autism dyads, n = 40) showed no differences in greeting compared with neurotypical dyads. Socioeconomic status partially mediated the difference between autism and neurotypical dyads. These findings show that autism and socioeconomic status were associated with the mutually adapted dynamics of dyadic interaction beginning in the first postnatal weeks. Importantly, differences in caregiver greeting observed in autism dyads are not interpreted as suboptimal behaviour from caregivers but rather indicate how early emerging social differences related to autism, years before overt features are present, may alter social learning opportunities elicited by the infant.
Collapse
Affiliation(s)
- Aiden Ford
- Neuroscience Graduate Program, Emory University , Atlanta, GA, USA
- Marcus Autism Center , Atlanta, GA, USA
| | - Hasse Walum
- Marcus Autism Center , Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine , Atlanta, GA, USA
| | - Beyonce Brice
- Emory College of Arts and Sciences, Emory University , Atlanta, GA, USA
| | - Hely Patel
- Emory College of Arts and Sciences, Emory University , Atlanta, GA, USA
| | - Sanjana Kunnikuru
- Emory College of Arts and Sciences, Emory University , Atlanta, GA, USA
| | - Warren Jones
- Neuroscience Graduate Program, Emory University , Atlanta, GA, USA
- Marcus Autism Center , Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine , Atlanta, GA, USA
| | - Gordon J Berman
- Neuroscience Graduate Program, Emory University , Atlanta, GA, USA
- Department of Biology, Emory University , Atlanta, GA, USA
| | - Sarah Shultz
- Neuroscience Graduate Program, Emory University , Atlanta, GA, USA
- Marcus Autism Center , Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine , Atlanta, GA, USA
| |
Collapse
|
10
|
Varley AN, Browning KN. Gastrointestinal dysfunction in the valproic acid induced model of social deficit in rats. Auton Neurosci 2024; 253:103161. [PMID: 38461695 PMCID: PMC11128350 DOI: 10.1016/j.autneu.2024.103161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024]
Abstract
Autism spectrum disorder (ASD) has increased in incidence over the past several decades, and is associated with a range of co-morbidities including gastrointestinal (GI) dysfunctions including gastroesophageal reflux, abdominal pain, bloating, constipation and/or diarrhea. Several animal models have been used that replicate several aspects of ASD but no single model has been able to replicate the entire disease pathophysiology. In humans, prenatal exposure to valproic acid (VPA) has been identified as a significant risk factor and rodent models have shown that in utero VPA exposure leads to behavioral deficits in offspring. The present study aimed to investigate whether in utero exposure to VPA induces GI dysfunction in rats. Timed pregnant Sprague-Dawley rats were injected with a single dose of VPA at embryonic day 12.5. Both male and female offspring subsequently underwent behavioral studies and assessment of GI function in adulthood. In utero VPA treatment induced social deficits in both male and female offspring, decreasing sociability and social novelty. Histological examination showed that VPA treated offspring had decreased thickness of GI muscle and mucosa, while immunohistochemical studies showed a decrease in myenteric neuron number in the fundus. Functional studies showed that both male and female VPA offspring had a delay in gastric emptying compared to vehicle treated offspring. Results of the current study suggest that the rat VPA model of behavioral deficits may be a convenient model by which both mechanistic and functional insights into GI dysfunction may be studied.
Collapse
Affiliation(s)
- Ashley N Varley
- Department of Comparative Medicine, Penn State College of Medicine, Hershey, PA, United States of America
| | - Kirsteen N Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States of America.
| |
Collapse
|
11
|
Mullin LJ, Rutsohn J, Gross JL, Caravella KE, Grzadzinski RL, Weisenfeld LA, Flake L, Botteron KN, Dager SR, Estes AM, Pandey J, Schultz RT, St John T, Wolff JJ, Shen MD, Piven J, Hazlett HC, Girault JB. Differential cognitive and behavioral development from 6 to 24 months in autism and fragile X syndrome. J Neurodev Disord 2024; 16:12. [PMID: 38509470 PMCID: PMC10953146 DOI: 10.1186/s11689-024-09519-y] [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: 03/15/2023] [Accepted: 02/14/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Specifying early developmental differences among neurodevelopmental disorders with distinct etiologies is critical to improving early identification and tailored intervention during the first years of life. Recent studies have uncovered important differences between infants with fragile X syndrome (FXS) and infants with familial history of autism spectrum disorder who go on to develop autism themselves (FH-ASD), including differences in brain development and behavior. Thus far, there have been no studies longitudinally investigating differential developmental skill profiles in FXS and FH-ASD infants. METHODS The current study contrasted longitudinal trajectories of verbal (expressive and receptive language) and nonverbal (gross and fine motor, visual reception) skills in FXS and FH-ASD infants, compared to FH infants who did not develop ASD (FH-nonASD) and typically developing controls. RESULTS Infants with FXS showed delays on a nonverbal composite compared to FH-ASD (as well as FH-nonASD and control) infants as early as 6 months of age. By 12 months an ordinal pattern of scores was established between groups on all domains tested, such that controls > FH-nonASD > FH-ASD > FXS. This pattern persisted through 24 months. Cognitive level differentially influenced developmental trajectories for FXS and FH-ASD. CONCLUSIONS Our results demonstrate detectable group differences by 6 months between FXS and FH-ASD as well as differential trajectories on each domain throughout infancy. This work further highlights an earlier onset of global cognitive delays in FXS and, conversely, a protracted period of more slowly emerging delays in FH-ASD. Divergent neural and cognitive development in infancy between FXS and FH-ASD contributes to our understanding of important distinctions in the development and behavioral phenotype of these two groups.
Collapse
Affiliation(s)
- Lindsay J Mullin
- Carolina Institute for Developmental Disabilities, the University of North Carolina at Chapel Hill, Chapel Hill, USA.
| | - Joshua Rutsohn
- Department of Biostatistics, the University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Julia L Gross
- Carolina Institute for Developmental Disabilities, the University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Kelly E Caravella
- Carolina Institute for Developmental Disabilities, the University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Rebecca L Grzadzinski
- Carolina Institute for Developmental Disabilities, the University of North Carolina at Chapel Hill, Chapel Hill, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Leigh Anne Weisenfeld
- Carolina Institute for Developmental Disabilities, the University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Lisa Flake
- Department of Psychiatry, Washington University School of Medicine, St. Louis, USA
| | - Kelly N Botteron
- Department of Psychiatry, Washington University School of Medicine, St. Louis, USA
| | - Stephen R Dager
- Department of Radiology, University of Washington, Seattle, USA
- Center On Human Development and Disability, University of Washington, Seattle, USA
| | - Annette M Estes
- Center On Human Development and Disability, University of Washington, Seattle, USA
- Department of Speech and Hearing Sciences, University of Washington, Seattle, USA
| | - Juhi Pandey
- The Children's Hospital of Philadelphia and University of Pennsylvania, Center for Autism Research, Philadelphia, USA
| | - Robert T Schultz
- The Children's Hospital of Philadelphia and University of Pennsylvania, Center for Autism Research, Philadelphia, USA
| | - Tanya St John
- Center On Human Development and Disability, University of Washington, Seattle, USA
- Department of Speech and Hearing Sciences, University of Washington, Seattle, USA
| | - Jason J Wolff
- Department of Educational Psychology, University of Minnesota, Minneapolis, USA
| | - Mark D Shen
- Carolina Institute for Developmental Disabilities, the University of North Carolina at Chapel Hill, Chapel Hill, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities, the University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Heather C Hazlett
- Carolina Institute for Developmental Disabilities, the University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Jessica B Girault
- Carolina Institute for Developmental Disabilities, the University of North Carolina at Chapel Hill, Chapel Hill, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, USA
| |
Collapse
|
12
|
Loffi RG, Cruz TKF, Paiva GM, Souto DO, Barreto SR, Santana PAN, Nascimento AAAC, Costa FRM, Cota EB, Haase VG. Theoretical-Methodological Foundations for the Global Integration Method (Método de Integração Global-MIG) in the Treatment of Autism Spectrum Disorder. CHILDREN (BASEL, SWITZERLAND) 2024; 11:191. [PMID: 38397303 PMCID: PMC10887636 DOI: 10.3390/children11020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Currently, there is no intervention model for autism spectrum disorder (ASD) that addresses all levels and factors of the International Classification of Functioning, Disability and Health (ICF, WHO). The most researched programs focus on naturalistic, developmental and behavioral approaches to socio-communication. Less attention has been paid to motor and environmental reactivity aspects (behavior/interest restriction and sensory reactivity). The evidence rationale for the Global Integration Method (MIG, "Método de Integração Global"), a model addressing sensorimotor reactivity in addition to socio-communication, is presented. MIG is an integrative, interdisciplinary, family-oriented intervention and naturalistic program that addresses all levels and moderating factors of ASD's impact. MIG's theoretical rationale is based on the predictive coding impairment and embodied cognition hypotheses. MIG incorporates both bottom-up (flexible therapeutic suit, social-motor synchronization) and top-down (schematic social information processing, narratives, imagery) strategies to promote the building and use of accurate, flexible and context-sensitive internal predictive models. MIG is based on the premises that predictive coding improves both socio-communication and environmental reactivity, and that the postural stabilization provided by the flexible therapeutic suit frees information processing resources for socio-cognitive learning. MIG builds on interdisciplinary, professionally and parentally mediated work based on behavioral principles of intensive training in a situated environment.
Collapse
Affiliation(s)
- Renato Guimarães Loffi
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
| | - Thalita Karla Flores Cruz
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Giulia Moreira Paiva
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Deisiane Oliveira Souto
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
| | - Simone Rosa Barreto
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
- Programa de Pós-Graduação em Ciências Fonoaudiológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Patrícia Aparecida Neves Santana
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Amanda Aparecida Alves Cunha Nascimento
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Fabiana Rachel Martins Costa
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
- Programa de Pós-Graduação em Psicologia: Cognição e Comportamento, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Elisa Braz Cota
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
| | - Vitor Geraldi Haase
- Instituto de Neurodesenvolvimento, Cognição e Educação Inclusiva (INCEI), TREINITEC Ltda., Rua Carmélia Loffi 17, Justinópolis, Ribeirão das Neves 33900-730, MG, Brazil; (R.G.L.); (G.M.P.); (D.O.S.); (S.R.B.); (P.A.N.S.); (A.A.A.C.N.); (F.R.M.C.); (E.B.C.); (V.G.H.)
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
- Programa de Pós-Graduação em Psicologia: Cognição e Comportamento, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| |
Collapse
|
13
|
Modafferi S, Lupo G, Tomasello M, Rampulla F, Ontario M, Scuto M, Salinaro AT, Arcidiacono A, Anfuso CD, Legmouz M, Azzaoui FZ, Palmeri A, Spano' S, Biamonte F, Cammilleri G, Fritsch T, Sidenkova A, Calabrese E, Wenzel U, Calabrese V. Antioxidants, Hormetic Nutrition, and Autism. Curr Neuropharmacol 2024; 22:1156-1168. [PMID: 37592816 PMCID: PMC10964097 DOI: 10.2174/1570159x21666230817085811] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 08/19/2023] Open
Abstract
Autism spectrum disorder (ASD) includes a heterogeneous group of complex neurodevelopmental disorders characterized by atypical behaviors with two core pathological manifestations: deficits in social interaction/communication and repetitive behaviors, which are associated with disturbed redox homeostasis. Modulation of cellular resilience mechanisms induced by low levels of stressors represents a novel approach for the development of therapeutic strategies, and in this context, neuroprotective effects of a wide range of polyphenol compounds have been demonstrated in several in vitro and in vivo studies and thoroughly reviewed. Mushrooms have been used in traditional medicine for many years and have been associated with a long list of therapeutic properties, including antitumor, immunomodulatory, antioxidant, antiviral, antibacterial, and hepatoprotective effects. Our recent studies have strikingly indicated the presence of polyphenols in nutritional mushrooms and demonstrated their protective effects in different models of neurodegenerative disorders in humans and rats. Although their therapeutic effects are exerted through multiple mechanisms, increasing attention is focusing on their capacity to induce endogenous defense systems by modulating cellular signaling processes such as nuclear factor erythroid 2 related factor 2 (Nrf2) and nuclear factor-kappa B (NF-κB) pathways. Here we discuss the protective role of hormesis and its modulation by hormetic nutrients in ASD.
Collapse
Affiliation(s)
- Sergio Modafferi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Gabriella Lupo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Mario Tomasello
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Francesco Rampulla
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Marialaura Ontario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Maria Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Antonio Arcidiacono
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Carmelina Daniela Anfuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Maria Legmouz
- Department of Biologie, Laboratory of Biologie and Health, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| | - Fatima-Zahra Azzaoui
- Department of Biologie, Laboratory of Biologie and Health, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| | - Agostino Palmeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Sestina Spano'
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Francesca Biamonte
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| | - Gaetano Cammilleri
- Food Department, Istituto Zooprofilattico Sperimentale della Sicilia, via Gino Marinuzzi, 3 90129, Palermo, Italy
| | | | - Alena Sidenkova
- Department of Psychiatry, Ural State Medical University, Ekaterinburg, Russia
| | - Edward Calabrese
- Department of Environmental Health Sciences; Morrill I, N344, University of Massachusetts, Amherst, MA, 01003, USA
| | - Uwe Wenzel
- Institut für Ernährungswissenschaft, Justus Liebig Universitat Giessen, Germany
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 950125, Italy
| |
Collapse
|
14
|
Anshu K, Nair AK, Srinath S, Laxmi TR. Altered Developmental Trajectory in Male and Female Rats in a Prenatal Valproic Acid Exposure Model of Autism Spectrum Disorder. J Autism Dev Disord 2023; 53:4390-4411. [PMID: 35976506 DOI: 10.1007/s10803-022-05684-y] [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] [Accepted: 07/13/2022] [Indexed: 10/15/2022]
Abstract
Early motor and sensory developmental delays precede Autism Spectrum Disorder (ASD) diagnosis and may serve as early indicators of ASD. The literature on sensorimotor development in animal models is sparse, male centered, and has mixed findings. We characterized early development in a prenatal valproic acid (VPA) model of ASD and found sex-specific developmental delays in VPA rats. We created a developmental composite score combining 15 test readouts, yielding a reliable gestalt measure spanning physical, sensory, and motor development, that effectively discriminated between VPA and control groups. Considering the heterogeneity in ASD phenotype, the developmental composite offers a robust metric that can enable comparison across different animal models of ASD and can serve as an outcome measure for early intervention studies.
Collapse
Affiliation(s)
- Kumari Anshu
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Main Road, Bengaluru, Karnataka, 560029, India
- Waisman Center, University of Wisconsin-Madison, Madison, 53705, WI, USA
| | - Ajay Kumar Nair
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Main Road, Bengaluru, Karnataka, 560029, India
- Center for Healthy Minds, University of Wisconsin-Madison, Madison, 53703, WI, USA
| | - Shoba Srinath
- Department of Child and Adolescent Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Main Road, Bengaluru, Karnataka, 560029, India
| | - T Rao Laxmi
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Main Road, Bengaluru, Karnataka, 560029, India.
| |
Collapse
|
15
|
Statsenko Y, Habuza T, Smetanina D, Simiyu GL, Meribout S, King FC, Gelovani JG, Das KM, Gorkom KNV, Zaręba K, Almansoori TM, Szólics M, Ismail F, Ljubisavljevic M. Unraveling Lifelong Brain Morphometric Dynamics: A Protocol for Systematic Review and Meta-Analysis in Healthy Neurodevelopment and Ageing. Biomedicines 2023; 11:1999. [PMID: 37509638 PMCID: PMC10377186 DOI: 10.3390/biomedicines11071999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
A high incidence and prevalence of neurodegenerative diseases and neurodevelopmental disorders justify the necessity of well-defined criteria for diagnosing these pathologies from brain imaging findings. No easy-to-apply quantitative markers of abnormal brain development and ageing are available. We aim to find the characteristic features of non-pathological development and degeneration in distinct brain structures and to work out a precise descriptive model of brain morphometry in age groups. We will use four biomedical databases to acquire original peer-reviewed publications on brain structural changes occurring throughout the human life-span. Selected publications will be uploaded to Covidence systematic review software for automatic deduplication and blinded screening. Afterwards, we will manually review the titles, abstracts, and full texts to identify the papers matching eligibility criteria. The relevant data will be extracted to a 'Summary of findings' table. This will allow us to calculate the annual rate of change in the volume or thickness of brain structures and to model the lifelong dynamics in the morphometry data. Finally, we will adjust the loss of weight/thickness in specific brain areas to the total intracranial volume. The systematic review will synthesise knowledge on structural brain change across the life-span.
Collapse
Affiliation(s)
- Yauhen Statsenko
- Radiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Medical Imaging Platform, ASPIRE Precision Medicine Research Institute Abu Dhabi, Al Ain P.O. Box 15551, United Arab Emirates
- Big Data Analytics Center, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Tetiana Habuza
- Big Data Analytics Center, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Department of Computer Science and Software Engineering, College of Information Technology, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Darya Smetanina
- Radiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Gillian Lylian Simiyu
- Radiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Sarah Meribout
- Radiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Medical Imaging Platform, ASPIRE Precision Medicine Research Institute Abu Dhabi, Al Ain P.O. Box 15551, United Arab Emirates
- Internal Medicine Department, Maimonides Medical Center, New York, NY 11219, USA
| | - Fransina Christina King
- Physiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Neuroscience Platform, ASPIRE Precision Medicine Research Institute Abu Dhabi, Al Ain P.O. Box 15551, United Arab Emirates
| | - Juri G Gelovani
- Radiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Biomedical Engineering Department, College of Engineering, Wayne State University, Detroit, MI 48202, USA
- Siriraj Hospital, Mahidol University, Nakhon Pathom 73170, Thailand
- Provost Office, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Karuna M Das
- Radiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Klaus N-V Gorkom
- Radiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Kornelia Zaręba
- Obstetrics & Gynecology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Taleb M Almansoori
- Radiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Miklós Szólics
- Neurology Division, Medicine Department, Tawam Hospital, Al Ain, P.O. Box 15258, United Arab Emirates
- Internal Medicine Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Fatima Ismail
- Pediatric Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Milos Ljubisavljevic
- Physiology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Neuroscience Platform, ASPIRE Precision Medicine Research Institute Abu Dhabi, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
16
|
Leming MJ, Bron EE, Bruffaerts R, Ou Y, Iglesias JE, Gollub RL, Im H. Challenges of implementing computer-aided diagnostic models for neuroimages in a clinical setting. NPJ Digit Med 2023; 6:129. [PMID: 37443276 DOI: 10.1038/s41746-023-00868-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Advances in artificial intelligence have cultivated a strong interest in developing and validating the clinical utilities of computer-aided diagnostic models. Machine learning for diagnostic neuroimaging has often been applied to detect psychological and neurological disorders, typically on small-scale datasets or data collected in a research setting. With the collection and collation of an ever-growing number of public datasets that researchers can freely access, much work has been done in adapting machine learning models to classify these neuroimages by diseases such as Alzheimer's, ADHD, autism, bipolar disorder, and so on. These studies often come with the promise of being implemented clinically, but despite intense interest in this topic in the laboratory, limited progress has been made in clinical implementation. In this review, we analyze challenges specific to the clinical implementation of diagnostic AI models for neuroimaging data, looking at the differences between laboratory and clinical settings, the inherent limitations of diagnostic AI, and the different incentives and skill sets between research institutions, technology companies, and hospitals. These complexities need to be recognized in the translation of diagnostic AI for neuroimaging from the laboratory to the clinic.
Collapse
Affiliation(s)
- Matthew J Leming
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| | - Esther E Bron
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Rose Bruffaerts
- Computational Neurology, Experimental Neurobiology Unit (ENU), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Yangming Ou
- Boston Children's Hospital, 300 Longwood Ave, Boston, MA, USA
| | - Juan Eugenio Iglesias
- Center for Medical Image Computing, University College London, London, UK
- Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Randy L Gollub
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hyungsoon Im
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
17
|
Li JH, Zhao JZ, Hua L, Hu XL, Tang LN, Yang T, He TY, Hu C, Yu XQ, Xiong TT, Wu DD, Hao Y. Efficacy of Children Neuropsychological and Behavioral Scale in Screening for Autism Spectrum Disorders through a Combination of Developmental Surveillance. Curr Med Sci 2023:10.1007/s11596-023-2698-5. [PMID: 37115393 DOI: 10.1007/s11596-023-2698-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/22/2022] [Indexed: 04/29/2023]
Abstract
OBJECTIVE This study aimed to explore the clinical value of Children Neuropsychological and Behavioral Scale-Revision 2016 (CNBS-R2016) for Autism Spectrum Disorder (ASD) screening in the presence of developmental surveillance. METHODS All participants were evaluated by the CNBS-R2016 and Gesell Developmental Schedules (GDS). Spearman's correlation coefficients and Kappa values were obtained. Taking GDS as a reference assessment, the performance of the CNBS-R2016 for detecting the developmental delays of children with ASD was analyzed with receiver operating characteristic (ROC) curves. The efficacy of the CNBS-R2016 to screen for ASD was explored by comparing Communication Warning Behavior with Autism Diagnostic Observation Schedule, Second Edition (ADOS-2). RESULTS In total, 150 children aged 12-42 months with ASD were enrolled. The developmental quotients of the CNBS-R2016 were correlated with those of the GDS (r=0.62-0.94). The CNBS-R2016 and GDS had good diagnostic agreement for developmental delays (Kappa=0.73-0.89), except for Fine Motor. There was a significant difference between the proportions of Fine Motor, delays detected by the CNBS-R2016 and GDS (86.0% vs. 77.3%). With GDS as a standard, the areas under the ROC curves of the CNBS-R2016 were above 0.95 for all the domains except Fine Motor, which was 0.70. In addition, the positive rate of ASD was 100.0% and 93.5% when the cut-off points of 7 and 12 in the Communication Warning Behavior subscale were used, respectively. CONCLUSION The CNBS-R2016 performed well in developmental assessment and screening for children with ASD, especially by Communication Warning Behaviors subscale. Therefore, the CNBS-R2016 is worthy of clinical application in children with ASD in China.
Collapse
Affiliation(s)
- Jin-Hui Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jin-Zhu Zhao
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Hua
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Lin Hu
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Na Tang
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ting Yang
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Hospital of Chongqing Medical University, Chongqing, 404100, China
| | - Tian-Yi He
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chen Hu
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Qun Yu
- Future Health Biotechnologies, Chengdu, 61000, China
| | - Ting-Ting Xiong
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan-Dan Wu
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Hao
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
18
|
Kumar M, Hiremath C, Khokhar SK, Bansal E, Sagar KJV, Padmanabha H, Girimaji AS, Narayan S, Kishore MT, Yamini BK, Jac Fredo AR, Saini J, Bharath RD. Altered cerebellar lobular volumes correlate with clinical deficits in siblings and children with ASD: evidence from toddlers. J Transl Med 2023; 21:246. [PMID: 37029372 PMCID: PMC10080978 DOI: 10.1186/s12967-023-04090-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/26/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterized by impaired social and communication skills, narrow interests, and repetitive behavior. It is known that the cerebellum plays a vital role in controlling movement and gait posture. However, recently, researchers have reported that the cerebellum may also be responsible for other functions, such as social cognition, reward, anxiety, language, and executive functions. METHODS In this study, we ascertained volumetric differences from cerebellar lobular analysis from children with ASD, ASD siblings, and typically developing healthy controls. In this cross-sectional study, a total of 30 children were recruited, including children with ASD (N = 15; mean age = 27.67 ± 5.1 months), ASD siblings (N = 6; mean age = 17.5 ± 3.79 months), and typically developing children (N = 9; mean age = 17.67 ± 3.21 months). All the MRI data was acquired under natural sleep without using any sedative medication. We performed a correlation analysis with volumetric data and developmental and behavioral measures obtained from these children. Two-way ANOVA and Pearson correlation was performed for statistical data analysis. RESULTS We observed intriguing findings from this study, including significantly increased gray matter lobular volumes in multiple cerebellar regions including; vermis, left and right lobule I-V, right CrusII, and right VIIb and VIIIb, respectively, in children with ASD, compared to typically developing healthy controls and ASD siblings. Multiple cerebellar lobular volumes were also significantly correlated with social quotient, cognition, language, and motor scores with children with ASD, ASD siblings, and healthy controls, respectively. CONCLUSIONS This research finding helps us understand the neurobiology of ASD and ASD-siblings, and critically advances current knowledge about the cerebellar role in ASD. However, results need to be replicated for a larger cohort from longitudinal research study in future.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029, India.
| | - Chandrakanta Hiremath
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029, India
| | - Sunil Kumar Khokhar
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029, India
| | - Eshita Bansal
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029, India
| | - Kommu John Vijay Sagar
- Department of Child and Adolescent Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, India
| | - Hansashree Padmanabha
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, India
| | - Akhila S Girimaji
- Department of Speech Pathology and Audiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, India
| | - Shweta Narayan
- Department of Clinical Psychology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, India
| | - M Thomas Kishore
- Department of Clinical Psychology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, India
| | - B K Yamini
- Department of Speech Pathology and Audiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, India
| | - A R Jac Fredo
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Jitender Saini
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029, India
| | - Rose Dawn Bharath
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029, India
| |
Collapse
|
19
|
Shahrbabaki SSV, Moslemizadeh A, Amiresmaili S, Tezerji SS, Juybari KB, Sepehri G, Meymandi MS, Bashiri H. Ameliorating age-dependent effects of resveratrol on VPA-induced social impairments and anxiety-like behaviors in a rat model of Neurodevelopmental Disorder. Neurotoxicology 2023; 96:154-165. [PMID: 36933665 DOI: 10.1016/j.neuro.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 01/12/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023]
Abstract
Although anxiety disorders, as well as difficulties in social interaction, are documented in children with autism spectrum disorder (ASD) as a neurodevelopmental disorder, the effectiveness of potential therapeutic procedures considering age and sex differences is under serious discussion. The present study aimed to investigate the effect of resveratrol (RSV) on anxiety-like behaviors and social interaction in juvenile and adult rats of both sex in a valproic acid (VPA)-induced autistic-like model. Prenatal exposure to VPA was associated with increased anxiety, also causing a significant reduction in social interaction in juvenile male subjects. Further administration of RSV attenuated VPA-induced anxiety symptoms in both sexes of adult animals and significantly increased the sociability index in male and female juvenile rats. Taken together, it can be concluded that treatment with RSV can attenuate some of the harsh effects of VPA. This treatment was especially effective on anxiety-like traits in adult subjects of both sexes regarding their performance in open field and EPM. We encourage future research to consider the sex and age-specific mechanisms behind the RSV treatment in the prenatal VPA model of autism.
Collapse
Affiliation(s)
| | | | | | | | - Kobra Bahrampour Juybari
- Department of Pharmacology, Shcool of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Gholamreza Sepehri
- neuroscience Research Center, Institute of Neuropharmacology, Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Manzume Shamsi Meymandi
- neuroscience Research Center, Institute of Neuropharmacology, Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- neuroscience Research Center, Institute of Neuropharmacology, Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
20
|
Risks of attention-deficit/hyperactivity disorder, autism spectrum disorder, and intellectual disability in children delivered by caesarean section: A population-based cohort study. Asian J Psychiatr 2023; 80:103334. [PMID: 36436450 DOI: 10.1016/j.ajp.2022.103334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/05/2022] [Accepted: 11/13/2022] [Indexed: 11/17/2022]
Abstract
This population-based study investigated the risks of attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder (ASD), and intellectual disabilities among children delivered by Cesarean section (CS) in comparison with those who were delivered by vaginal delivery (VD). The Taiwan Maternal and Child Health Database from 2004 to 2016 registered 675,718 and 1,208,983 children delivered by CS and by VD, respectively. The results of Cox proportional hazards regression model demonstrated that children delivered by CS had significantly higher risks of ADHD, ASD, and intellectual disability than those delivered by VD after the confounding effects of maternal and child factors were controlled for.
Collapse
|
21
|
Advancing research on early autism through an integrated risk and resilience perspective. Dev Psychopathol 2023; 35:44-61. [PMID: 35379370 DOI: 10.1017/s0954579421001437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
To date, a deficit-oriented approach dominates autism spectrum disorder (ASD) research, including studies of infant siblings of children with ASD at high risk (HR) for the disabilities associated with this disorder. Despite scientific advances regarding early ASD-related risk, there remains little systematic investigation of positive development, limiting the scope of research and quite possibly a deeper understanding of pathways toward and away from ASD-related impairments. In this paper, we argue that integrating a resilience framework into early ASD research has the potential to enhance knowledge on prodromal course, phenotypic heterogeneity, and developmental processes of risk and adaptation. We delineate a developmental systems resilience framework with particular reference to HR infants. To illustrate the utility of a resilience perspective, we consider the "female protective effect" and other evidence of adaptation in the face of ASD-related risk. We suggest that a resilience framework invites focal questions about the nature, timing, levels, interactions, and mechanisms by which positive adaptation occurs in relation to risk and developmental pathways toward and away from ASD-related difficulties. We conclude with recommendations for future research, including more focus on adaptive development and multisystem processes, pathways away from disorder, and reconsideration of extant evidence within an integrated risk-and-resilience framework.
Collapse
|
22
|
McEwan F, Glazier JD, Hager R. The impact of maternal immune activation on embryonic brain development. Front Neurosci 2023; 17:1146710. [PMID: 36950133 PMCID: PMC10025352 DOI: 10.3389/fnins.2023.1146710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
The adult brain is a complex structure with distinct functional sub-regions, which are generated from an initial pool of neural epithelial cells within the embryo. This transition requires a number of highly coordinated processes, including neurogenesis, i.e., the generation of neurons, and neuronal migration. These take place during a critical period of development, during which the brain is particularly susceptible to environmental insults. Neurogenesis defects have been associated with the pathogenesis of neurodevelopmental disorders (NDDs), such as autism spectrum disorder and schizophrenia. However, these disorders have highly complex multifactorial etiologies, and hence the underlying mechanisms leading to aberrant neurogenesis continue to be the focus of a significant research effort and have yet to be established. Evidence from epidemiological studies suggests that exposure to maternal infection in utero is a critical risk factor for NDDs. To establish the biological mechanisms linking maternal immune activation (MIA) and altered neurodevelopment, animal models have been developed that allow experimental manipulation and investigation of different developmental stages of brain development following exposure to MIA. Here, we review the changes to embryonic brain development focusing on neurogenesis, neuronal migration and cortical lamination, following MIA. Across published studies, we found evidence for an acute proliferation defect in the embryonic MIA brain, which, in most cases, is linked to an acceleration in neurogenesis, demonstrated by an increased proportion of neurogenic to proliferative divisions. This is accompanied by disrupted cortical lamination, particularly in the density of deep layer neurons, which may be a consequence of the premature neurogenic shift. Although many aspects of the underlying pathways remain unclear, an altered epigenome and mitochondrial dysfunction are likely mechanisms underpinning disrupted neurogenesis in the MIA model. Further research is necessary to delineate the causative pathways responsible for the variation in neurogenesis phenotype following MIA, which are likely due to differences in timing of MIA induction as well as sex-dependent variation. This will help to better understand the underlying pathogenesis of NDDs, and establish therapeutic targets.
Collapse
|
23
|
Girault JB, Donovan K, Hawks Z, Talovic M, Forsen E, Elison JT, Shen MD, Swanson MR, Wolff JJ, Kim SH, Nishino T, Davis S, Snyder AZ, Botteron KN, Estes AM, Dager SR, Hazlett HC, Gerig G, McKinstry R, Pandey J, Schultz RT, St John T, Zwaigenbaum L, Todorov A, Truong Y, Styner M, Pruett JR, Constantino JN, Piven J. Infant Visual Brain Development and Inherited Genetic Liability in Autism. Am J Psychiatry 2022; 179:573-585. [PMID: 35615814 PMCID: PMC9356977 DOI: 10.1176/appi.ajp.21101002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Autism spectrum disorder (ASD) is heritable, and younger siblings of ASD probands are at higher likelihood of developing ASD themselves. Prospective MRI studies of siblings report that atypical brain development precedes ASD diagnosis, although the link between brain maturation and genetic factors is unclear. Given that familial recurrence of ASD is predicted by higher levels of ASD traits in the proband, the authors investigated associations between proband ASD traits and brain development among younger siblings. METHODS In a sample of 384 proband-sibling pairs (89 pairs concordant for ASD), the authors examined associations between proband ASD traits and sibling brain development at 6, 12, and 24 months in key MRI phenotypes: total cerebral volume, cortical surface area, extra-axial cerebrospinal fluid, occipital cortical surface area, and splenium white matter microstructure. Results from primary analyses led the authors to implement a data-driven approach using functional connectivity MRI at 6 months. RESULTS Greater levels of proband ASD traits were associated with larger total cerebral volume and surface area and larger surface area and reduced white matter integrity in components of the visual system in siblings who developed ASD. This aligned with weaker functional connectivity between several networks and the visual system among all siblings during infancy. CONCLUSIONS The findings provide evidence that specific early brain MRI phenotypes of ASD reflect quantitative variation in familial ASD traits. Multimodal anatomical and functional convergence on cortical regions, fiber pathways, and functional networks involved in visual processing suggest that inherited liability has a role in shaping the prodromal development of visual circuitry in ASD.
Collapse
Affiliation(s)
- Jessica B Girault
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Educational Psychology (Wolff), University of Minnesota, Minneapolis;Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Kevin Donovan
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Zoë Hawks
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Muhamed Talovic
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Elizabeth Forsen
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Jed T Elison
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Mark D Shen
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Meghan R Swanson
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Jason J Wolff
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Sun Hyung Kim
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Tomoyuki Nishino
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Savannah Davis
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Abraham Z Snyder
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Kelly N Botteron
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Annette M Estes
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Stephen R Dager
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Heather C Hazlett
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Guido Gerig
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Robert McKinstry
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Juhi Pandey
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Robert T Schultz
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Tanya St John
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Lonnie Zwaigenbaum
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Alexandre Todorov
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Young Truong
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Martin Styner
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - John R Pruett
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - John N Constantino
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities (Girault, Forsen, Shen, Hazlett, Piven), Department of Psychiatry (Girault, Shen, Kim, Hazlett, Styner, Piven), Department of Biostatistics (Donovan, Truong), and ; Department of Psychological and Brain Sciences (Hawks) and Department of Psychiatry (Talovic, Nishino, Davis, Botteron, Todorov, Pruett, Constantino), Washington University School of Medicine in St. Louis; Institute of Child Development (Elison) and Department of Psychology, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Tex. (Swanson); Department of Radiology, Washington University in St. Louis (Snyder, McKinstry); Department of Speech and Hearing Science, University of Washington, Seattle (Estes, St. John); Department of Radiology, University of Washington Medical Center, Seattle (Dager); Tandon School of Engineering, New York University, New York (Gerig); Center for Autism Research, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Pandey, Schultz); Department of Pediatrics, University of Alberta, Edmonton, Canada (Zwaigenbaum)
| |
Collapse
|
24
|
Ayoub MJ, Keegan L, Tager-Flusberg H, Gill SV. Neuroimaging Techniques as Descriptive and Diagnostic Tools for Infants at Risk for Autism Spectrum Disorder: A Systematic Review. Brain Sci 2022; 12:602. [PMID: 35624989 PMCID: PMC9139416 DOI: 10.3390/brainsci12050602] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
Autism Spectrum Disorder (ASD) has traditionally been evaluated and diagnosed via behavioral assessments. However, increasing research suggests that neuroimaging as early as infancy can reliably identify structural and functional differences between autistic and non-autistic brains. The current review provides a systematic overview of imaging approaches used to identify differences between infants at familial risk and without risk and predictive biomarkers. Two primary themes emerged after reviewing the literature: (1) neuroimaging methods can be used to describe structural and functional differences between infants at risk and infants not at risk for ASD (descriptive), and (2) neuroimaging approaches can be used to predict ASD diagnosis among high-risk infants and developmental outcomes beyond infancy (predicting later diagnosis). Combined, the articles highlighted that several neuroimaging studies have identified a variety of neuroanatomical and neurological differences between infants at high and low risk for ASD, and among those who later receive an ASD diagnosis. Incorporating neuroimaging into ASD evaluations alongside traditional behavioral assessments can provide individuals with earlier diagnosis and earlier access to supportive resources.
Collapse
Affiliation(s)
- Maria J. Ayoub
- College of Health and Rehabilitation Sciences: Sargent College, Boston University, 635 Commonwealth Avenue, Boston, MA 02215, USA; (M.J.A.); (L.K.)
| | - Laura Keegan
- College of Health and Rehabilitation Sciences: Sargent College, Boston University, 635 Commonwealth Avenue, Boston, MA 02215, USA; (M.J.A.); (L.K.)
| | - Helen Tager-Flusberg
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA;
| | - Simone V. Gill
- College of Health and Rehabilitation Sciences: Sargent College, Boston University, 635 Commonwealth Avenue, Boston, MA 02215, USA; (M.J.A.); (L.K.)
| |
Collapse
|
25
|
McDonald NM. Studying the early emergence of autism: what is the goal of baby siblings research? Dev Med Child Neurol 2022; 64:534. [PMID: 34807457 DOI: 10.1111/dmcn.15114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Nicole M McDonald
- UCLA Semel Institute for Neuroscience & Human Behavior, Los Angeles, CA, USA
| |
Collapse
|
26
|
Silverman JL, Thurm A, Ethridge SB, Soller MM, Petkova SP, Abel T, Bauman MD, Brodkin ES, Harony‐Nicolas H, Wöhr M, Halladay A. Reconsidering animal models used to study autism spectrum disorder: Current state and optimizing future. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12803. [PMID: 35285132 PMCID: PMC9189007 DOI: 10.1111/gbb.12803] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 12/15/2022]
Abstract
Neurodevelopmental disorders (NDDs), including autism spectrum disorder (ASD) and intellectual disability (ID), are pervasive, often lifelong disorders, lacking evidence-based interventions for core symptoms. With no established biological markers, diagnoses are defined by behavioral criteria. Thus, preclinical in vivo animal models of NDDs must be optimally utilized. For this reason, experts in the field of behavioral neuroscience convened a workshop with the goals of reviewing current behavioral studies, reports, and assessments in rodent models. Goals included: (a) identifying the maximal utility and limitations of behavior in animal models with construct validity; (b) providing recommendations for phenotyping animal models; and (c) guidelines on how in vivo models should be used and reported reliably and rigorously while acknowledging their limitations. We concluded by recommending minimal criteria for reporting in manuscripts going forward. The workshop elucidated a consensus of potential solutions to several problems, including revisiting claims made about animal model links to ASD (and related conditions). Specific conclusions included: mice (or other rodent or preclinical models) are models of the neurodevelopmental insult, not specifically any disorder (e.g., ASD); a model that perfectly recapitulates a disorder such as ASD is untenable; and greater attention needs be given to validation of behavioral testing methods, data analysis, and critical interpretation.
Collapse
Affiliation(s)
- Jill L. Silverman
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping ServiceNational Institute of Mental HealthBethesdaMarylandUSA
| | - Sarah B. Ethridge
- Neurodevelopmental and Behavioral Phenotyping ServiceNational Institute of Mental HealthBethesdaMarylandUSA
| | - Makayla M. Soller
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Stela P. Petkova
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Ted Abel
- Department of Neuroscience and PharmacologyIowa Neuroscience Institute, University of IowaIowa CityIowaUSA
| | - Melissa D. Bauman
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Edward S. Brodkin
- Department of PsychiatryPerelman School of Medicine at the University of Pennsylvania, Translational Research LaboratoryPhiladelphiaPennsylvaniaUSA
| | - Hala Harony‐Nicolas
- Seaver Autism Center for Research and TreatmentIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Markus Wöhr
- Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological PsychologySocial and Affective Neuroscience Research Group, KU LeuvenLeuvenBelgium,Leuven Brain InstituteKU LeuvenLeuvenBelgium,Faculty of Psychology, Experimental and Biological Psychology, Behavioral NeurosciencePhilipps‐University of MarburgMarburgGermany,Center for Mind, Brain, and BehaviorPhilipps‐University of MarburgMarburgGermany
| | - Alycia Halladay
- Autism Science FoundationUSA,Department of Pharmacology and ToxicologyRutgers UniversityPiscatawayNew JerseyUSA
| |
Collapse
|
27
|
Al Dera H. Cellular and molecular mechanisms underlying autism spectrum disorders and associated comorbidities: A pathophysiological review. Biomed Pharmacother 2022; 148:112688. [PMID: 35149383 DOI: 10.1016/j.biopha.2022.112688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 12/31/2022] Open
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders that develop in early life due to interaction between several genetic and environmental factors and lead to alterations in brain function and structure. During the last decades, several mechanisms have been placed to explain the pathogenesis of autism. Unfortunately, these are reported in several studies and reviews which make it difficult to follow by the reader. In addition, some recent molecular mechanisms related to ASD have been unrevealed. This paper revises and highlights the major common molecular mechanisms responsible for the clinical symptoms seen in people with ASD, including the roles of common genetic factors and disorders, neuroinflammation, GABAergic signaling, and alterations in Ca+2 signaling. Besides, it covers the major molecular mechanisms and signaling pathways involved in initiating the epileptic seizure, including the alterations in the GABAergic and glutamate signaling, vitamin and mineral deficiency, disorders of metabolism, and autoimmunity. Finally, this review also discusses sleep disorder patterns and the molecular mechanisms underlying them.
Collapse
Affiliation(s)
- Hussain Al Dera
- Department of Basic Medical Sciences, College of Medicine at King Saud, Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia.
| |
Collapse
|
28
|
Wilder L, Semendeferi K. Infant Brain Development and Plasticity from an Evolutionary Perspective. EVOLUTIONARY PSYCHOLOGY 2022. [DOI: 10.1007/978-3-030-76000-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
29
|
Guma E, Snook E, Spring S, Lerch JP, Nieman BJ, Devenyi GA, Chakravarty MM. Subtle alterations in neonatal neurodevelopment following early or late exposure to prenatal maternal immune activation in mice. Neuroimage Clin 2021; 32:102868. [PMID: 34749289 PMCID: PMC8573196 DOI: 10.1016/j.nicl.2021.102868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022]
Abstract
Prenatal exposure to maternal immune activation (MIA) is a risk factor for a variety of neurodevelopmental and psychiatric disorders. The timing of MIA-exposure has been shown to affect adolescent and adult offspring neurodevelopment, however, less is known about these effects in the neonatal period. To better understand the impact of MIA-exposure on neonatal brain development in a mouse model, we assess neonate communicative abilities with the ultrasonic vocalization task, followed by high-resolution ex vivo magnetic resonance imaging (MRI) on the neonatal (postnatal day 8) mouse brain. Early exposed offspring displayed decreased communicative ability, while brain anatomy appeared largely unaffected, apart from some subtle alterations. By integrating MRI and behavioural assays to investigate the effects of MIA-exposure on neonatal neurodevelopment we show that offspring neuroanatomy and behaviour are only subtly affected by both early and late exposure. This suggests that the deficits often observed in later stages of life may be dormant, not yet developed in the neonatal period, or not as easily detectable using a cross-sectional approach.
Collapse
Affiliation(s)
- Elisa Guma
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada.
| | - Emily Snook
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shoshana Spring
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Brian J Nieman
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Gabriel A Devenyi
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
30
|
Peterson M, Prigge MBD, Bigler ED, Zielinski B, King JB, Lange N, Alexander A, Lainhart JE, Nielsen JA. Evidence for normal extra-axial cerebrospinal fluid volume in autistic males from middle childhood to adulthood. Neuroimage 2021; 240:118387. [PMID: 34260891 PMCID: PMC8485737 DOI: 10.1016/j.neuroimage.2021.118387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 07/01/2021] [Accepted: 07/10/2021] [Indexed: 12/03/2022] Open
Abstract
Autism spectrum disorder has long been associated with a variety of organizational and developmental abnormalities in the brain. An increase in extra-axial cerebrospinal fluid volume in autistic individuals between the ages of 6 months and 4 years has been reported in recent studies. Increased extra-axial cerebrospinal fluid volume was predictive of the diagnosis and severity of the autistic symptoms in all of them, irrespective of genetic risk for developing the disorder. In the present study, we explored the trajectory of extra-axial cerebrospinal fluid volume from childhood to adulthood in both autism and typical development. We hypothesized that an elevated extra-axial cerebrospinal fluid volume would be found in autism persisting throughout the age range studied. We tested the hypothesis by employing an accelerated, multi-cohort longitudinal data set of 189 individuals (97 autistic, 92 typically developing). Each individual had been scanned between 1 and 5 times, with scanning sessions separated by 2-3 years, for a total of 439 T1-weighted MRI scans. A linear mixed-effects model was used to compare developmental, age-related changes in extra-axial cerebrospinal fluid volume between groups. Inconsistent with our hypothesis, we found no group differences in extra-axial cerebrospinal fluid volume in this cohort of individuals 3 to 42 years of age. Our results suggest that extra-axial cerebrospinal fluid volume in autistic individuals is not increased compared with controls beyond four years of age.
Collapse
Affiliation(s)
- Madeline Peterson
- Department of Psychology, Brigham Young University, Provo, UT, 84602, United States
| | - Molly B D Prigge
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, 84108, United States; Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Erin D Bigler
- Department of Psychology, Brigham Young University, Provo, UT, 84602, United States; Neuroscience Center, Brigham Young University, Provo, UT, 84604, United States; Department of Neurology, University of Utah, Salt Lake City, UT, 84108, United States; Department of Neurology, University of California-Davis, Davis, CA United States
| | - Brandon Zielinski
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, 84108, United States; Department of Neurology, University of Utah, Salt Lake City, UT, 84108, United States; Department of Pediatrics, University of Utah, Salt Lake City, UT, 84108, United States
| | - Jace B King
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, 84108, United States
| | - Nicholas Lange
- Department of Psychiatry, Harvard Medical School, Boston, MA 02115, United States
| | - Andrew Alexander
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, United States; Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, 53719, United States; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Janet E Lainhart
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, United States; Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, 53719, United States
| | - Jared A Nielsen
- Department of Psychology, Brigham Young University, Provo, UT, 84602, United States; Neuroscience Center, Brigham Young University, Provo, UT, 84604, United States.
| |
Collapse
|
31
|
Heimann M, Holmer E. Neonatal Imitation, Intersubjectivity, and Children With Atypical Development: Do Observations on Autism and Down Syndrome Change Our Understanding? Front Psychol 2021; 12:701795. [PMID: 34512459 PMCID: PMC8430258 DOI: 10.3389/fpsyg.2021.701795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/30/2021] [Indexed: 11/26/2022] Open
Abstract
Almost all studies on neonatal imitation to date seem to have focused on typically developing children, and we thus lack information on the early imitative abilities of children who follow atypical developmental trajectories. From both practical and theoretical perspectives, these abilities might be relevant to study in children who develop a neuropsychiatric diagnosis later on or in infants who later show impaired ability to imitate. Theoretical in the sense that it will provide insight into the earliest signs of intersubjectivity—i.e., primary intersubjectivity—and how this knowledge might influence our understanding of children following atypical trajectories of development. Practical in the sense that it might lead to earlier detection of certain disabilities. In the present work, we screen the literature for empirical studies on neonatal imitation in children with an Autism spectrum disorder (ASD) or Down syndrome (DS) as well as present an observation of neonatal imitation in an infant that later was diagnosed with autism and a re-interpretation of previously published data on the phenomenon in a small group of infants with DS. Our findings suggest that the empirical observations to date are too few to draw any definite conclusions but that the existing data suggests that neonatal imitation can be observed both in children with ASD and in children with DS. Thus, neonatal imitation might not represent a useful predictor of a developmental deficit. Based on current theoretical perspectives advocating that neonatal imitation is a marker of primary intersubjectivity, we propose tentatively that an ability to engage in purposeful exchanges with another human being exists in these populations from birth.
Collapse
Affiliation(s)
- Mikael Heimann
- Infant and Child Lab, Department of Behavioural Sciences and Learning, Linköping University, Linköping, Sweden
| | - Emil Holmer
- Department of Behavioural Sciences and Learning, The Swedish Institute for Disability Research, Linköping University, Linköping, Sweden
| |
Collapse
|
32
|
Sacrey LAR, Zwaigenbaum L, Bryson S, Brian J, Smith IM, Roberts W, Szatmari P, Vaillancourt T, Roncadin C, Garon N. Screening for Behavioral Signs of Autism Spectrum Disorder in 9-Month-Old Infant Siblings. J Autism Dev Disord 2021; 51:839-848. [PMID: 31939081 DOI: 10.1007/s10803-020-04371-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite considerable progress in characterizing the early signs of autism spectrum disorder (ASD), more remains to be learned about how symptoms emerge in the first year of life. Parents with a new baby who already had at least one biological child diagnosed with ASD (high-risk) or no family history of ASD (low-risk) completed two measures when their baby was 9 months of age, the Autism Parent Screen for Infants (APSI) questionnaire and the interview-based Parent Concerns Form. Children underwent a blinded independent diagnostic assessment for ASD at age 3 years. Total scores on the APSI and the Parent Concerns Form were both able to independently differentiate high-risk children who were later diagnosed with ASD from other high-risk and low-risk children who were not. Using logistic regression, we found that the total score on the APSI predicted ASD outcomes at age 3 with 70% accuracy, but the Parent Concerns Form did not contribute any unique variance when the APSI was already in the model. The results suggest that the APSI identifies early features predictive of ASD in high-risk infants and can be used to flag them for targeted follow-up and screening.
Collapse
Affiliation(s)
- Lori-Ann R Sacrey
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada. .,Department of Pediatrics, Autism Research Centre - E209, Glenrose Rehabilitation Hospital, 10230-111 Avenue, Edmonton, AB, T5G 0B7, Canada.
| | - Lonnie Zwaigenbaum
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Department of Pediatrics, Autism Research Centre - E209, Glenrose Rehabilitation Hospital, 10230-111 Avenue, Edmonton, AB, T5G 0B7, Canada
| | - Susan Bryson
- Dalhousie University/IWK Health Centre, Halifax, NS, Canada
| | - Jessica Brian
- Bloorview Research Institute, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Isabel M Smith
- Dalhousie University/IWK Health Centre, Halifax, NS, Canada
| | | | - Peter Szatmari
- University of Toronto, Toronto, ON, Canada.,The Hospital for Sick Children, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | | | - Caroline Roncadin
- University of Toronto, Toronto, ON, Canada.,McMaster Children's Hospital, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Nancy Garon
- Mount Allison University, Sackville, NB, Canada
| |
Collapse
|
33
|
Nair A, Jalal R, Liu J, Tsang T, McDonald NM, Jackson L, Ponting C, Jeste SS, Bookheimer SY, Dapretto M. Altered Thalamocortical Connectivity in 6-Week-Old Infants at High Familial Risk for Autism Spectrum Disorder. Cereb Cortex 2021; 31:4191-4205. [PMID: 33866373 DOI: 10.1093/cercor/bhab078] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Converging evidence from neuroimaging studies has revealed altered connectivity in cortical-subcortical networks in youth and adults with autism spectrum disorder (ASD). Comparatively little is known about the development of cortical-subcortical connectivity in infancy, before the emergence of overt ASD symptomatology. Here, we examined early functional and structural connectivity of thalamocortical networks in infants at high familial risk for ASD (HR) and low-risk controls (LR). Resting-state functional connectivity and diffusion tensor imaging data were acquired in 52 6-week-old infants. Functional connectivity was examined between 6 cortical seeds-prefrontal, motor, somatosensory, temporal, parietal, and occipital regions-and bilateral thalamus. We found significant thalamic-prefrontal underconnectivity, as well as thalamic-occipital and thalamic-motor overconnectivity in HR infants, relative to LR infants. Subsequent structural connectivity analyses also revealed atypical white matter integrity in thalamic-occipital tracts in HR infants, compared with LR infants. Notably, aberrant connectivity indices at 6 weeks predicted atypical social development between 9 and 36 months of age, as assessed with eye-tracking and diagnostic measures. These findings indicate that thalamocortical connectivity is disrupted at both the functional and structural level in HR infants as early as 6 weeks of age, providing a possible early marker of risk for ASD.
Collapse
Affiliation(s)
- Aarti Nair
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA 92354, USA
| | - Rhideeta Jalal
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Janelle Liu
- Interdepartmental Neuroscience Program, University of California, Los Angeles, CA 90095, USA
| | - Tawny Tsang
- Department of Psychology, University of California, Los Angeles, CA 90095, USA
| | - Nicole M McDonald
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Lisa Jackson
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Carolyn Ponting
- Department of Psychology, University of California, Los Angeles, CA 90095, USA
| | - Shafali S Jeste
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Susan Y Bookheimer
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Mirella Dapretto
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
34
|
Zhong H, Xiao R, Ruan R, Liu H, Li X, Cai Y, Zhao J, Fan X. Neonatal curcumin treatment restores hippocampal neurogenesis and improves autism-related behaviors in a mouse model of autism. Psychopharmacology (Berl) 2020; 237:3539-3552. [PMID: 32803366 DOI: 10.1007/s00213-020-05634-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
RATIONALE Autism spectrum disorders (ASDs) are highly prevalent neurodevelopmental disorders characterized by deficits in social communication and interaction, repetitive stereotyped behaviors, and cognitive impairments. Curcumin has been indicated to be neuroprotective against neurological and psychological disorders. However, the role of curcumin in autistic phenotypes remains unclear. OBJECTIVES In the current study, we evaluated the effects of neonatal curcumin treatment on behavior and hippocampal neurogenesis in BTBRT+ltpr3tf/J (BTBR) mice, a model of autism. METHODS C57BL/6J (C57) and BTBR mouse pups were treated with 0.1% dimethyl sulfoxide (DMSO) or curcumin (20 mg/kg) from postnatal day 6 (P6) to P8. Neural progenitor cells (NPCs) in the hippocampal dentate gyrus (DG) were evaluated on P8, and neurogenesis was measured on P24 by immunofluorescence. A battery of behavioral tests was carried out when the mice were 8 weeks of age. RESULTS Neonatal curcumin treatment improved autism-related symptoms in BTBR mice, enhancing sociability, reducing repetitive behaviors, and ameliorating cognitive impairments. Furthermore, the suppression of hippocampal neurogenesis in BTBR mice was greatly rescued after neonatal curcumin treatment, leading to an increase in neurogenic processes and an increase in NPC proliferation concomitant with an expansion of the NPC pool on P8, and NPC differentiation towards the neuronal lineage was promoted in the DG of BTBR mice on P24. CONCLUSIONS Our findings suggest that neonatal curcumin treatment elicits a therapeutic response through the restoration of hippocampal neurogenesis in BTBR mice and thus may represent a promising novel pharmacological strategy for ASD treatment.
Collapse
Affiliation(s)
- Hongyu Zhong
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Rui Xiao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Ruotong Ruan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Hui Liu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Xin Li
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Yun Cai
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Jinghui Zhao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China. .,Institute of Brain and Intelligence, Chongqing, 400038, China.
| |
Collapse
|
35
|
Samanta D. An Updated Review of Tuberous Sclerosis Complex-Associated Autism Spectrum Disorder. Pediatr Neurol 2020; 109:4-11. [PMID: 32563542 DOI: 10.1016/j.pediatrneurol.2020.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 01/30/2023]
Abstract
Tuberous sclerosis complex (TSC) is a neurocutaneous disorder caused by mutations of either the TSC1 or TSC2 gene. Various neuropsychiatric features, including autism, are prevalent in TSC. Recently, significant progress has been possible with the prospective calculation of the prevalence of autism in TSC, identification of early clinical and neurophysiological biomarkers to predict autism, and investigation of different therapies to prevent autism in this high-risk population. The author provides a narrative review of recent findings related to biomarkers for diagnosis of autism in TSC, as well as recent studies related to the management of TSC-associated autism. Further sophisticated modeling and analysis are required to understand the role of different models-tuber models, seizures and related neurophysiological factors models, genotype models, and brain connectivity models-to unravel the neurobiological basis of autism in TSC. Early neuropsychologic assessments may be beneficial in this high-risk group. Targeted intervention to improve visual skill, cognition, and fine motor skills with later addition of social skill training can be helpful. Multicenter, prospective studies are ongoing to identify if presymptomatic treatment with vigabatrin in patients with TSC can improve outcomes, including autism. Several studies indicated reasonable safety of everolimus in young children, and its potential application in high-risk infants with TSC, before the closure of the temporal window of permanent changes, maybe undertaken shortly.
Collapse
Affiliation(s)
- Debopam Samanta
- Child Neurology Section, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
36
|
Haartsen R, van der Velde B, Jones EJH, Johnson MH, Kemner C. Using multiple short epochs optimises the stability of infant EEG connectivity parameters. Sci Rep 2020; 10:12703. [PMID: 32728099 PMCID: PMC7391718 DOI: 10.1038/s41598-020-68981-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
Atypicalities in connectivity between brain regions have been implicated in a range of neurocognitive disorders. We require metrics to assess stable individual differences in connectivity in the developing brain, while facing the challenge of limited data quality and quantity. Here, we examine how varying core processing parameters can optimise the test–retest reliability of EEG connectivity measures in infants. EEG was recorded twice with a 1-week interval between sessions in 10-month-olds. EEG alpha connectivity was measured across different epoch lengths and numbers, with the phase lag index (PLI) and debiased weighted PLI (dbWPLI), for both whole-head connectivity and graph theory metrics. We calculated intra-class correlations between sessions for infants with sufficient data for both sessions (N’s = 19–41, depending on the segmentation method). Reliability for the whole brain dbWPLI was higher across many short epochs, whereas reliability for the whole brain PLI was higher across fewer long epochs. However, the PLI is confounded by the number of available segments. Reliability was higher for whole brain connectivity than graph theory metrics. Thus, segmenting available data into a high number of short epochs and calculating the dbWPLI is most appropriate for characterising connectivity in populations with limited availability of EEG data.
Collapse
Affiliation(s)
- Rianne Haartsen
- Department of Psychological Sciences (BMA), Centre for Brain and Cognitive Development, Birkbeck College, University of London, Malet Street, London, WC1E 7HX, UK.
| | - Bauke van der Velde
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Heidelberglaan 1, 3584 CS, Utrecht, The Netherlands.,Department of Developmental Psychology, Utrecht University, Utrecht, The Netherlands.,Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Emily J H Jones
- Department of Psychological Sciences (BMA), Centre for Brain and Cognitive Development, Birkbeck College, University of London, Malet Street, London, WC1E 7HX, UK
| | - Mark H Johnson
- Department of Psychological Sciences (BMA), Centre for Brain and Cognitive Development, Birkbeck College, University of London, Malet Street, London, WC1E 7HX, UK.,Department of Psychology, University of Cambridge, Cambridge, UK
| | - Chantal Kemner
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Heidelberglaan 1, 3584 CS, Utrecht, The Netherlands.,Department of Developmental Psychology, Utrecht University, Utrecht, The Netherlands.,Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| |
Collapse
|
37
|
Sex dependent alterations of resveratrol on social behaviors and nociceptive reactivity in VPA-induced autistic-like model in rats. Neurotoxicol Teratol 2020; 81:106905. [PMID: 32534151 DOI: 10.1016/j.ntt.2020.106905] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 06/01/2020] [Accepted: 06/08/2020] [Indexed: 01/20/2023]
Abstract
INTRODUCTION The present study was designed to clarify the effects of resveratrol (RSV) on social behavioral alterations and nociceptive reactivity in valproic acid (VPA)-induced autistic-like model in female and male rats. METHODS Pregnant Wistar rats were randomly divided in five groups. Animals received saline, DMSO, VPA, RSV and RSV + VPA. VPA was administered (600 mg/kg, i. p.) on embryonic day 12.5 (E12.5) and pretreatment by resveratrol (3.6 mg/kg, s. c.) was applied on E6.5 until E18.5. All offspring were weaned on postnatal day 21 and the experiments were done in male and female rats on day 60. Social interaction, hot plate and tail flick tests were set out to assess social deficits and pain threshold, respectively. Sociability index (SI), Social novelty index (SNI) and latency time were calculated as the standard indices of social behaviors and pain threshold, respectively. RESULTS The results indicated that systemic intraperitoneal administration of VPA (600 mg/kg) significantly decreased SI and SNI in social interaction test (SIT) especially in male rats, indicating the social impairments caused by VPA. RSV (3.6 mg/kg, s. c.) reversed VPA-induced social deficits in male rats, but not in female group. VPA administration resulted in significant increase in latency time in the hot plate and tail flick tests in male rats, whereas it had no such dramatic effect in females. RSV administration in combination with VPA had no significant effect on latency time compared to the valproic acid group in male rats. It is important to note that RSV by itself had no significant effect on SI, SNI and latency time in female and male rats. CONCLUSION It can be concluded that valproic acid produces autistic-like behaviors and increases pain threshold in male rats which may be ameliorated at least in part by resveratrol administration. Further studies are needed to elucidate the molecular mechanisms involved in valproic acid and resveratrol-induced effects.
Collapse
|
38
|
Geng X, Kang X, Wong PCM. Autism spectrum disorder risk prediction: A systematic review of behavioral and neural investigations. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 173:91-137. [PMID: 32711819 DOI: 10.1016/bs.pmbts.2020.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
A reliable diagnosis of autism spectrum disorder (ASD) is difficult to make until after toddlerhood. Detection in an earlier age enables early intervention, which is typically more effective. Recent studies of the development of brain and behavior in infants and toddlers have provided important insights in the diagnosis of autism. This extensive review focuses on published studies of predicting the diagnosis of autism during infancy and toddlerhood younger than 3 years using behavioral and neuroimaging approaches. After screening a total of 782 papers, 17 neuroimaging and 43 behavioral studies were reviewed. The features for prediction consist of behavioral measures using screening tools, observational and experimental methods, brain volumetric measures, and neural functional activation and connectivity patterns. The classification approaches include logistic regression, linear discriminant function, decision trees, support vector machine, and deep learning based methods. Prediction performance has large variance across different studies. For behavioral studies, the sensitivity varies from 20% to 100%, and specificity ranges from 48% to 100%. The accuracy rates range from 61% to 94% in neuroimaging studies. Possible factors contributing to this inconsistency may be partially due to the heterogeneity of ASD, different targeted populations (i.e., high-risk group for ASD and general population), age when the features were collected, and validation procedures. The translation to clinical practice requires extensive further research including external validation with large sample size and optimized feature selection. The use of multi-modal features, e.g., combination of neuroimaging and behavior, is worth further investigation to improve the prediction accuracy.
Collapse
Affiliation(s)
- Xiujuan Geng
- Brain and Mind Institute, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Xin Kang
- Brain and Mind Institute, The Chinese University of Hong Kong, Shatin, Hong Kong; Department of Linguistics and Modern Languages, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Patrick C M Wong
- Brain and Mind Institute, The Chinese University of Hong Kong, Shatin, Hong Kong; Department of Linguistics and Modern Languages, The Chinese University of Hong Kong, Shatin, Hong Kong; Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Impairments in social interaction/communication become apparent after 12 months of age in children who develop Autism spectrum disorder (ASD). Studies of baby siblings of children with ASD provide the means to detect changes in the brain that are present before behavioral symptoms appear. In this review, advances from brain imaging studies of infant siblings over the past 18 months are highlighted. RECENT FINDINGS During the first 2 months of life, functional differences in social brain regions and microstructural differences in dorsal language tracks are found in some high-risk baby siblings. At 4-6 months of age, differences in subcortical and cerebellum volumes and atypical cortical responses to social stimuli are evident. At 6 months, extra-axial cerebrospinal fluid is increased, and at 8 months there is evidence of cortical hyper-reactivity. Patterns of functional connectivity are distinct in infant siblings and suggest dysfunctional activation and integration of information across the cortex and neural networks underlying social behaviors. SUMMARY Further replication in very large independent samples is needed to verify the majority of the findings discussed and understand how they are related within individual infants. Much more research is needed before translation to clinical practice.
Collapse
|
40
|
Dickinson A, Varcin KJ, Sahin M, Nelson CA, Jeste SS. Early patterns of functional brain development associated with autism spectrum disorder in tuberous sclerosis complex. Autism Res 2019; 12:1758-1773. [PMID: 31419043 PMCID: PMC6898751 DOI: 10.1002/aur.2193] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 01/12/2023]
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic disorder that confers a high risk for autism spectrum disorders (ASD), with behavioral predictors of ASD emerging early in life. Deviations in structural and functional neural connectivity are highly implicated in both TSC and ASD. For the first time, we explore whether electroencephalographic (EEG) measures of neural network function precede or predict the emergence of ASD in TSC. We determine whether altered brain function (a) is present in infancy in TSC, (b) differentiates infants with TSC based on ASD diagnostic status, and (c) is associated with later cognitive function. We studied 35 infants with TSC (N = 35), and a group of typically developing infants (N = 20) at 12 and 24 months of age. Infants with TSC were later subdivided into ASD and non-ASD groups based on clinical evaluation. We measured features of spontaneous alpha oscillations (6-12 Hz) that are closely associated with neural network development: alpha power, alpha phase coherence (APC), and peak alpha frequency (PAF). Infants with TSC demonstrated reduced interhemispheric APC compared to controls at 12 months of age, and these differences were found to be most pronounced at 24 months in the infants who later developed ASD. Across all infants, PAF at 24 months was associated with verbal and nonverbal cognition at 36 months. Associations between early network function and later neurodevelopmental and cognitive outcomes highlight the potential utility of early scalable EEG markers to identify infants with TSC requiring additional targeted intervention initiated very early in life. Autism Res 2019, 12: 1758-1773. © 2019 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Approximately half of infants with tuberous sclerosis complex (TSC) develop autism. Here, using EEG, we find that there is a reduction in communication between brain regions during infancy in TSC, and that the infants who show the largest reductions are those who later develop autism. Being able to identify infants who show early signs of disrupted brain development may improve the timing of early prediction and interventions in TSC, and also help us to understand how early brain changes lead to autism.
Collapse
Affiliation(s)
- Abigail Dickinson
- UCLA Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, Los Angeles, California
| | - Kandice J Varcin
- Telethon Kids Institute, University of Western Australia, Subiaco, Western Australia, Australia
| | - Mustafa Sahin
- Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Charles A Nelson
- Division of Developmental Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
- Harvard Graduate School of Education, Cambridge, Massachusetts
| | - Shafali S Jeste
- UCLA Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, Los Angeles, California
| |
Collapse
|
41
|
Douglas PS. Pre-emptive Intervention for Autism Spectrum Disorder: Theoretical Foundations and Clinical Translation. Front Integr Neurosci 2019; 13:66. [PMID: 31798425 PMCID: PMC6877903 DOI: 10.3389/fnint.2019.00066] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 11/04/2019] [Indexed: 12/28/2022] Open
Abstract
Autism spectrum disorders (ASD) are an emergent public health problem, placing significant burden upon the individual, family and health system. ASD are polygenetic spectrum disorders of neural connectome development, in which one or more feedback loops amplify small genetic, structural, or functional variations in the very early development of motor and sensory-motor pathways. These perturbations trigger a 'butterfly effect' of unpredictable cascades of structural and functional imbalances in the global neuronal workspace, resulting in atypical behaviors, social communication, and cognition long-term. The first 100 days post-term are critically neuroplastic and comprise an injury-sensitive developmental window, characterized by a neural biomarker, the persistence of the cortical subplate, and a behavioral biomarker, the crying diathesis. By the time potential diagnostic signs are identified, from 6 months of age, ASD neuropathy is already entrenched. The International Society for Autism Research Special Interest Group has called for pre-emptive intervention, based upon rigorous theoretical frames, and real world translation and evaluation. This paper responds to that call. It synthesizes heterogenous evidence concerning ASD etiologies from both psychosocial and biological research literatures with complexity science and evolutionary biology, to propose a theoretical framework for pre-emptive intervention. This paper hypothesizes that environmental factors resulting from a mismatch between environment of evolutionary adaptedness and culture initiate or perpetuate early motor and sensory-motor lesions, triggering a butterfly effect of multi-directional cascades of atypical developmental in the complex adaptive system of the parent and ASD-susceptible infant. Chronic sympathetic nervous system/hypothalamic-pituitary-adrenal axis hyperarousal and disrupted parent-infant biobehavioral synchrony are the key biologic and behavioral mechanisms perpetuating these atypical developmental cascades. A clinical translation of this evidence is proposed, for application antenatally and in the first 6 months of life, as pre-emptive intervention for ASD.
Collapse
Affiliation(s)
- Pamela S. Douglas
- Transforming Maternity Care Collaborative, Griffith University, Brisbane, QLD, Australia
- Discipline of General Practice, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
42
|
Girault JB, Piven J. The Neurodevelopment of Autism from Infancy Through Toddlerhood. Neuroimaging Clin N Am 2019; 30:97-114. [PMID: 31759576 DOI: 10.1016/j.nic.2019.09.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Autism spectrum disorder (ASD) emerges during early childhood and is marked by a relatively narrow window in which infants transition from exhibiting normative behavioral profiles to displaying the defining features of the ASD phenotype in toddlerhood. Prospective brain imaging studies in infants at high familial risk for autism have revealed important insights into the neurobiology and developmental unfolding of ASD. In this article, we review neuroimaging studies of brain development in ASD from birth through toddlerhood, relate these findings to candidate neurobiological mechanisms, and discuss implications for future research and translation to clinical practice.
Collapse
Affiliation(s)
- Jessica B Girault
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill School of Medicine, 101 Renee Lynne Court, Chapel Hill, NC 27599, USA.
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill School of Medicine, 101 Renee Lynne Court, Chapel Hill, NC 27599, USA
| |
Collapse
|
43
|
Strunecka A, Strunecky O. Chronic Fluoride Exposure and the Risk of Autism Spectrum Disorder. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E3431. [PMID: 31527457 PMCID: PMC6765894 DOI: 10.3390/ijerph16183431] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/07/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022]
Abstract
The continuous rise of autism spectrum disorder (ASD) prevalent in the past few decades is causing an increase in public health and socioeconomic concern. A consensus suggests the involvement of both genetic and environmental factors in the ASD etiopathogenesis. Fluoride (F) is rarely recognized among the environmental risk factors of ASD, since the neurotoxic effects of F are not generally accepted. Our review aims to provide evidence of F neurotoxicity. We assess the risk of chronic F exposure in the ASD etiopathology and investigate the role of metabolic and mitochondrial dysfunction, oxidative stress and inflammation, immunoexcitotoxicity, and decreased melatonin levels. These symptoms have been observed both after chronic F exposure as well as in ASD. Moreover, we show that F in synergistic interactions with aluminum's free metal cation (Al3+) can reinforce the pathological symptoms of ASD. This reinforcement takes place at concentrations several times lower than when acting alone. A high ASD prevalence has been reported from countries with water fluoridation as well as from endemic fluorosis areas. We suggest focusing the ASD prevention on the reduction of the F and Al3+ burdens from daily life.
Collapse
Affiliation(s)
- Anna Strunecka
- The Institute of Technology and Business, Okružní 517/10, 370 01 České Budějovice, Czech Republic.
| | - Otakar Strunecky
- The Institute of Technology and Business, Okružní 517/10, 370 01 České Budějovice, Czech Republic.
| |
Collapse
|
44
|
Linking social motivation with social skill: The role of emotion dysregulation in autism spectrum disorder. Dev Psychopathol 2019; 31:931-943. [PMID: 30957732 DOI: 10.1017/s0954579419000361] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Autism spectrum disorder (ASD) is associated with pervasive social deficits as well as marked emotion dysregulation across the life span. Decreased social motivation accounts in part for social difficulties, but factors moderating its influence are not fully understood. In this paper, we (a) characterize social and emotional functioning among children and adolescents with ASD, (b) explore contributions of social motivation and emotion dysregulation to social skill, and (c) consider biological sex and intellectual functioning as moderators of these associations. In a sample of 2,079 children and adolescents with ASD from the Simons Simplex Collection, we document direct effects of social motivation, internalizing symptoms, aggression, attention problems, irritability, and self-injurious behavior on children's social skills. Furthermore, dysregulation in several domains moderated the association between social motivation and social skill, suggesting a blunting effect on social motivation in the context of emotional difficulties. Moreover, when considering only individuals with intellectual skills in the average range or higher, biological sex further moderated these associations. Findings add to our understanding of social-emotional processes in ASD, suggest emotion dysregulation as a target of intervention in the service of social skill improvements, and build on efforts to understand sources of individual difference that contribute to heterogeneity among individuals with ASD.
Collapse
|
45
|
Petanjek Z, Sedmak D, Džaja D, Hladnik A, Rašin MR, Jovanov-Milosevic N. The Protracted Maturation of Associative Layer IIIC Pyramidal Neurons in the Human Prefrontal Cortex During Childhood: A Major Role in Cognitive Development and Selective Alteration in Autism. Front Psychiatry 2019; 10:122. [PMID: 30923504 PMCID: PMC6426783 DOI: 10.3389/fpsyt.2019.00122] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
The human specific cognitive shift starts around the age of 2 years with the onset of self-awareness, and continues with extraordinary increase in cognitive capacities during early childhood. Diffuse changes in functional connectivity in children aged 2-6 years indicate an increase in the capacity of cortical network. Interestingly, structural network complexity does not increase during this time and, thus, it is likely to be induced by selective maturation of a specific neuronal subclass. Here, we provide an overview of a subclass of cortico-cortical neurons, the associative layer IIIC pyramids of the human prefrontal cortex. Their local axonal collaterals are in control of the prefrontal cortico-cortical output, while their long projections modulate inter-areal processing. In this way, layer IIIC pyramids are the major integrative element of cortical processing, and changes in their connectivity patterns will affect global cortical functioning. Layer IIIC neurons have a unique pattern of dendritic maturation. In contrast to other classes of principal neurons, they undergo an additional phase of extensive dendritic growth during early childhood, and show characteristic molecular changes. Taken together, circuits associated with layer IIIC neurons have the most protracted period of developmental plasticity. This unique feature is advanced but also provides a window of opportunity for pathological events to disrupt normal formation of cognitive circuits involving layer IIIC neurons. In this manuscript, we discuss how disrupted dendritic and axonal maturation of layer IIIC neurons may lead into global cortical disconnectivity, affecting development of complex communication and social abilities. We also propose a model that developmentally dictated incorporation of layer IIIC neurons into maturing cortico-cortical circuits between 2 to 6 years will reveal a previous (perinatal) lesion affecting other classes of principal neurons. This "disclosure" of pre-existing functionally silent lesions of other neuronal classes induced by development of layer IIIC associative neurons, or their direct alteration, could be found in different forms of autism spectrum disorders. Understanding the gene-environment interaction in shaping cognitive microcircuitries may be fundamental for developing rehabilitation and prevention strategies in autism spectrum and other cognitive disorders.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Domagoj Džaja
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mladen Roko Rašin
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Nataša Jovanov-Milosevic
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Medical Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
46
|
Drozd HP, Karathanasis SF, Molosh AI, Lukkes JL, Clapp DW, Shekhar A. From bedside to bench and back: Translating ASD models. PROGRESS IN BRAIN RESEARCH 2018; 241:113-158. [PMID: 30447753 DOI: 10.1016/bs.pbr.2018.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autism spectrum disorders (ASD) represent a heterogeneous group of disorders defined by deficits in social interaction/communication and restricted interests, behaviors, or activities. Models of ASD, developed based on clinical data and observations, are used in basic science, the "bench," to better understand the pathophysiology of ASD and provide therapeutic options for patients in the clinic, the "bedside." Translational medicine creates a bridge between the bench and bedside that allows for clinical and basic science discoveries to challenge one another to improve the opportunities to bring novel therapies to patients. From the clinical side, biomarker work is expanding our understanding of possible mechanisms of ASD through measures of behavior, genetics, imaging modalities, and serum markers. These biomarkers could help to subclassify patients with ASD in order to better target treatments to a more homogeneous groups of patients most likely to respond to a candidate therapy. In turn, basic science has been responding to developments in clinical evaluation by improving bench models to mechanistically and phenotypically recapitulate the ASD phenotypes observed in clinic. While genetic models are identifying novel therapeutics targets at the bench, the clinical efforts are making progress by defining better outcome measures that are most representative of meaningful patient responses. In this review, we discuss some of these challenges in translational research in ASD and strategies for the bench and bedside to bridge the gap to achieve better benefits to patients.
Collapse
Affiliation(s)
- Hayley P Drozd
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sotirios F Karathanasis
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrei I Molosh
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jodi L Lukkes
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - D Wade Clapp
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anantha Shekhar
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Clinical and Translation Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
47
|
Dadalko OI, Travers BG. Evidence for Brainstem Contributions to Autism Spectrum Disorders. Front Integr Neurosci 2018; 12:47. [PMID: 30337860 PMCID: PMC6180283 DOI: 10.3389/fnint.2018.00047] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/18/2018] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition that affects one in 59 children in the United States. Although there is a mounting body of knowledge of cortical and cerebellar contributions to ASD, our knowledge about the early developing brainstem in ASD is only beginning to accumulate. Understanding how brainstem neurotransmission is implicated in ASD is important because many of this condition’s sensory and motor symptoms are consistent with brainstem pathology. Therefore, the purpose of this review was to integrate epidemiological, behavioral, histological, neuroimaging, and animal evidence of brainstem contributions to ASD. Because ASD is a neurodevelopmental condition, we examined the available data through a lens of hierarchical brain development. The review of the literature suggests that developmental alterations of the brainstem could have potential cascading effects on cortical and cerebellar formation, ultimately leading to ASD symptoms. This view is supported by human epidemiology findings and data from animal models of ASD, showing that perturbed development of the brainstem substructures, particularly during the peak formation of the brainstem’s monoaminergic centers, may relate to ASD or ASD-like behaviors. Furthermore, we review evidence from human histology, psychophysiology, and neuroimaging suggesting that brainstem development and maturation may be atypical in ASD and may be related to key ASD symptoms, such as atypical sensorimotor features and social responsiveness. From this review there emerges the need of future research to validate early detection of the brainstem-based somatosensory and psychophysiological behaviors that emerge in infancy, and to examine the brainstem across the life span, while accounting for age. In all, there is preliminary evidence for brainstem involvement in ASD, but a better understanding of the brainstem’s role would likely pave the way for earlier diagnosis and treatment of ASD.
Collapse
Affiliation(s)
- Olga I Dadalko
- Motor and Brain Development Lab, Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Brittany G Travers
- Motor and Brain Development Lab, Occupational Therapy Program in the Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|