1
|
Castellano G, Bonnet Da Silva J, Pietropaolo S. The role of gene-environment interactions in social dysfunction: Focus on preclinical evidence from mouse studies. Neuropharmacology 2024; 261:110179. [PMID: 39369849 DOI: 10.1016/j.neuropharm.2024.110179] [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: 07/28/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
Human and animal research has demonstrated that genetic and environmental factors can strongly modulate behavioral function, including the expression of social behaviors and their dysfunctionalities. Several genes have been linked to pathologies characterized by alterations in social behaviors, e.g., aggressive/antisocial personality disorder (ASPD), or autism spectrum disorder (ASD). Environmental stimulation (e.g., physical exercise, environmental enrichment) or adversity (e.g., chronic stress, social isolation) may respectively improve or impair social interactions. While the independent contribution of genetic and environmental factors to social behaviors has been assessed in a variety of human and animal studies, the impact of their interactive effects on social functions has been less extensively investigated. Genetic mutations and environmental changes can indeed influence each other through complex mutual effects, e.g., inducing synergistic, antagonistic or interactive behavioral outcomes. This complexity is difficult to be disentangled in human populations, thus encouraging studies in animal models, especially in the mouse species which is the most suitable for genetic manipulations. Here we review the available preclinical evidence on the impact of gene-environment interactions on social behaviors and their dysfunction, focusing on studies in laboratory mice. We included findings combining naturally occurring mutations, selectively bred or transgenic mice with multiple environmental manipulations, including positive (environmental enrichment, physical exercise) and aversive (social isolation, maternal separation, and stress) experiences. The impact of these results is critically discussed in terms of their generalizability across mouse models and social tests, as well as their implications for human studies on social dysfunction.
Collapse
Affiliation(s)
- Giulia Castellano
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, F-33000, Bordeaux, France
| | | | | |
Collapse
|
2
|
Mediane DH, Basu S, Cahill EN, Anastasiades PG. Medial prefrontal cortex circuitry and social behaviour in autism. Neuropharmacology 2024; 260:110101. [PMID: 39128583 DOI: 10.1016/j.neuropharm.2024.110101] [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: 04/15/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Autism spectrum disorder (ASD) has proven to be highly enigmatic due to the diversity of its underlying genetic causes and the huge variability in symptom presentation. Uncovering common phenotypes across people with ASD and pre-clinical models allows us to better understand the influence on brain function of the many different genetic and cellular processes thought to contribute to ASD aetiology. One such feature of ASD is the convergent evidence implicating abnormal functioning of the medial prefrontal cortex (mPFC) across studies. The mPFC is a key part of the 'social brain' and may contribute to many of the changes in social behaviour observed in people with ASD. Here we review recent evidence for mPFC involvement in both ASD and social behaviours. We also highlight how pre-clinical mouse models can be used to uncover important cellular and circuit-level mechanisms that may underly atypical social behaviours in ASD. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Diego H Mediane
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Shinjini Basu
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Emma N Cahill
- Department of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, United Kingdom
| | - Paul G Anastasiades
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom.
| |
Collapse
|
3
|
Nehzomi ZS, Shirani K. Investigating the role of food pollutants in autism spectrum disorder: a comprehensive analysis of heavy metals, pesticides, and mycotoxins. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03551-4. [PMID: 39466439 DOI: 10.1007/s00210-024-03551-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
Food pollutants, including heavy metals, pesticides, and mycotoxins, have been proposed as potential risk factors for autism spectrum disorder (ASD) during pregnancy and early childhood. This paper examines the impact of food pollutants on ASD risk. A systematic search through PubMed, Google Scholar, and Sciverse yielded studies from 1990 to present. Research indicates elevated levels of heavy metals in children with ASD, linking pesticides and toxins to brain development disruptions. Mycotoxins, specifically, show a correlation with ASD and can contaminate food, posing a threat to neurodevelopment. Strategies like choosing organic foods and reducing exposure to toxins may benefit individuals with ASD and those vulnerable to the disorder. Further research is essential to comprehend the food pollutant-ASD relationship and devise effective exposure reduction strategies.
Collapse
Affiliation(s)
| | - Kobra Shirani
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
4
|
Luglio D, Kleeman MJ, Yu X, Lin JC, Chow T, Martinez MP, Chen Z, Chen JC, Eckel SP, Schwartz J, Lurmann F, McConnell R, Xiang AH, Rahman MM. Prenatal Exposure to Source-Specific Fine Particulate Matter and Autism Spectrum Disorder. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:18566-18577. [PMID: 39392704 PMCID: PMC11500427 DOI: 10.1021/acs.est.4c05563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
In this study, associations between prenatal exposure to fine particulate matter (PM2.5) from 9 sources and development of autism spectrum disorder (ASD) were assessed in a population-based retrospective pregnancy cohort in southern California. The cohort included 318,750 mother-child singleton pairs. ASD cases (N = 4559) were identified by ICD codes. Source-specific PM2.5 concentrations were estimated from a chemical transport model with a 4 × 4 km2 resolution and assigned to maternal pregnancy residential addresses. Cox proportional hazard models were used to estimate the hazard ratios (HR) of ASD development for each individual source. We also adjusted for total PM2.5 mass and in a separate model for all other sources simultaneously. Increased ASD risk was observed with on-road gasoline (HR [CI]: 1.18 [1.13, 1.24]), off-road gasoline (1.15 [1.12, 1.19]), off-road diesel (1.08 [1.05, 1.10]), food cooking (1.05 [1.02, 1.08]), aircraft (1.04 [1.01, 1.06]), and natural gas combustion (1.09 [1.06, 1.11]), each scaled to standard deviation increases in concentration. On-road gasoline and off-road gasoline were robust for other pollutant groups. PM2.5 emitted from different sources may have different impacts on ASD. The results also identify PM source mixtures for toxicological investigations that may provide evidence for future public health policies.
Collapse
Affiliation(s)
- David
G. Luglio
- Department
of Environmental Health Sciences, Tulane
University School of Public Health and Tropical Medicine, New Orleans, Louisiana 70118, United States
| | - Michael J. Kleeman
- Department
of Civil and Environmental Engineering, University of California, Davis, Davis, California 95616, United States
| | - Xin Yu
- Spatial
Science Institute, University of Southern
California, Los Angeles, California 90089, United States
| | - Jane C. Lin
- Department
of Research & Evaluation, Kaiser Permanente
Southern California, Pasadena, California 91101, United States
| | - Ting Chow
- Department
of Research & Evaluation, Kaiser Permanente
Southern California, Pasadena, California 91101, United States
| | - Mayra P. Martinez
- Department
of Research & Evaluation, Kaiser Permanente
Southern California, Pasadena, California 91101, United States
| | - Zhanghua Chen
- Department
of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Jiu-Chiuan Chen
- Department
of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Sandrah Proctor Eckel
- Department
of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Joel Schwartz
- Department
of Environmental Health, Harvard T.H. Chan
School of Public Health, Boston, Massachusetts 02115, United States
- Department
of Epidemiology, Harvard T.H. Chan School
of Public Health, Boston, Massachusetts 02115, United States
| | | | - Rob McConnell
- Department
of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Anny H. Xiang
- Department
of Research & Evaluation, Kaiser Permanente
Southern California, Pasadena, California 91101, United States
| | - Md Mostafijur Rahman
- Department
of Environmental Health Sciences, Tulane
University School of Public Health and Tropical Medicine, New Orleans, Louisiana 70118, United States
- Department
of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
5
|
Ashley SA, Merritt K, Solmi F, Laguna PL, Reichenberg A, David AS. A Longitudinal Study of Head Circumference Trajectories in Autism and Autistic Traits. J Autism Dev Disord 2024:10.1007/s10803-024-06578-x. [PMID: 39395125 DOI: 10.1007/s10803-024-06578-x] [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] [Accepted: 09/19/2024] [Indexed: 10/14/2024]
Abstract
Increased head circumference is an established finding in autism spectrum disorder (ASD); however, it is unclear when this increase occurs, if it persists and whether it manifests across the whole ASD spectrum. Head circumference is a strong predictor of brain size and can therefore provide key insights into brain development in ASD. We used data from the Avon Longitudinal Study of Parents and Children to compare head circumference trajectories from birth to 15 years in children with an ASD diagnosis (N = 78, controls = 6,404) or elevated autistic traits as measured using the Social Communication Disorder Checklist (N = 639, controls = 6,230). Exploratory analyses were conducted in those with ASD and co-morbid cognitive learning needs (CLN). Children with an ASD diagnosis had larger head circumference from birth across childhood and adolescence compared to controls in univariable (B = 0.69, 95% confidence interval [CI]: 0.28-1.09, p = 0.001) and multivariable models (B = 0.38, 95% CI: 0.003-0.75, p = 0.048). Differences were more marked in those with co-morbid CLN. Children with elevated autistic traits had significantly smaller head circumference compared to controls. There was weak evidence of group differences when height was included as a covariate. Head circumference trajectories in ASD deviate from control children and persist until adolescence. Autistic traits were associated with smaller head circumference, suggesting distinct growth trajectories between clinical cases from those with non-clinical traits.
Collapse
Affiliation(s)
- Sarah A Ashley
- Division of Psychiatry, University College London, London, UK.
| | - Kate Merritt
- Division of Psychiatry, University College London, London, UK
| | - Francesca Solmi
- Division of Psychiatry, University College London, London, UK
| | | | - Abraham Reichenberg
- Department of Psychiatry, Department of Environmental Medicine and Public Health, MINDICH Child and Health and Development Institute, and Seaver Center for Autism Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Anthony S David
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
6
|
King C, Rogers LG, Jansen J, Sivayokan B, Neyhard J, Warnes E, Hall SE, Plakke B. Adolescent treadmill exercise enhances hippocampal brain-derived neurotrophic factor (BDNF) expression and improves cognition in autism-modeled rats. Physiol Behav 2024; 284:114638. [PMID: 39004196 DOI: 10.1016/j.physbeh.2024.114638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by repetitive behaviors and altered communication abilities. Exercise is a low-cost intervention that could improve cognitive function and improve brain plasticity mechanisms. Here, the valproic acid (VPA) model was utilized to induce ASD-like phenotypes in rodents. Animals were exercised on a treadmill and performance was evaluated on a cognitive flexibility task. Biomarkers related to exercise and plasticity regulation were quantified from the prefrontal cortex, hippocampus, and skeletal muscle. Exercised VPA animals had higher levels of hippocampal BDNF compared to sedentary VPA animals and upregulated antioxidant enzyme expression in skeletal muscle. Cognitive improvements were demonstrated in both sexes, but in different domains of cognitive flexibility. This research demonstrates the benefits of exercise and provides evidence that molecular responses to exercise occur in both the central nervous system and in the periphery. These results suggest that improving regulation of BDNF via exercise, even at low intensity, could provide better synaptic regulation and cognitive benefits for individuals with ASD.
Collapse
Affiliation(s)
- Cole King
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Liza G Rogers
- Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Jeremy Jansen
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Bhavana Sivayokan
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Jenna Neyhard
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Ellie Warnes
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Stephanie E Hall
- Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Bethany Plakke
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
7
|
Liu Q, Yu D. Interaction and association between multiple vitamins and social adaptability and severity of autism: A large-scale retrospective study from China. Autism Res 2024. [PMID: 39327156 DOI: 10.1002/aur.3241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
Since children with autism spectrum disorder (ASD) often exhibit selective eating behaviors, it is generally believed that they may have abnormal nutrient structure, leading to aberrant concentrations of some serum vitamins. However, previous studies on serum vitamins in individuals with ASD are mixed. Additionally, the interaction and association between multiple serum vitamin and ASD-related symptoms remain unclear. This study utilized a cross-sectional survey with a large sample size (n = 1235) from China to clarify previous mixed findings, and examine the interaction and association between multiple serum vitamins (including folic acid [FA], vitamin A [VA], vitamin E [VE], vitamin B12 [VB12], and vitamin D [VD]) and social adaptability and symptom severity in children with ASD. Findings found that symptom severity was negatively associated with concentrations of serum VA, VE, VB12, and VD; while, social adaptability was significantly associated with the natural log-transformed concentrations of FA and VB12. Finding also revealed the interaction of VA and VE on the association between both vitamins and severity of ASD symptoms, as well as the interaction of VB12 and FA on the association between both vitamins and social adaptability. In particular, the combination of low concentration of VA and high concentration of VE is associated with the lowest risk of being "severely autistic"; while, the combination of low concentration of FA and high concentration of VB12 is associated with the lowest risk of being "poor social adaptability". This study offers the evidence for the requirement of considering multiple vitamins comprehensively, as well as valuable references for revealing the association between vitamin disparities and food selectivity in children with ASD.
Collapse
Affiliation(s)
- Qi Liu
- Henan Provincial Medical Key Lab of Child Developmental Behavior and Learning, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Engineering Research Center of Children's Digital Rehabilitation, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongchuan Yu
- Henan Provincial Medical Key Lab of Child Developmental Behavior and Learning, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Engineering Research Center of Children's Digital Rehabilitation, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, China
| |
Collapse
|
8
|
Sameea AA, Abd El-Wahab EW, Osman SO. Risk of Autism Spectrum Disorder (ASD) Among 18 to 48 Month Old Children: A Multi-Center Study in Qatar. Glob Pediatr Health 2024; 11:2333794X241284476. [PMID: 39323455 PMCID: PMC11423378 DOI: 10.1177/2333794x241284476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 08/07/2024] [Accepted: 09/02/2024] [Indexed: 09/27/2024] Open
Abstract
Background. Little is known about autism spectrum disorder (ASD) in Qatar. The lack of consensus in ASD screening has led to differences in the reported prevalence with escalating rates over time. Objective(s). To screen for ASD and to identify associate factors among Qatary children aged 18 to 48 months. Methods. A cross-sectional study included 600 eligible children at 10 Primary Health Care Centers (PHCCs) in Qatar. Identification of ASD was based on the M-CHAT™ criteria. Results. The number of ASD screen-positive children in the M-CHAT™ was 25.13 per 10.000, and was significantly associated with older maternal age (10.5%) and history of neonatal hyperbilirubinemia (20.0%) [AOR] = 4.88; 95%[CI]: 1.50-16.30)]. The Odds of detecting ASD was lower in children below 2 years of age (AOR = 0.92; 95% CI: 0.87-0.98]). Conclusion. This study demonstrates the utility of M-CHAT™ for ASD screening in PHCCs. Identification of the factors associated with positive ASD screening can highlight areas suitable for future intervention.
Collapse
Affiliation(s)
| | - Ekram Wassim Abd El-Wahab
- Tropical Health Department, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Sherif Omar Osman
- Tropical Health Department, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| |
Collapse
|
9
|
Baghdadli A, Peries M, Loubersac J, Michelon C, Rattaz C, Ferrando L, David A, Munir K, Picot MC. Contributions of the ELENA Cohort to Study Autism Spectrum Disorder in Children and Adolescents from a Biopsychosocial Framework. J Autism Dev Disord 2024:10.1007/s10803-024-06519-8. [PMID: 39230780 DOI: 10.1007/s10803-024-06519-8] [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] [Accepted: 08/08/2024] [Indexed: 09/05/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition influenced by a myriad of developmental, biological, psychological, and socio-demographic factors. The ELENA cohort seeks to delineate the intricate interplay of these factors, facilitating the identification of risk factors and the development of targeted interventions. This paper emphasizes the clinical profiles of children and outlines key findings from a biopsychosocial perspective. The ELENA cohort, a multicenter initiative across French regional centers, conducted a systematic prospective analysis on children newly diagnosed with DSM-5 ASD between 2012 and 2019. This encompassed direct assessments and parent-reported questionnaires covering a broad spectrum of developmental, biological, psychological and socio-demographic measures. Embedded case-control studies further examined risk and protective factors, alongside specific environmental and psychosocial influences during pregnancy and early childhood. A subset of participants also contributed biospecimens, with data enhancement via linkage to French National Administrative Healthcare Databases. The study unveils baseline clinical characteristics for 876 children, average age 6 (SD ± 3.3) previously unreported in protocol descriptions. It highlights the study's developmental biopsychosocial approach and its novel findings on children's socio-adaptive functioning, ASD severity, comorbidities, quality of life and interventions. Employing developmental biopsychosocial insights offers a promising pathway to integrating health, social care, and experiential insights, ultimately aiming to enhance the future well-being and outcomes for children with ASD. This approach underscores the need of a holistic, interdisciplinary strategy in encouraging and supporting the ASD community.
Collapse
Affiliation(s)
- Amaria Baghdadli
- Centre de Ressource Autisme Languedoc-Roussillon & Centre d'Excellence sur l'Autisme et les Troubles Neurodéveloppementaux (CeAND), CHU Montpellier, 39 Avenue Charles Flahaut, 34295, Montpellier Cedex 05, France.
- Centre d'Excellence sur l'Autisme et les Troubles du Neurodéveloppement (CeAND), CHU Montpellier, Montpellier, France.
- Université Paris-Saclay, UVSQ, Inserm, CESP, Team DevPsy, 94807, Villejuif, France.
- Faculté de Médecine, Université de Montpellier, Montpellier, France.
| | - Marianne Peries
- Centre de Ressource Autisme Languedoc-Roussillon & Centre d'Excellence sur l'Autisme et les Troubles Neurodéveloppementaux (CeAND), CHU Montpellier, 39 Avenue Charles Flahaut, 34295, Montpellier Cedex 05, France
- Centre d'Excellence sur l'Autisme et les Troubles du Neurodéveloppement (CeAND), CHU Montpellier, Montpellier, France
- Université Paris-Saclay, UVSQ, Inserm, CESP, Team DevPsy, 94807, Villejuif, France
| | - Julie Loubersac
- Centre de Ressource Autisme Languedoc-Roussillon & Centre d'Excellence sur l'Autisme et les Troubles Neurodéveloppementaux (CeAND), CHU Montpellier, 39 Avenue Charles Flahaut, 34295, Montpellier Cedex 05, France
- Centre d'Excellence sur l'Autisme et les Troubles du Neurodéveloppement (CeAND), CHU Montpellier, Montpellier, France
- Université Paris-Saclay, UVSQ, Inserm, CESP, Team DevPsy, 94807, Villejuif, France
| | - Cécile Michelon
- Centre de Ressource Autisme Languedoc-Roussillon & Centre d'Excellence sur l'Autisme et les Troubles Neurodéveloppementaux (CeAND), CHU Montpellier, 39 Avenue Charles Flahaut, 34295, Montpellier Cedex 05, France
- Centre d'Excellence sur l'Autisme et les Troubles du Neurodéveloppement (CeAND), CHU Montpellier, Montpellier, France
- Université Paris-Saclay, UVSQ, Inserm, CESP, Team DevPsy, 94807, Villejuif, France
| | - Cécile Rattaz
- Centre de Ressource Autisme Languedoc-Roussillon & Centre d'Excellence sur l'Autisme et les Troubles Neurodéveloppementaux (CeAND), CHU Montpellier, 39 Avenue Charles Flahaut, 34295, Montpellier Cedex 05, France
- Centre d'Excellence sur l'Autisme et les Troubles du Neurodéveloppement (CeAND), CHU Montpellier, Montpellier, France
| | - Laetitia Ferrando
- Centre de Ressource Autisme Languedoc-Roussillon & Centre d'Excellence sur l'Autisme et les Troubles Neurodéveloppementaux (CeAND), CHU Montpellier, 39 Avenue Charles Flahaut, 34295, Montpellier Cedex 05, France
- Centre d'Excellence sur l'Autisme et les Troubles du Neurodéveloppement (CeAND), CHU Montpellier, Montpellier, France
| | - Aurore David
- Centre de Ressource Autisme Languedoc-Roussillon & Centre d'Excellence sur l'Autisme et les Troubles Neurodéveloppementaux (CeAND), CHU Montpellier, 39 Avenue Charles Flahaut, 34295, Montpellier Cedex 05, France
- Centre d'Excellence sur l'Autisme et les Troubles du Neurodéveloppement (CeAND), CHU Montpellier, Montpellier, France
- Université Paris-Saclay, UVSQ, Inserm, CESP, Team DevPsy, 94807, Villejuif, France
| | - Kerim Munir
- Developmental Medicine Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marie-Christine Picot
- Centre d'Excellence sur l'Autisme et les Troubles du Neurodéveloppement (CeAND), CHU Montpellier, Montpellier, France
- Université Paris-Saclay, UVSQ, Inserm, CESP, Team DevPsy, 94807, Villejuif, France
- Clinical Research and Epidemiology Unit (Public Health Department), INSERM, Centre d'Investigation Clinique 1411, CHU Montpellier, Univ Montpellier, 34295, Montpellier Cedex 5, France
| |
Collapse
|
10
|
Duan X, Shan X, Uddin LQ, Chen H. The Future of Disentangling the Heterogeneity of Autism With Neuroimaging Studies. Biol Psychiatry 2024:S0006-3223(24)01536-1. [PMID: 39181387 DOI: 10.1016/j.biopsych.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
Autism spectrum disorder (ASD) is a lifelong neurodevelopmental condition. Over the past decade, a considerable number of approaches have been developed to identify potential neuroimaging-based biomarkers of ASD that have uncovered specific neural mechanisms that underlie behaviors associated with ASD. However, the substantial heterogeneity among individuals who are diagnosed with ASD hinders the development of biomarkers. Disentangling the heterogeneity of ASD is pivotal to improving the quality of life for individuals with ASD by facilitating early diagnosis and individualized interventions for those who need support. In this review, we discuss recent advances in neuroimaging that have facilitated the characterization of the heterogeneity of this condition using 3 frameworks: neurosubtyping, dimensional models, and normative models. We also discuss the challenges, possible solutions, and clinical utility of these 3 frameworks. We argue that several factors need to be considered when parsing heterogeneity using neuroimaging, including co-occurring conditions, neurodevelopment, heredity and environment, and multisite and multimodal data. We close with a discussion of future directions for achieving a better understanding of the neural mechanisms that underlie neurodevelopmental heterogeneity and the future of precision medicine in ASD.
Collapse
Affiliation(s)
- Xujun Duan
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China.
| | - Xiaolong Shan
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Lucina Q Uddin
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California; Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Huafu Chen
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
11
|
Frazier T. Autism screening is critical for the most vulnerable children. Dev Med Child Neurol 2024. [PMID: 39166419 DOI: 10.1111/dmcn.16043] [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: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 08/22/2024]
Affiliation(s)
- Thomas Frazier
- Department of Psychology, John Carroll University, University Heights, OH, USA
| |
Collapse
|
12
|
Nutor C, Dickerson AS, Hsu T, Al-Jadiri A, Camargo CA, Schweitzer JB, Shuster CL, Karagas MR, Madan JC, Restrepo B, Schmidt RJ, Lugo-Candelas C, Neiderhiser J, Sathyanarayana S, Dunlop AL, Brennan PA. Examining the association between prenatal cannabis exposure and child autism traits: A multi-cohort investigation in the environmental influences on child health outcome program. Autism Res 2024; 17:1651-1664. [PMID: 38953698 PMCID: PMC11341247 DOI: 10.1002/aur.3185] [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: 12/07/2023] [Accepted: 05/29/2024] [Indexed: 07/04/2024]
Abstract
This study examined the association between prenatal cannabis exposure and autism spectrum disorder (ASD) diagnoses and traits. A total sample of 11,570 children (ages 1-18; 53% male; 25% Hispanic; 60% White) from 34 cohorts of the National Institutes of Health-funded environmental influences on child health outcomes consortium were included in analyses. Results from generalized linear mixed models replicated previous studies showing that associations between prenatal cannabis exposure and ASD traits in children are not significant when controlling for relevant covariates, particularly tobacco exposure. Child biological sex did not moderate the association between prenatal cannabis exposure and ASD. In a large sample and measuring ASD traits continuously, there was no evidence that prenatal cannabis exposure increases the risk for ASD. This work helps to clarify previous mixed findings by addressing concerns about statistical power and ASD measurement.
Collapse
Affiliation(s)
- Chaela Nutor
- Department of Psychology, Emory University, Atlanta, Georgia, USA
| | - Aisha S Dickerson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Tingju Hsu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Aseel Al-Jadiri
- Institute for Child Development, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Carlos A Camargo
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julie B Schweitzer
- School of Medicine, University of California, Sacramento, California, USA
| | - Coral L Shuster
- Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA
| | | | - Juliette C Madan
- Department of Pediatrics, Psychiatry & Epidemiology, Children's Hospital at Dartmouth, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Bibiana Restrepo
- Department of Pediatrics, University of California Davis School of Medicine, MIND Institute, Sacramento, California, USA
| | - Rebecca J Schmidt
- Department of Public Health Sciences and the MIND Institute, University of California Davis School of Medicine, Davis, California, USA
| | | | - Jenae Neiderhiser
- Department of Psychology, Pennsylvania State University, University Park, Pennsylvania, USA
| | | | - Anne L Dunlop
- Department of Gynecology and Obstetrics, Emory University, Atlanta, Georgia, USA
| | | |
Collapse
|
13
|
Dubey MJ, Ghosh R, Dubey S, Das S, Chakraborty AP, Chatterjee S, Sengupta S, Benito-León J. Integrating parental psychopathologies in autism spectrum disorder care: Toward a holistic family-centric approach. Indian J Psychiatry 2024; 66:751-754. [PMID: 39398514 PMCID: PMC11469559 DOI: 10.4103/indianjpsychiatry.indianjpsychiatry_352_24] [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: 04/19/2024] [Accepted: 07/23/2024] [Indexed: 10/15/2024] Open
Affiliation(s)
- Mahua J. Dubey
- Department of Psychiatry, Berhampore Mental Hospital, Berhampore, West Bengal, India
| | - Ritwik Ghosh
- Department of General Medicine, Burdwan Medical College and Hospital, Burdwan, West Bengal, India
| | - Souvik Dubey
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, West Bengal, India
| | - Shambaditya Das
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, West Bengal, India
| | - Arka P. Chakraborty
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, West Bengal, India
| | - Subham Chatterjee
- Department of Psychiatry, Institute of Psychiatry- Center of Excellence, Kolkata, West Bengal, India
| | - Samya Sengupta
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, West Bengal, India
| | - Julián Benito-León
- Department of Neurology, University Hospital “12 de Octubre”, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
14
|
Kearns RJ, Nelson SM, Rex S. Epidural analgesia in labour: separating fact from fiction for autism and neurodevelopment. Br J Anaesth 2024; 133:247-254. [PMID: 38876925 DOI: 10.1016/j.bja.2024.05.009] [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: 03/22/2024] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 06/16/2024] Open
Abstract
Having epidural analgesia in labour has been associated with a later diagnosis of autism spectrum disorder in the offspring, resulting in concerns about childhood wellbeing. Neurodevelopmental changes are inconsistently reported in the literature, creating challenges in the interpretation of these findings. Here we explore the limitations of the current evidence base, and why findings differ between studies, concluding that the current body of evidence does not support a causal association between use of epidural analgesia in labour and autism spectrum disorder.
Collapse
Affiliation(s)
- Rachel J Kearns
- Department of Anaesthesia, Glasgow Royal Infirmary, Glasgow, UK.
| | | | - Steffen Rex
- Department of Anaesthesiology, University Hospitals Leuven, Leuven, Belgium; Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Lagod PP, Abdelli LS, Naser SA. An In Vivo Model of Propionic Acid-Rich Diet-Induced Gliosis and Neuro-Inflammation in Mice (FVB/N-Tg(GFAPGFP)14Mes/J): A Potential Link to Autism Spectrum Disorder. Int J Mol Sci 2024; 25:8093. [PMID: 39125662 PMCID: PMC11311704 DOI: 10.3390/ijms25158093] [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: 07/01/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Evidence shows that Autism Spectrum Disorder (ASD) stems from an interplay of genetic and environmental factors, which may include propionic acid (PPA), a microbial byproduct and food preservative. We previously reported that in vitro treatment of neural stem cells with PPA leads to gliosis and neuroinflammation. In this study, mice were exposed ad libitum to a PPA-rich diet for four weeks before mating. The same diet was maintained through pregnancy and administered to the offspring after weaning. The brains of the offspring were studied at 1 and 5 months postpartum. Glial fibrillary acidic protein (astrocytic marker) was significantly increased (1.53 ± 0.56-fold at 1 M and 1.63 ± 0.49-fold at 5 M) in the PPA group brains. Tubulin IIIβ (neuronal marker) was significantly decreased in the 5 M group. IL-6 and TNF-α expression were increased in the brain of the PPA group (IL-6: 2.48 ± 1.25-fold at 5 M; TNF-α: 2.84 ± 1.16-fold at 1 M and 2.64 ± 1.42-fold, at 5 M), while IL-10 was decreased. GPR41 and p-Akt were increased, while PTEN (p-Akt inhibitor) was decreased in the PPA group. The data support the role of a PPA-rich diet in glia over-proliferation and neuro-inflammation mediated by the GPR41 receptor and PTEN/Akt pathway. These findings strongly support our earlier study on the role of PPA in ASD.
Collapse
Affiliation(s)
- Piotr P. Lagod
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA;
| | - Latifa S. Abdelli
- Health Sciences Department, College of Health Professions and Sciences, University of Central Florida, Orlando, FL 32816, USA;
| | - Saleh A. Naser
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA;
| |
Collapse
|
16
|
King C, Plakke B. Maternal choline supplementation in neurodevelopmental disorders: mechanistic insights from animal models and future directions. Nutr Neurosci 2024:1-20. [PMID: 39046330 DOI: 10.1080/1028415x.2024.2377084] [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: 07/25/2024]
Abstract
OBJECTIVES To synthesize evidence from animal models of neurodevelopmental disorders (NDD) using maternal choline supplementation, to characterize current knowledge on the mechanisms of choline's protective effects against NDD, and to identify gaps in knowledge for future study. METHODS A literature review was conducted in PubMed to identify studies using prenatal choline supplementation interventions in rodent models of neurodevelopmental disorders. 24 studies were identified, and behavioral and biological findings were extracted from each. Studies examining both genetic and environmental risk factors were included. RESULTS Maternal choline supplementation during gestation is protective against both genetic and environmental NDD risk factors. Maternal choline supplementation improves both cognitive and affective outcomes throughout the lifespan in NDD models. Prenatal choline improved these outcomes through its participation in processes like neurogenesis, epigenetic regulation, and anti-inflammatory signaling. DISCUSSION Maternal choline supplementation improves behavioral and neurobiological outcomes in animal models of NDD, paralleling findings in humans. Animal models provide a unique opportunity to study the mechanisms by which gestational choline improves neurodevelopmental outcomes. This is especially important since nearly 90% of pregnant people in the United States are deficient in choline intake. However, much is still unknown about the mechanisms through which choline and its derivatives act. Further research into this topic, especially mechanistic studies in animal models, is critical to modernize maternal choline intake guidelines and to develop interventions to increase maternal choline intake in vulnerable populations.
Collapse
Affiliation(s)
- Cole King
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| | - Bethany Plakke
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
17
|
Jones JP, Williamson L, Konsoula Z, Anderson R, Reissner KJ, Parker W. Evaluating the Role of Susceptibility Inducing Cofactors and of Acetaminophen in the Etiology of Autism Spectrum Disorder. Life (Basel) 2024; 14:918. [PMID: 39202661 PMCID: PMC11355895 DOI: 10.3390/life14080918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 09/03/2024] Open
Abstract
More than 20 previously reported lines of independent evidence from clinical observations, studies in laboratory animal models, pharmacokinetic considerations, and numerous temporal and spatial associations indicate that numerous genetic and environmental factors leading to inflammation and oxidative stress confer vulnerability to the aberrant metabolism of acetaminophen during early development, leading to autism spectrum disorder (ASD). Contrary to this conclusion, multivariate analyses of cohort data adjusting for inflammation-associated factors have tended to show little to no risk of acetaminophen use for neurodevelopment. To resolve this discrepancy, here we use in silico methods to create an ideal (virtual) population of 120,000 individuals in which 50% of all cases of virtual ASD are induced by oxidative stress-associated cofactors and acetaminophen use. We demonstrate that Cox regression analysis of this ideal dataset shows little to no risk of acetaminophen use if the cofactors that create aberrant metabolism of acetaminophen are adjusted for in the analysis. Further, under-reporting of acetaminophen use is shown to be a considerable problem for this analysis, leading to large and erroneously low calculated risks of acetaminophen use. In addition, we argue that factors that impart susceptibility to acetaminophen-induced injury, and propensity for acetaminophen use itself, can be shared between the prepartum, peripartum, and postpartum periods, creating additional difficulty in the analysis of existing datasets to determine risks of acetaminophen exposure for neurodevelopment during a specific time frame. It is concluded that risks of acetaminophen use for neurodevelopment obtained from multivariate analysis of cohort data depend on underlying assumptions in the analyses, and that other evidence, both abundant and robust, demonstrate the critical role of acetaminophen in the etiology of ASD.
Collapse
Affiliation(s)
- John P. Jones
- WPLab, Inc., Durham, NC 27707, USA; (J.P.J.III); (Z.K.); (R.A.)
| | - Lauren Williamson
- Department of Biological Sciences, Northern Kentucky University, Highland Heights, KY 41099, USA;
| | | | - Rachel Anderson
- WPLab, Inc., Durham, NC 27707, USA; (J.P.J.III); (Z.K.); (R.A.)
| | - Kathryn J. Reissner
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - William Parker
- WPLab, Inc., Durham, NC 27707, USA; (J.P.J.III); (Z.K.); (R.A.)
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC 27599, USA;
| |
Collapse
|
18
|
De Marzio M, Lasky-Su J, Chu SH, Prince N, Litonjua AA, Weiss ST, Kelly RS, Glass KR. The metabolic role of vitamin D in children's neurodevelopment: a network study. Sci Rep 2024; 14:16929. [PMID: 39043876 PMCID: PMC11266698 DOI: 10.1038/s41598-024-67835-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Neurodevelopmental disorders are rapidly increasing in prevalence and have been linked to various environmental risk factors. Mounting evidence suggests a potential role of vitamin D in child neurodevelopment, though the causal mechanisms remain largely unknown. Here, we investigate how vitamin D deficiency affects children's communication development, particularly in relation to Autism Spectrum Disorder (ASD). We do so by developing an integrative network approach that combines metabolomic profiles, clinical traits, and neurodevelopmental data from a pediatric cohort. Our results show that low levels of vitamin D are associated with changes in the metabolic networks of tryptophan, linoleic, and fatty acid metabolism. These changes correlate with distinct ASD-related phenotypes, including delayed communication skills and respiratory dysfunctions. Additionally, our analysis suggests the kynurenine and serotonin sub-pathways may mediate the effect of vitamin D on early life communication development. Altogether, our findings provide metabolome-wide insights into the potential of vitamin D as a therapeutic option for ASD and other communication disorders.
Collapse
Grants
- R01HL091528 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K01HL153941 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K01 HL153941 NHLBI NIH HHS
- UH3 OD023268 ODCDC CDC HHS
- K01HL146980 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL141826 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K25HL168157 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL155749 NHLBI NIH HHS
- R01HL155749 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL123915 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Margherita De Marzio
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA.
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Su H Chu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Nicole Prince
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Augusto A Litonjua
- Division of Pulmonary Medicine, Golisano Children's Hospital, University of Rochester, Rochester, NY, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Rachel S Kelly
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kimberly R Glass
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
19
|
Le Belle JE, Condro M, Cepeda C, Oikonomou KD, Tessema K, Dudley L, Schoenfield J, Kawaguchi R, Geschwind D, Silva AJ, Zhang Z, Shokat K, Harris NG, Kornblum HI. Acute rapamycin treatment reveals novel mechanisms of behavioral, physiological, and functional dysfunction in a maternal inflammation mouse model of autism and sensory over-responsivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602602. [PMID: 39026891 PMCID: PMC11257517 DOI: 10.1101/2024.07.08.602602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Maternal inflammatory response (MIR) during early gestation in mice induces a cascade of physiological and behavioral changes that have been associated with autism spectrum disorder (ASD). In a prior study and the current one, we find that mild MIR results in chronic systemic and neuro-inflammation, mTOR pathway activation, mild brain overgrowth followed by regionally specific volumetric changes, sensory processing dysregulation, and social and repetitive behavior abnormalities. Prior studies of rapamycin treatment in autism models have focused on chronic treatments that might be expected to alter or prevent physical brain changes. Here, we have focused on the acute effects of rapamycin to uncover novel mechanisms of dysfunction and related to mTOR pathway signaling. We find that within 2 hours, rapamycin treatment could rapidly rescue neuronal hyper-excitability, seizure susceptibility, functional network connectivity and brain community structure, and repetitive behaviors and sensory over-responsivity in adult offspring with persistent brain overgrowth. These CNS-mediated effects are also associated with alteration of the expression of several ASD-,ion channel-, and epilepsy-associated genes, in the same time frame. Our findings suggest that mTOR dysregulation in MIR offspring is a key contributor to various levels of brain dysfunction, including neuronal excitability, altered gene expression in multiple cell types, sensory functional network connectivity, and modulation of information flow. However, we demonstrate that the adult MIR brain is also amenable to rapid normalization of these functional changes which results in the rescue of both core and comorbid ASD behaviors in adult animals without requiring long-term physical alterations to the brain. Thus, restoring excitatory/inhibitory imbalance and sensory functional network modularity may be important targets for therapeutically addressing both primary sensory and social behavior phenotypes, and compensatory repetitive behavior phenotypes.
Collapse
|
20
|
Herrera ML, Paraíso-Luna J, Bustos-Martínez I, Barco Á. Targeting epigenetic dysregulation in autism spectrum disorders. Trends Mol Med 2024:S1471-4914(24)00162-X. [PMID: 38971705 DOI: 10.1016/j.molmed.2024.06.004] [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: 03/10/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 07/08/2024]
Abstract
Autism spectrum disorders (ASD) comprise a range of neurodevelopmental pathologies characterized by deficits in social interaction and repetitive behaviors, collectively affecting almost 1% of the worldwide population. Deciphering the etiology of ASD has proven challenging due to the intricate interplay of genetic and environmental factors and the variety of molecular pathways affected. Epigenomic alterations have emerged as key players in ASD etiology. Their research has led to the identification of biomarkers for diagnosis and pinpointed specific gene targets for therapeutic interventions. This review examines the role of epigenetic alterations, resulting from both genetic and environmental influences, as a central causative factor in ASD, delving into its contribution to pathogenesis and treatment strategies.
Collapse
Affiliation(s)
- Macarena L Herrera
- Instituto de Neurociencias (Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain
| | - Juan Paraíso-Luna
- Instituto de Neurociencias (Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain
| | - Isabel Bustos-Martínez
- Instituto de Neurociencias (Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain
| | - Ángel Barco
- Instituto de Neurociencias (Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain.
| |
Collapse
|
21
|
Shao W, Su Y, Liu J, Liu Y, Zhao J, Fan X. Understanding the link between different types of maternal diabetes and the onset of autism spectrum disorders. DIABETES & METABOLISM 2024; 50:101543. [PMID: 38761920 DOI: 10.1016/j.diabet.2024.101543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024]
Abstract
Autism spectrum disorders (ASD) encompass a collection of neurodevelopmental disorders that exhibit impaired social interactions and repetitive stereotypic behaviors. Although the exact cause of these disorders remains unknown, it is widely accepted that both genetic and environmental factors contribute to their onset and progression. Recent studies have highlighted the potential negative impact of maternal diabetes on embryonic neurodevelopment, suggesting that intrauterine hyperglycemia could pose an additional risk to early brain development and contribute to the development of ASD. This paper presents a comprehensive analysis of the current research on the relationship between various forms of maternal diabetes, such as type 1 diabetes mellitus, type 2 diabetes mellitus, and gestational diabetes mellitus, and the likelihood of ASD in offspring. The study elucidates the potential mechanisms through which maternal hyperglycemia affects fetal development, involving metabolic hormones, immune dysregulation, heightened oxidative stress, and epigenetic alterations. The findings of this review offer valuable insights for potential preventive measures and evidence-based interventions targeting ASD.
Collapse
Affiliation(s)
- Wenyu Shao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yichun Su
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiayin Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yulong Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jinghui Zhao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
22
|
Carey ME, Kivumbi A, Rando J, Mesaros AC, Melnyk S, James SJ, Croen LA, Volk H, Lyall K. The association between prenatal oxidative stress levels measured by isoprostanes and offspring neurodevelopmental outcomes at 36 months. Brain Behav Immun Health 2024; 38:100775. [PMID: 38706573 PMCID: PMC11067487 DOI: 10.1016/j.bbih.2024.100775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Oxidative stress during pregnancy has been a mechanistic pathway implicated in autism development, yet few studies have examined this association directly. Here, we examined the association of prenatal levels of 8-iso-PGF2α, a widely used measure of oxidative stress, and several neurodevelopmental outcomes related to autism in children. Participants included 169 mother-child pairs from the Early Autism Risk Longitudinal Investigation (EARLI), which enrolled mothers who had an autistic child from a previous pregnancy and followed them through a subsequent pregnancy and until that child reached age 3 years. Maternal urine samples were collected during the second trimester of pregnancy and were later measured for levels of isoprostanes. Child neurodevelopmental assessments included the Mullen Scales of Early Learning (MSEL), the Social Responsiveness Scale (SRS), and the Vineland Adaptive Behavior Scale (VABS), and were conducted around 36 months of age. Primary analyses examined associations between interquartile range (IQR) increases in 8-iso-PGF2α levels, and total composite scores from each assessment using quantile regression. In adjusted analyses, we did not observe statistically significant associations, though estimates suggested modestly lower cognitive scores (β for MSEL = -3.68, 95% CI: -10.09, 2.70), and minor increases in autism-related trait scores (β for SRS T score = 1.68, 95% CI: -0.24, 3.60) with increasing 8-iso-PGF2α. These suggestive associations between decreased cognitive scores and increased autism-related traits with increasing prenatal oxidative stress point to the need for continued investigation in larger samples of the role of oxidative stress as a mechanistic pathway in autism and related neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Meghan E. Carey
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
| | - Apollo Kivumbi
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
| | - Juliette Rando
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
| | - A. Clementina Mesaros
- Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA, 17104, USA
| | - Stepan Melnyk
- Arkansas Children’s Hospital Research Institute, 13 Childrens Way, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR, 72205, USA
| | - S. Jill James
- Department of Pediatrics, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR, 72205, USA
| | - Lisa A. Croen
- Division of Research Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| | - Heather Volk
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD, 21205, USA
| | - Kristen Lyall
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
| | - the Early Autism Risk Longitudinal Investigation (EARLI) team
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA, 17104, USA
- Arkansas Children’s Hospital Research Institute, 13 Childrens Way, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR, 72205, USA
- Division of Research Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD, 21205, USA
| |
Collapse
|
23
|
Zheng L, Jiao Y, Zhong H, Tan Y, Yin Y, Liu Y, Liu D, Wu M, Wang G, Huang J, Wang P, Qin M, Wang M, Xiao Y, Lv T, Luo Y, Hu H, Hou ST, Kui L. Human-derived fecal microbiota transplantation alleviates social deficits of the BTBR mouse model of autism through a potential mechanism involving vitamin B 6 metabolism. mSystems 2024; 9:e0025724. [PMID: 38780265 PMCID: PMC11237617 DOI: 10.1128/msystems.00257-24] [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: 02/25/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental condition characterized by social communication deficiencies and stereotypic behaviors influenced by hereditary and/or environmental risk factors. There are currently no approved medications for treating the core symptoms of ASD. Human fecal microbiota transplantation (FMT) has emerged as a potential intervention to improve autistic symptoms, but the underlying mechanisms are not fully understood. In this study, we evaluated the effects of human-derived FMT on behavioral and multi-omics profiles of the BTBR mice, an established model for ASD. FMT effectively alleviated the social deficits in the BTBR mice and normalized their distinct plasma metabolic profile, notably reducing the elevated long-chain acylcarnitines. Integrative analysis linked these phenotypic changes to specific Bacteroides species and vitamin B6 metabolism. Indeed, vitamin B6 supplementation improved the social behaviors in BTBR mice. Collectively, these findings shed new light on the interplay between FMT and vitamin B6 metabolism and revealed a potential mechanism underlying the therapeutic role of FMT in ASD.IMPORTANCEAccumulating evidence supports the beneficial effects of human fecal microbiota transplantation (FMT) on symptoms associated with autism spectrum disorder (ASD). However, the precise mechanism by which FMT induces a shift in the microbiota and leads to symptom improvement remains incompletely understood. This study integrated data from colon-content metagenomics, colon-content metabolomics, and plasma metabolomics to investigate the effects of FMT treatment on the BTBR mouse model for ASD. The analysis linked the amelioration of social deficits following FMT treatment to the restoration of mitochondrial function and the modulation of vitamin B6 metabolism. Bacterial species and compounds with beneficial roles in vitamin B6 metabolism and mitochondrial function may further contribute to improving FMT products and designing novel therapies for ASD treatment.
Collapse
Affiliation(s)
- Lifeng Zheng
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
- Xbiome Co. Ltd., Shenzhen, China
| | - Yinming Jiao
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Haolin Zhong
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
| | - Yan Tan
- Xbiome Co. Ltd., Shenzhen, China
| | | | | | - Ding Liu
- Xbiome Co. Ltd., Shenzhen, China
| | - Manli Wu
- Xbiome Co. Ltd., Shenzhen, China
| | - Guoyun Wang
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | | | - Ping Wang
- Shenzhen Institute for Drug Control, Shenzhen, China
| | - Meirong Qin
- Shenzhen Institute for Drug Control, Shenzhen, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children’s Hospital of Fudan University, National Center for Children’s Health, Shanghai, China
| | - Yang Xiao
- Department of Hematology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Tiying Lv
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yangzi Luo
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Han Hu
- Xbiome Co. Ltd., Shenzhen, China
| | - Sheng-Tao Hou
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
| | - Ling Kui
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
24
|
Milanković V, Tasić T, Leskovac A, Petrović S, Mitić M, Lazarević-Pašti T, Novković M, Potkonjak N. Metals on the Menu-Analyzing the Presence, Importance, and Consequences. Foods 2024; 13:1890. [PMID: 38928831 PMCID: PMC11203375 DOI: 10.3390/foods13121890] [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: 05/07/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Metals are integral components of the natural environment, and their presence in the food supply is inevitable and complex. While essential metals such as sodium, potassium, magnesium, calcium, iron, zinc, and copper are crucial for various physiological functions and must be consumed through the diet, others, like lead, mercury, and cadmium, are toxic even at low concentrations and pose serious health risks. This study comprehensively analyzes the presence, importance, and consequences of metals in the food chain. We explore the pathways through which metals enter the food supply, their distribution across different food types, and the associated health implications. By examining current regulatory standards for maximum allowable levels of various metals, we highlight the importance of ensuring food safety and protecting public health. Furthermore, this research underscores the need for continuous monitoring and management of metal content in food, especially as global agricultural and food production practices evolve. Our findings aim to inform dietary recommendations, food fortification strategies, and regulatory policies, ultimately contributing to safer and more nutritionally balanced diets.
Collapse
Affiliation(s)
- Vedran Milanković
- VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia; (V.M.); (T.T.); (A.L.); (S.P.); (M.M.); (T.L.-P.)
| | - Tamara Tasić
- VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia; (V.M.); (T.T.); (A.L.); (S.P.); (M.M.); (T.L.-P.)
| | - Andreja Leskovac
- VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia; (V.M.); (T.T.); (A.L.); (S.P.); (M.M.); (T.L.-P.)
| | - Sandra Petrović
- VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia; (V.M.); (T.T.); (A.L.); (S.P.); (M.M.); (T.L.-P.)
| | - Miloš Mitić
- VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia; (V.M.); (T.T.); (A.L.); (S.P.); (M.M.); (T.L.-P.)
| | - Tamara Lazarević-Pašti
- VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia; (V.M.); (T.T.); (A.L.); (S.P.); (M.M.); (T.L.-P.)
| | - Mirjana Novković
- Group for Muscle Cellular and Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11000 Belgrade, Serbia;
| | - Nebojša Potkonjak
- VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia; (V.M.); (T.T.); (A.L.); (S.P.); (M.M.); (T.L.-P.)
| |
Collapse
|
25
|
Wikarska A, Roszak K, Roszek K. Mesenchymal Stem Cells and Purinergic Signaling in Autism Spectrum Disorder: Bridging the Gap between Cell-Based Strategies and Neuro-Immune Modulation. Biomedicines 2024; 12:1310. [PMID: 38927517 PMCID: PMC11201695 DOI: 10.3390/biomedicines12061310] [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: 04/28/2024] [Revised: 05/26/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of autism spectrum disorder (ASD) is still increasing, which means that this neurodevelopmental lifelong pathology requires special scientific attention and efforts focused on developing novel therapeutic approaches. It has become increasingly evident that neuroinflammation and dysregulation of neuro-immune cross-talk are specific hallmarks of ASD, offering the possibility to treat these disorders by factors modulating neuro-immunological interactions. Mesenchymal stem cell-based therapy has already been postulated as one of the therapeutic approaches for ASD; however, less is known about the molecular mechanisms of stem cell influence. One of the possibilities, although still underestimated, is the paracrine purinergic activity of MSCs, by which stem cells ameliorate inflammatory reactions. Modulation of adenosine signaling may help restore neurotransmitter balance, reduce neuroinflammation, and improve overall brain function in individuals with ASD. In our review article, we present a novel insight into purinergic signaling, including but not limited to the adenosinergic pathway and its role in neuroinflammation and neuro-immune cross-talk modulation. We anticipate that by achieving a greater understanding of the purinergic signaling contribution to ASD and related disorders, novel therapeutic strategies may be devised for patients with autism in the near future.
Collapse
Affiliation(s)
| | | | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100 Torun, Poland; (A.W.); (K.R.)
| |
Collapse
|
26
|
El-Ansary A, Alfawaz HA, Bacha AB, Al-Ayadhi LY. Combining Anti-Mitochondrial Antibodies, Anti-Histone, and PLA2/COX Biomarkers to Increase Their Diagnostic Accuracy for Autism Spectrum Disorders. Brain Sci 2024; 14:576. [PMID: 38928576 PMCID: PMC11201962 DOI: 10.3390/brainsci14060576] [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: 05/09/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in social interaction and restricted and repetitive behaviors. Oxidative stress may be a critical link between mitochondrial dysfunction and ASD as reactive oxygen species (ROS) generated from pro-oxidant environmental toxicants and activated immune cells can result in mitochondrial failure. Recently, mitochondrial dysfunction, autoimmunity, and abnormal lipid mediators have been identified in multiple investigations as an acknowledged etiological mechanism of ASD that can be targeted for therapeutic intervention. METHODS The relationship between lipid mediator markers linked to inflammation induction, such as phospholipase A2/cyclooxygenase-2 (PLA2/Cox-2), and the mitochondrial dysfunction marker anti-mitochondrial antibodies (AMA-M2), and anti-histone autoantibodies in the etiology of ASD was investigated in this study using combined receiver operating characteristic (ROC) curve analyses. This study also sought to identify the linear combination for a given set of markers that optimizes the partial area under ROC curves. This study included 40 age- and sex-matched controls and 40 ASD youngsters. The plasma of both groups was tested for PLA2/COX-2, AMA-M2, and anti-histone autoantibodies' levels using ELISA kits. ROC curves and logistic regression models were used in the statistical analysis. RESULTS Using the integrated ROC curve analysis, a notable rise in the area under the curve was noticed. Additionally, the combined markers had markedly improved specificity and sensitivity. CONCLUSIONS The current study suggested that measuring the predictive value of selected biomarkers related to mitochondrial dysfunction, autoimmunity, and lipid metabolism in children with ASD using a ROC curve analysis could lead to a better understanding of the etiological mechanism of ASD as well as its relationship with metabolism.
Collapse
Affiliation(s)
- Afaf El-Ansary
- Autism Center, Lotus Holistic Alternative Medical Center, Abu Dhabi P.O. Box 110281, United Arab Emirates
| | - Hanan A. Alfawaz
- Department of Food Science and Nutrition, College of Food & Agriculture Sciences, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia;
| | - Abir Ben Bacha
- Department of Biochemistry, College of Science, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia;
| | - Laila Y. Al-Ayadhi
- Department of Physiology, Faculty of Medicine, King Saud University, P.O. Box 2925, Riyadh 11461, Saudi Arabia
| |
Collapse
|
27
|
Nutor C, Dunlop A, Sadler O, Brennan PA. Prenatal Cannabis Use and Offspring Autism-Related Behaviors: Examining Maternal Stress as a Moderator in a Black American Cohort. J Autism Dev Disord 2024; 54:2355-2367. [PMID: 37097527 PMCID: PMC10127191 DOI: 10.1007/s10803-023-05982-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 04/26/2023]
Abstract
Prenatal cannabis use and maternal stress have been proposed as risk factors for autism spectrum disorder (ASD). Black mothers and mothers of lower socioeconomic status (SES) may be especially likely to experience high levels of stress. This study examined the impact of prenatal cannabis use and maternal stress (i.e., prenatal distress, racial discrimination, and lower SES) on child ASD-related behaviors in a sample of 172 Black mother-child pairs. We found that prenatal stress was significantly associated with ASD-related behaviors. Prenatal cannabis use did not predict ASD-related behaviors and did not interact with maternal stress to predict ASD-related behaviors. These findings replicate previous work on prenatal stress-ASD associations and add to the limited literature on prenatal cannabis-ASD associations in Black samples.
Collapse
Affiliation(s)
- C Nutor
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA.
| | - A Dunlop
- Department of Gynecology and Obstetrics, Emory University, 1365 E Clifton Rd NE, Atlanta, GA, 30322, USA
| | - O Sadler
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA
| | - P A Brennan
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA
| |
Collapse
|
28
|
Dudas A, Nakahara TS, Pellissier LP, Chamero P. Parenting behaviors in mice: Olfactory mechanisms and features in models of autism spectrum disorders. Neurosci Biobehav Rev 2024; 161:105686. [PMID: 38657845 DOI: 10.1016/j.neubiorev.2024.105686] [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: 11/30/2023] [Revised: 03/24/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Rodents, along with numerous other mammals, heavily depend on olfactory cues to navigate their social interactions. Processing of olfactory sensory inputs is mediated by conserved brain circuits that ultimately trigger social behaviors, such as social interactions and parental care. Although innate, parenting is influenced by internal states, social experience, genetics, and the environment, and any significant disruption of these factors can impact the social circuits. Here, we review the molecular mechanisms and social circuits from the olfactory epithelium to central processing that initiate parental behaviors and their dysregulations that may contribute to the social impairments in mouse models of autism spectrum disorders (ASD). We discuss recent advances of the crucial role of olfaction in parental care, its consequences for social interactions, and the reciprocal influence on social interaction impairments in mouse models of ASD.
Collapse
Affiliation(s)
- Ana Dudas
- Team biology of GPCR Signaling systems (BIOS), CNRS, INRAE, University of Tours, PRC, Nouzilly F-37380, France
| | - Thiago S Nakahara
- Team Neuroendocrine Integration of Reproduction and Behavior (INERC), CNRS, INRAE, University of Tours, PRC, Nouzilly F-37380, France
| | - Lucie P Pellissier
- Team biology of GPCR Signaling systems (BIOS), CNRS, INRAE, University of Tours, PRC, Nouzilly F-37380, France.
| | - Pablo Chamero
- Team Neuroendocrine Integration of Reproduction and Behavior (INERC), CNRS, INRAE, University of Tours, PRC, Nouzilly F-37380, France.
| |
Collapse
|
29
|
Wang Z, Zhang B, Mu C, Qiao D, Chen H, Zhao Y, Cui H, Zhang R, Li S. Androgen levels in autism spectrum disorders: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 2024; 15:1371148. [PMID: 38779452 PMCID: PMC11109388 DOI: 10.3389/fendo.2024.1371148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/04/2024] [Indexed: 05/25/2024] Open
Abstract
Background Accumulating evidence suggests that the autism spectrum disorder (ASD) population exhibits altered hormone levels, including androgens. However, studies on the regulation of androgens, such as testosterone and dehydroepiandrosterone (DHEA), in relation to sex differences in individuals with ASD are limited and inconsistent. We conducted the systematic review with meta-analysis to quantitatively summarise the blood, urine, or saliva androgen data between individuals with ASD and controls. Methods A systematic search was conducted for eligible studies published before 16 January 2023 in six international and two Chinese databases. We computed summary statistics with a random-effects model. Publication bias was assessed using funnel plots and heterogeneity using I2 statistics. Subgroup analysis was performed by age, sex, sample source, and measurement method to explain the heterogeneity. Results 17 case-control studies (individuals with ASD, 825; controls, 669) were assessed. Androgen levels were significantly higher in individuals with ASD than that in controls (SMD: 0.27, 95% CI: 0.06-0.48, P=0.01). Subgroup analysis showed significantly elevated levels of urinary total testosterone, urinary DHEA, and free testosterone in individuals with ASD. DHEA level was also significantly elevated in males with ASD. Conclusion Androgen levels, especially free testosterone, may be elevated in individuals with ASD and DHEA levels may be specifically elevated in males.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Bohan Zhang
- Department of Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Chenyu Mu
- Department of Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Dan Qiao
- Department of Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Huan Chen
- Department of Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Yan Zhao
- School of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Huixian Cui
- Department of Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Rong Zhang
- Neuroscience Research Institute, Key Laboratory for Neuroscience, Ministry of Education of China, Peking University, Beijing, China
- Key Laboratory for Neuroscience, National Committee of Health, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Autism Research Center of Peking University Health Science Center, Beijing, China
| | - Sha Li
- Department of Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| |
Collapse
|
30
|
Yin W, Pulakka A, Reichenberg A, Kolevzon A, Ludvigsson JF, Risnes K, Lahti-Pulkkinen M, Persson M, Silverman ME, Åden U, Kajantie E, Sandin S. Association between parental psychiatric disorders and risk of offspring autism spectrum disorder: a Swedish and Finnish population-based cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2024; 40:100902. [PMID: 38689608 PMCID: PMC11059471 DOI: 10.1016/j.lanepe.2024.100902] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 05/02/2024]
Abstract
Background Roughly more than one in six adults worldwide suffer from psychiatric conditions. Sporadic studies have associated parental psychiatric disorders with autism spectrum disorder in offspring. Comprehensively examining the association between parental psychiatric disorders and offspring autism spectrum disorder is needed to guide health policies, and to inform etiologic studies. Methods We included all children born in Sweden and Finland 1997-2016. Diagnoses were clinically ascertained from National Registers through 2017. We calculated adjusted hazard ratios (aHRs) and 95% confidence intervals (CIs) for autism spectrum disorder in offspring of fathers and mothers with psychiatric disorders, in both parents jointly and across co-occurring conditions. Findings Among 2,505,842 children, 33,612 were diagnosed with autism spectrum disorder, of which 20% had a parent with psychiatric disorders. The risk of autism spectrum disorder was increased across all psychiatric disorders in fathers (Sweden: aHR = 2.02, 95% CI = 1.92-2.12; Finland: aHR = 1.63, 95% CI = 1.50-1.77), mothers (Sweden: aHR = 2.34, 95% CI = 2.24-2.43; Finland aHR = 2.12, 95% CI = 1.92-2.28), or both parents (Sweden: aHR = 3.76, 95% CI = 3.48-4.07; Finland aHR = 3.61, 95% CI = 3.20-4.07), compared to neither parents. Co-occurrence of parental psychiatric disorders further increased risk (e.g., Sweden: for one, two or ≥three different diagnostic categories compared to no diagnosis, in fathers aHR = 1.81, 2.07, 2.52; in mothers aHR = 2.05, 2.63, 3.57). Interpretation Psychiatric disorders in both parents conveyed the highest risk of offspring autism spectrum disorder, followed by mothers and then fathers. The risk increased with number of co-occurring disorders. All parental psychiatric disorders were associated with increased the risk of autism spectrum disorder. To reliably assess the risk of autism spectrum disorder in children, a comprehensive history incorporating the full range of parental psychiatric disorders is needed beyond solely focusing on familial autism spectrum disorder. Funding Swedish-Research-Council-2021-0214.
Collapse
Affiliation(s)
- Weiyao Yin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Anna Pulakka
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki and Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Abraham Reichenberg
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Seaver Center for Autism Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Kolevzon
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Seaver Center for Autism Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonas F. Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatrics, Örebro University Hospital, Örebro, Sweden
| | - Kari Risnes
- Department of Clinical and Molecular Medicine, NTNU, Trondheim, Norway
- Children’s Clinic, St Olav University Hospital, Trondheim, Norway
| | - Marius Lahti-Pulkkinen
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki and Oulu, Finland
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Finland
| | - Martina Persson
- Department of Medicine, Clinical Epidemiological Unit, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Science and Education, Division of Pediatrics, Karolinska Institutet, Stockholm, Sweden
- Sachsska Childrens’ and Youth Hospital, Stockholm, Sweden
| | - Michael E. Silverman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ulrika Åden
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Eero Kajantie
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki and Oulu, Finland
- Department of Clinical and Molecular Medicine, NTNU, Trondheim, Norway
- Clinical Medicine Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Sven Sandin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Seaver Center for Autism Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
31
|
King C, Mali I, Strating H, Fangman E, Neyhard J, Payne M, Bossmann SH, Plakke B. Region-Specific Brain Volume Changes Emerge in Adolescence in the Valproic Acid Model of Autism and Parallel Human Findings. Dev Neurosci 2024:1-12. [PMID: 38679020 PMCID: PMC11511791 DOI: 10.1159/000538932] [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: 01/11/2024] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and communication deficits, cognitive dysfunction, and stereotyped repetitive behaviors. Regional volume changes are commonly observed in individuals with ASD. To examine volumetric dysregulation across adolescence, the valproic acid (VPA) model was used to induce ASD-like phenotypes in rats. METHOD Regional volumes were obtained via magnetic resonance imaging at either postnatal day 28 or postnatal day 40 (P40), which correspond to early and late adolescence, respectively. RESULTS Consistent with prior research, VPA animals had reduced total brain volume compared to control animals. A novel outcome was that VPA animals had overgrown right hippocampi at P40. Differences in the pattern of development of the anterior cingulate cortex were also observed in VPA animals. Differences for the posterior cingulate were only observed in males, but not females. CONCLUSION These results demonstrate differences in region-specific developmental trajectories between control and VPA animals and suggest that the VPA model may capture regional volume changes consistent with human ASD.
Collapse
Affiliation(s)
- Cole King
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506
| | - Ivina Mali
- Kansas State University, Department of Chemistry, 1212 Mid-Campus Dr. N, Manhattan, KS 66506
| | - Hunter Strating
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506
| | - Elizabeth Fangman
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506
| | - Jenna Neyhard
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506
| | - Macy Payne
- Kansas State University, Department of Chemistry, 1212 Mid-Campus Dr. N, Manhattan, KS 66506
| | - Stefan H. Bossmann
- Kansas State University, Department of Chemistry, 1212 Mid-Campus Dr. N, Manhattan, KS 66506
| | - Bethany Plakke
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506
| |
Collapse
|
32
|
Blasco-Fontecilla H, Li C, Vizcaino M, Fernández-Fernández R, Royuela A, Bella-Fernández M. A Nomogram for Predicting ADHD and ASD in Child and Adolescent Mental Health Services (CAMHS). J Clin Med 2024; 13:2397. [PMID: 38673670 PMCID: PMC11051553 DOI: 10.3390/jcm13082397] [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: 03/05/2024] [Revised: 04/08/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Objectives: To enhance the early detection of Attention Deficit/Hyperactivity Disorder (ADHD) and Autism Spectrum Disorder (ASD) by leveraging clinical variables collected at child and adolescent mental health services (CAMHS). Methods: This study included children diagnosed with ADHD and/or ASD (n = 857). Three logistic regression models were developed to predict the presence of ADHD, its subtypes, and ASD. The analysis began with univariate logistic regression, followed by a multicollinearity diagnostic. A backward logistic regression selection strategy was then employed to retain variables with p < 0.05. Ethical approval was obtained from the local ethics committee. The models' internal validity was evaluated based on their calibration and discriminative abilities. Results: The study produced models that are well-calibrated and validated for predicting ADHD (incorporating variables such as physical activity, history of bone fractures, and admissions to pediatric/psychiatric services) and ASD (including disability, gender, special education needs, and Axis V diagnoses, among others). Conclusions: Clinical variables can play a significant role in enhancing the early identification of ADHD and ASD.
Collapse
Affiliation(s)
- Hilario Blasco-Fontecilla
- Instituto de Investigación, Transferencia e Innovación, Ciencias de la Saludy Escuela de Doctorado, Universidad Internacional de La Rioja, 26006 Logroño, Spain
- Center of Biomedical Network Research on Mental Health (CIBERSAM), Carlos III Institute of Health, 28029 Madrid, Spain
| | - Chao Li
- Faculty of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| | | | | | - Ana Royuela
- Biostatistics Unit, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Majadahonda, Spain;
| | - Marcos Bella-Fernández
- Puerta de Hierro University Hospital, 28222 Majadahonda, Spain;
- Faculty of Psychology, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
33
|
Baia-da-Silva DC, Mendes PFS, Silva DCBD, Chemelo VS, Bittencourt LO, Padilha PM, Oriá RB, Aschner M, Lima RR. What does scientometry tell us about mercury toxicology and its biological impairments? Heliyon 2024; 10:e27526. [PMID: 38586377 PMCID: PMC10998116 DOI: 10.1016/j.heliyon.2024.e27526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 04/09/2024] Open
Abstract
Mercury is a toxic pollutant that poses risks to both human and environmental health, making it a pressing public health concern. This study aimed to summarize the knowledge on mercury toxicology and the biological impairments caused by exposure to mercury in experimental studies and/or diagnosis in humans. The research was conducted on the main collection of Web of Science, employing as a methodological tool a bibliometric analysis. The selected articles were analyzed, and extracted data such as publication year, journal, author, title, number of citations, corresponding author's country, keywords, and the knowledge mapping was performed about the type of study, chemical form of mercury, exposure period, origin of exposure, tissue/fluid of exposure measurement, mercury concentration, evaluation period (age), mercury effect, model experiments, dose, exposure pathway, and time of exposure. The selected articles were published between 1965 and 2021, with Clarkson TW being the most cited author who has also published the most articles. A total of 38% of the publications were from the USA. These studies assessed the prenatal and postnatal effects of mercury, emphasizing the impact of methylmercury on neurodevelopment, including motor and cognitive evaluations, the association between mercury and autism, and an evaluation of its protective effects against mercury toxicity. In observational studies, the blood, umbilical cord, and hair were the most frequently used for measuring mercury levels. Our data analysis reveals that mercury neurotoxicology has been extensively explored, but the association among the outcomes evaluated in experimental studies has yet to be strengthened. Providing metric evidence on what is unexplored allows for new studies that may help governmental and non-governmental organizations develop guidelines and policies.
Collapse
Affiliation(s)
- Daiane Claydes Baia-da-Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Paulo Fernando Santos Mendes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Diane Cleydes Baia da Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Victória Santos Chemelo
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Pedro Magalhães Padilha
- School of Veterinary Medicine and Animal Science, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Reinaldo Barreto Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| |
Collapse
|
34
|
Caballero M, Satterstrom FK, Buxbaum JD, Mahjani B. Identification of moderate effect size genes in autism spectrum disorder through a novel gene pairing approach. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.03.24305278. [PMID: 38633780 PMCID: PMC11023658 DOI: 10.1101/2024.04.03.24305278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Autism Spectrum Disorder (ASD) arises from complex genetic and environmental factors, with inherited genetic variation playing a substantial role. This study introduces a novel approach to uncover moderate effect size (MES) genes in ASD, which individually do not meet the ASD liability threshold but collectively contribute when paired with specific other MES genes. Analyzing 10,795 families from the SPARK dataset, we identified 97 MES genes forming 50 significant gene pairs, demonstrating a substantial association with ASD when considered in tandem, but not individually. Our method leverages familial inheritance patterns and statistical analyses, refined by comparisons against control cohorts, to elucidate these gene pairs' contribution to ASD liability. Furthermore, expression profile analyses of these genes in brain tissues underscore their relevance to ASD pathology. This study underscores the complexity of ASD's genetic landscape, suggesting that gene combinations, beyond high impact single-gene mutations, significantly contribute to the disorder's etiology and heterogeneity. Our findings pave the way for new avenues in understanding ASD's genetic underpinnings and developing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Madison Caballero
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - F Kyle Satterstrom
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph D. Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Behrang Mahjani
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
35
|
Okabe M, Sato T, Takahashi M, Honjo A, Okawa M, Ishida M, Kukimoto-Niino M, Shirouzu M, Miyamoto Y, Yamauchi J. Autism Spectrum Disorder- and/or Intellectual Disability-Associated Semaphorin-5A Exploits the Mechanism by Which Dock5 Signalosome Molecules Control Cell Shape. Curr Issues Mol Biol 2024; 46:3092-3107. [PMID: 38666924 PMCID: PMC11049140 DOI: 10.3390/cimb46040194] [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: 02/12/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that includes autism, Asperger's syndrome, and pervasive developmental disorder. Individuals with ASD may exhibit difficulties in social interactions, communication challenges, repetitive behaviors, and restricted interests. While genetic mutations in individuals with ASD can either activate or inactivate the activities of the gene product, impacting neuronal morphogenesis and causing symptoms, the underlying mechanism remains to be fully established. Herein, for the first time, we report that genetically conserved Rac1 guanine-nucleotide exchange factor (GEF) Dock5 signalosome molecules control process elongation in the N1E-115 cell line, a model line capable of achieving neuronal morphological changes. The increased elongation phenotypes observed in ASD and intellectual disability (ID)-associated Semaphorin-5A (Sema5A) Arg676-to-Cys [p.R676C] were also mediated by Dock5 signalosome molecules. Indeed, knockdown of Dock5 using clustered regularly interspaced short palindromic repeat (CRISPR)/CasRx-based guide(g)RNA specifically recovered the mutated Sema5A-induced increase in process elongation in cells. Knockdown of Elmo2, an adaptor molecule of Dock5, also exhibited similar recovery. Comparable results were obtained when transfecting the interaction region of Dock5 with Elmo2. The activation of c-Jun N-terminal kinase (JNK), one of the primary signal transduction molecules underlying process elongation, was ameliorated by either their knockdown or transfection. These results suggest that the Dock5 signalosome comprises abnormal signaling involved in the process elongation induced by ASD- and ID-associated Sema5A. These molecules could be added to the list of potential therapeutic target molecules for abnormal neuronal morphogenesis in ASD at the molecular and cellular levels.
Collapse
Affiliation(s)
- Miyu Okabe
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Takanari Sato
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Mikito Takahashi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Asahi Honjo
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Maho Okawa
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Miki Ishida
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Mutsuko Kukimoto-Niino
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, Yokohama 230-0045, Japan; (M.K.-N.); (M.S.)
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, Yokohama 230-0045, Japan; (M.K.-N.); (M.S.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, Yokohama 230-0045, Japan; (M.K.-N.); (M.S.)
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
36
|
Zamstein O, Sheiner E, Binyamin Y, Pariente G, Wainstock T. Examining the relationship between autism spectrum disorder in children whose mother had labour epidural analgesia for their birth: A retrospective cohort study. Eur J Anaesthesiol 2024; 41:282-287. [PMID: 38084085 DOI: 10.1097/eja.0000000000001932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
BACKGROUND Controversy exists regarding the association between autism spectrum disorder (ASD) in children whose mother had labour epidural analgesia for their birth, as the few existing investigations have reported mixed findings. OBJECTIVE This study aims to evaluate the possibility of an association in our heterogeneous population. DESIGN A retrospective population-based cohort study. SETTING Vaginal deliveries that took place between the years 2005 and 2017 at Soroka University Medical Center, a tertiary referral hospital in Israel, and a follow-up on the incidence of ASD in the children. PATIENTS A hundred and thirty-nine thousand, nine hundred and eighty-one labouring patients and their offspring. MAIN OUTCOME MEASURES The incidence of children diagnosed with ASD (both hospital and community-based diagnoses) was compared based on whether their mothers had received labour epidural analgesia during their labour. A Kaplan-Meier survival curve compared cumulative incidence of ASD. A Cox proportional hazards model was used to control for relevant confounders. RESULTS Labour epidural analgesia was administered to 33 315 women. Epidural analgesia was more common among high-risk pregnancy groups (including pregnancies complicated with diabetes mellitus, hypertensive disorders, intrauterine growth restriction, and oligohydramnios; P < 0.001). In a Cox proportional hazards model, the association between epidural analgesia during labour and ASD in the children lost statistical significance following adjustment for confounders such as maternal age, gestational age, hypertensive disorders, diabetes mellitus, and ethnicity [adjusted hazard ratio = 1.13, 95% confidence interval (CI), 0.96 to 1.34, P = 0.152]. CONCLUSION In our population, after adjusting for confounders, epidural analgesia is not independently associated with autism spectrum disorder in the children. These findings enhance our knowledge regarding the safety of epidural analgesia and enable patients to make informed decisions about their pain relief techniques during labour.
Collapse
Affiliation(s)
- Omri Zamstein
- From the Obstetrics and Gynecology Division, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel (OZ, ES, GP), Department of Anesthesiology, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel (YB) and Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel (TW)
| | | | | | | | | |
Collapse
|
37
|
Wei Q, Xiao Y, Yang T, Chen J, Chen L, Wang K, Zhang J, Li L, Jia F, Wu L, Hao Y, Ke X, Yi M, Hong Q, Chen J, Fang S, Wang Y, Wang Q, Jin C, Xu X, Li T. Predicting autism spectrum disorder using maternal risk factors: A multi-center machine learning study. Psychiatry Res 2024; 334:115789. [PMID: 38452495 DOI: 10.1016/j.psychres.2024.115789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 02/02/2024] [Accepted: 02/11/2024] [Indexed: 03/09/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with a complex environmental etiology involving maternal risk factors, which have been combined with machine learning to predict ASD. However, limited studies have considered the factors throughout preconception, perinatal, and postnatal periods, and even fewer have been conducted in multi-center. In this study, five predictive models were developed using 57 maternal risk factors from a cohort across ten cities (ASD:1232, typically developing[TD]: 1090). The extreme gradient boosting model performed best, achieving an accuracy of 66.2 % on the external cohort from three cities (ASD:266, TD:353). The most important risk factors were identified as unstable emotions and lack of multivitamin supplementation using Shapley values. ASD risk scores were calculated based on predicted probabilities from the optimal model and divided into low, medium, and high-risk groups. The logistic analysis indicated that the high-risk group had a significantly increased risk of ASD compared to the low-risk group. Our study demonstrated the potential of machine learning models in predicting the risk for ASD based on maternal factors. The developed model provided insights into the maternal emotion and nutrition factors associated with ASD and highlighted the potential clinical applicability of the developed model in identifying high-risk populations.
Collapse
Affiliation(s)
- Qiuhong Wei
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China
| | - Yuanjie Xiao
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China
| | - Ting Yang
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China
| | - Jie Chen
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China
| | - Li Chen
- Department of Children's Healthcare, Children's Hospital of Chongqing Medical University, China
| | - Ke Wang
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China; Big Data Center for Children's Medical Care, Children's Hospital of Chongqing Medical University, No. 136. Zhongshan Er Rd, Yuzhong District, Chongqing 400014, China
| | - Jie Zhang
- Xi'an Children's Hospital, Xi'an, China
| | - Ling Li
- Department of Children Rehabilitation, Hainan Women and Children's Medical Center, Haikou, China
| | - Feiyong Jia
- Department of Developmental and Behavioral Pediatric, The First Hospital of Jilin University, Changchun, China
| | - Lijie Wu
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin, China
| | - Yan Hao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Ke
- Child Mental Health Research Center of Nanjing Brain Hospital, Nanjing, China
| | - Mingji Yi
- Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qi Hong
- Maternal and Child Health Hospital of Baoan, Shenzhen, China
| | - Jinjin Chen
- Department of Child Healthcare, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shuanfeng Fang
- Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yichao Wang
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Qi Wang
- Deyang Maternity & Child Healthcare Hospital, Deyang, China
| | - Chunhua Jin
- Department of Children Health Care, Capital Institute of Pediatrics, Beijing, China
| | - Ximing Xu
- Big Data Center for Children's Medical Care, Children's Hospital of Chongqing Medical University, No. 136. Zhongshan Er Rd, Yuzhong District, Chongqing 400014, China.
| | - Tingyu Li
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China.
| |
Collapse
|
38
|
Smirnova L, Modafferi S, Schlett C, Osborne LM, Payne JL, Sabunciyan S. Blood extracellular vesicles carrying brain-specific mRNAs are potential biomarkers for detecting gene expression changes in the female brain. Mol Psychiatry 2024; 29:962-973. [PMID: 38212371 DOI: 10.1038/s41380-023-02384-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 12/07/2023] [Accepted: 12/13/2023] [Indexed: 01/13/2024]
Abstract
The absence of non-invasive tests that can monitor the status of the brain is a major obstacle for psychiatric care. In order to address this need, we assessed the feasibility of using tissue-specific gene expression to determine the origin of extracellular vesicle (EV) mRNAs in peripheral blood. Using the placenta as a model, we discovered that 26 messenger RNAs that are specifically expressed in the placenta are present in EVs circulating in maternal blood. Twenty-three of these transcripts were either exclusively or highly expressed in maternal blood during pregnancy only and not in the postpartum period, verifying the feasibility of using tissue-specific gene expression to infer the tissue of origin for EV mRNAs. Using the same bioinformatic approach, which provides better specificity than isolating L1 cell-adhesion molecule containing EVs, we discovered that 181 mRNAs that are specifically expressed in the female brain are also present in EVs circulating in maternal blood. Gene set enrichment analysis revealed that these transcripts, which are involved in synaptic functions and myelination, are enriched for genes implicated in mood disorders, schizophrenia, and substance use disorders. The EV mRNA levels of 13 of these female brain-specific transcripts are associated with postpartum depression (adjusted p-vals = 3 × 10-5 to 0.08), raising the possibility that they can be used to infer the state of the brain. In order to determine the extent to which EV mRNAs reflect transcription in the brain, we compared mRNAs isolated from cells and EVs in an iPSC-derived brain microphysiological system differentiated for 3 and 9 weeks. We discovered that, although cellular and extracellular mRNA levels are not identical, they do correlate, and it is possible to extrapolate cellular RNA expression changes in the brain via EV mRNA levels. Our findings bring EV mRNAs to the forefront of peripheral biomarker development efforts in psychiatric diseases by demonstrating the feasibility of inferring transcriptional changes in the brain via blood EV mRNA levels.
Collapse
Affiliation(s)
- Lena Smirnova
- Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Sergio Modafferi
- Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Charlotte Schlett
- Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Lauren M Osborne
- Departments of Obstetrics & Gynecology and of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Jennifer L Payne
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, USA
| | - Sarven Sabunciyan
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
39
|
Cai Y, Deng W, Yang Q, Pan G, Liang Z, Yang X, Li S, Xiao X. High-fat diet-induced obesity causes intestinal Th17/Treg imbalance that impairs the intestinal barrier and aggravates anxiety-like behavior in mice. Int Immunopharmacol 2024; 130:111783. [PMID: 38514921 DOI: 10.1016/j.intimp.2024.111783] [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: 11/08/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
The prevalence of autism spectrum disorders (ASD) has been steadily increasing, and growing evidence suggests a link between high-fat diet (HFD), obesity, and ASD; however, the mechanism underlying this association remains elusive. Herein, BTBR T + tf/J (BTBR) inbred mice (a mouse ASD model) and C57Bl/6J (C57) mice were fed an HFD and normal diet (ND) for 8 weeks (groups: C57 + ND, C57 + HFD, BTBR + ND, and BTBR + HFD). Subsequently, mice underwent behavioral assessments, followed by intestinal tissues harvesting to detect expression of intestinal barrier proteins and inflammatory factors and immune cell numbers, and a correlation analysis. HFD-fed BTBR mice developed obesity, elevated blood sugar, significantly aggravated anxiety-like behaviors, impaired intestinal barrier function, intestinal inflammation with elevated CD4+IL17+ T (Th17) cells and reduced CD4+Foxp3+ T (Treg) cells, exhibiting reduced expression of proteins related to AMPK regulatory pathway (AMPK, p-AMPK, SIRT1). Correlation analysis revealed that the degree of behavioral anxiety, the degree of intestinal barrier damage, the severity of intestinal inflammation, and the degree of immune cell imbalance positively correlated with each other. Accordingly, HFD-induced obesity may cause intestinal Th17/Treg imbalance via the AMPK-SIRT1 pathway, leading to an inflammatory environment in the intestine, impairing intestinal barrier function, and ultimately aggravating anxiety-like behaviors in mice.
Collapse
Affiliation(s)
- Yao Cai
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Wenlin Deng
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Qiuping Yang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Guixian Pan
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Zao Liang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Ximei Yang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Sitao Li
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China.
| | - Xin Xiao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China.
| |
Collapse
|
40
|
Zhou T, Shen Y, Lyu J, Yang L, Wang HJ, Hong S, Ji Y. Medication Usage Record-Based Predictive Modeling of Neurodevelopmental Abnormality in Infants under One Year: A Prospective Birth Cohort Study. Healthcare (Basel) 2024; 12:713. [PMID: 38610136 PMCID: PMC11011488 DOI: 10.3390/healthcare12070713] [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: 01/18/2024] [Revised: 03/16/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Early identification of children with neurodevelopmental abnormality is a major challenge, which is crucial for improving symptoms and preventing further decline in children with neurodevelopmental abnormality. This study focuses on developing a predictive model with maternal sociodemographic, behavioral, and medication-usage information during pregnancy to identify infants with abnormal neurodevelopment before the age of one. In addition, an interpretable machine-learning approach was utilized to assess the importance of the variables in the model. In this study, artificial neural network models were developed for the neurodevelopment of five areas of infants during the first year of life and achieved good predictive efficacy in the areas of fine motor and problem solving, with median AUC = 0.670 (IQR: 0.594, 0.764) and median AUC = 0.643 (IQR: 0.550, 0.731), respectively. The final model for neurodevelopmental abnormalities in any energy region of one-year-old children also achieved good prediction performance. The sensitivity is 0.700 (IQR: 0.597, 0.797), the AUC is 0.821 (IQR: 0.716, 0.833), the accuracy is 0.721 (IQR: 0.696, 0.739), and the specificity is 0.742 (IQR: 0.680, 0.748). In addition, interpretable machine-learning methods suggest that maternal exposure to drugs such as acetaminophen, ferrous succinate, and midazolam during pregnancy affects the development of specific areas of the offspring during the first year of life. This study established predictive models of neurodevelopmental abnormality in infants under one year and underscored the prediction value of medication exposure during pregnancy for the neurodevelopmental outcomes of the offspring.
Collapse
Affiliation(s)
- Tianyi Zhou
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China; (T.Z.); (Y.S.); (J.L.); (H.-J.W.)
- Peking University Health Science Center-Weifang Joint Research Center for Maternal and Child Health, Beijing 100191, China
| | - Yaojia Shen
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China; (T.Z.); (Y.S.); (J.L.); (H.-J.W.)
- Peking University Health Science Center-Weifang Joint Research Center for Maternal and Child Health, Beijing 100191, China
| | - Jinlang Lyu
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China; (T.Z.); (Y.S.); (J.L.); (H.-J.W.)
- Peking University Health Science Center-Weifang Joint Research Center for Maternal and Child Health, Beijing 100191, China
| | - Li Yang
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China;
| | - Hai-Jun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China; (T.Z.); (Y.S.); (J.L.); (H.-J.W.)
- Peking University Health Science Center-Weifang Joint Research Center for Maternal and Child Health, Beijing 100191, China
| | - Shenda Hong
- National Institute of Health Data Science, Peking University, Beijing 100191, China;
| | - Yuelong Ji
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China; (T.Z.); (Y.S.); (J.L.); (H.-J.W.)
- Peking University Health Science Center-Weifang Joint Research Center for Maternal and Child Health, Beijing 100191, China
| |
Collapse
|
41
|
Gailer J. Toward a Mechanism-Driven Integrated Framework to Link Human Exposure to Multiple Toxic Metal(loid) Species with Environmental Diseases. Int J Mol Sci 2024; 25:3393. [PMID: 38542366 PMCID: PMC10969815 DOI: 10.3390/ijms25063393] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 11/11/2024] Open
Abstract
The ongoing anthropogenic pollution of the biosphere with As, Cd, Hg and Pb will inevitably result in an increased influx of their corresponding toxic metal(loid) species into the bloodstream of human populations, including children and pregnant women. To delineate whether the measurable concentrations of these inorganic pollutants in the bloodstream are tolerable or implicated in the onset of environmental diseases urgently requires new insight into their dynamic bioinorganic chemistry in the bloodstream-organ system. Owing to the human exposure to multiple toxic metal(loid) species, the mechanism of chronic toxicity of each of these needs to be integrated into a framework to better define the underlying exposure-disease relationship. Accordingly, this review highlights some recent advances into the bioinorganic chemistry of the Cd2+, Hg2+ and CH3Hg+ in blood plasma, red blood cells and target organs and provides a first glimpse of their emerging mechanisms of chronic toxicity. Although many important knowledge gaps remain, it is essential to design experiments with the intent of refining these mechanisms to eventually establish a framework that may allow us to causally link the cumulative exposure of human populations to multiple toxic metal(loid) species with environmental diseases of unknown etiology that do not appear to have a genetic origin. Thus, researchers from a variety of scientific disciplines need to contribute to this interdisciplinary effort to rationally address this public health threat which may require the implementation of stronger regulatory requirements to improve planetary and human health, which are fundamentally intertwined.
Collapse
Affiliation(s)
- Jürgen Gailer
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
42
|
Zhang P, Wang X, Xu Y, Zhao X, Zhang X, Zhao Z, Wang H, Xiong Z. Association between interpregnancy interval and risk of autism spectrum disorder: a systematic review and Bayesian network meta-analysis. Eur J Pediatr 2024; 183:1209-1221. [PMID: 38085281 DOI: 10.1007/s00431-023-05364-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 03/20/2024]
Abstract
Although the risk of autism spectrum disorder (ASD) has been reported to be associated with interpregnancy intervals (IPIs), their association remains debatable due to inconsistent findings in existing studies. Therefore, the present study aimed to explore their association. PubMed, Embase, Web of Science, and the Cochrane Library were systematically retrieved up to May 25, 2022. An updated search was performed on May 25, 2023, to encompass recent studies. The quality of the included studies was assessed using the Newcastle-Ottawa Scale (NOS). Our primary outcome measures were expressed as adjusted odds ratios (ORs). Given various control measures for IPI and diverse IPI thresholds in the included studies, a Bayesian network meta-analysis was performed. Eight studies were included, involving 24,865 children with ASD and 2,890,289 children without ASD. Compared to an IPI of 24 to 35 months, various IPIs were significantly associated with a higher risk of ASD (IPIs < 6 months: OR = 1.63, 95% CI 1.53-1.74, n = 5; IPIs of 6-11 months: OR = 1.50, 95% CI 1.42-1.59, n = 4; IPIs of 12-23 months: OR = 1.19, 95% CI 1.12-1.23, n = 10; IPIs of 36-59 months: OR = 0.96, 95% CI 0.94-0.99, n = 2; IPIs of 60-119 months: OR = 1.15, 95% CI 1.10-1.20, n = 4; IPIs > 120 months: OR = 1.57, 95% CI 1.43-1.72, n = 4). After adjusting confounding variables, our analysis delineated a U-shaped restricted cubic spline curve, underscoring that both substantially short (< 24 months) and excessively long IPIs (> 72 months) are significantly correlated with an increased risk of ASD. Conclusion: Our analysis indicates that both shorter and longer IPIs might predispose children to a higher risk of ASD. Optimal childbearing health and neurodevelopmental outcomes appear to be associated with a moderate IPI, specifically between 36 and 60 months. What is Known: • An association between autism spectrum disorder (ASD) and interpregnancy intervals (IPIs) has been speculated in some reports. • This association remains debatable due to inconsistent findings in available studies. What is New: • Our study delineated a U-shaped restricted cubic spline curve, suggesting that both shorter and longer IPIs predispose children to a higher risk of ASD. • Optimal childbearing health and neurodevelopmental outcomes appear to be associated with a moderate IPI, specifically between 36 and 60 months.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Xiaoyan Wang
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Yufen Xu
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Xiaoming Zhao
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Xuan Zhang
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Zhiwei Zhao
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Hong Wang
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China.
| | - Zhonggui Xiong
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China.
| |
Collapse
|
43
|
Raghavan R, Wang X. Early Life Origins of Neurodevelopmental Disabilities in the Boston Birth Cohort: Research findings and future directions. PRECISION NUTRITION 2024; 3:e00062. [PMID: 39184946 PMCID: PMC11343508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Neurodevelopmental disabilities (NDD) are a group of conditions with onset in early development period and is characterized by limitations in several functional domains. Autism spectrum disorder (ASD) and Attention-Deficit Hyperactivity Disorders (ADHD), the most common NDDs, have complex etiologies and possibly multiple pathways leading up to the manifestation of these disorders. Boston Birth Cohort (BBC) is a preterm enriched birth cohort, and over the years, researchers have used the BBC dataset to study a broad spectrum of early life protective and risk factors in the context of NDDs. Broadly, some of them include: 1) nutrition (e.g. maternal folate, vitamin B12, cord folate species, selenium), 2) metabolic factors (e.g. role of maternal diabetes, obesity, branched chain amino acids and other essential amino acids), 3) lipid metabolism (e.g. maternal cholesterol), 4) immune activation and/or systematic inflammation (including maternal immune activation, inflammation of the placenta, inflammatory markers, maternal antibiotic use and acetaminophen use), and 5) other factors associated with NDDs (e.g. maternal stress, sickle cell disease). The findings from these studies are discussed in this review. BBC studies have advanced the field of NDD in the following important ways: 1) generating evidence that sheds light on new exposures, 2) furthering the existing knowledge using better methodological approaches, 3) analyzing novel mechanistic pathways on already proven relationship, and 4) advancing knowledge on the under-studied minority population in the U.S. BBC researchers are involved in ongoing efforts to characterize NDD developmental trajectories across the life stages by integrating multi-omics data (genome, epigenome, and metabolome) to gain a deeper understanding of the molecular pathways by which early life factors drive or shape the developmental trajectories of NDDs.
Collapse
Affiliation(s)
- Ramkripa Raghavan
- Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Xiaobin Wang
- Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
44
|
Aran A, Cayam Rand D. Cannabinoid treatment for the symptoms of autism spectrum disorder. Expert Opin Emerg Drugs 2024; 29:65-79. [PMID: 38226593 DOI: 10.1080/14728214.2024.2306290] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is a neurodevelopmental disorder affecting approximately 3% of school-age children. The core symptoms are deficits in social communication and restricted and repetitive patterns of behavior. Associated problems in cognition, language, behavior, sleep and mood are prevalent. Currently, no established pharmacological treatment exists for core ASD symptoms. Risperidone and aripiprazole are used to manage associated irritability, but their effectiveness is limited and adverse events are common. AREAS COVERED This mini-review summarizes existing scientific literature and ongoing clinical trials concerning cannabinoid treatment for ASD. Uncontrolled case series have documented improvements in both core ASD symptoms and related behavioral challenges in children treated with cannabis extracts rich in cannabidiol (CBD). Placebo-controlled studies involving CBD-rich cannabis extracts and/or pure CBD in children with ASD have demonstrated mixed efficacy results. A similar outcome was observed in a placebo-controlled study of pure CBD addressing social avoidance in Fragile X syndrome. Importantly, these studies have shown relatively high safety and tolerability. EXPERT OPINION While current clinical data suggest the potential of CBD and CBD-rich cannabis extract in managing core and behavioral deficits in ASD, it is prudent to await the results of ongoing placebo-controlled trials before considering CBD treatment for ASD.
Collapse
Affiliation(s)
- Adi Aran
- Neuropediatric Unit, Shaare Zedek Medical Center, Jerusalem, Israel
- Israel School of Medicine, Hebrew university of Jerusalem, Jerusalem, Israel
| | - Dalit Cayam Rand
- Neuropediatric Unit, Shaare Zedek Medical Center, Jerusalem, Israel
- Israel School of Medicine, Hebrew university of Jerusalem, Jerusalem, Israel
| |
Collapse
|
45
|
Ou XX, Wang X, Zhan XL, Shen SL, Karatela S, Jing J, Cai L, Liu RQ, Lin LZ, Dong GH. The associations of secondhand smoke exposure with neurodevelopmental disorders and critical time window identification: A systematic review and meta-analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 913:169649. [PMID: 38159763 DOI: 10.1016/j.scitotenv.2023.169649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/11/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Secondhand smoke (SHS) exposure was harmful for brain development. However, the association between SHS exposure and NDDs diagnosis were unclear. OBJECTIVES To evaluate associations between SHS exposure and NDDs diagnosis, identify critical time windows, and summarize the strength of evidence. METHODS To investigate the associations of SHS exposure and the development of NDDs, we searched Ovid, EMBASE, Web of Science, Cochrane Library, and PubMed for all the relevant studies up to 31 March 2023. The risk estimates and standardized mean differences (SMD) for the individuals with any NDDs who were exposed to SHS exposure compared with those unexposed or low-exposed. RESULTS The results showed that a total of 31,098 citations were identified, of which 54 studies were included. We identified significant associations between SHS exposure and the risks of NDDs including specific types of NDDs like attention deficit hyperactivity disorder (ADHD) and learning disabilities (LD) despite the observed heterogeneity for NDDs and ADHD. We also observed a significant association between cotinine exposure and ADHD. However, inconsistent ratings between the two quality-of-evidence methods for all the meta-analyses indicated the current evidence of the associations and the potential exposure window remained inconclusive. DISCUSSION Our findings suggested that SHS exposure was associated with a higher risk of developing ADHD and LD, with inconclusive quality-of-evidence. In addition, period-specific associations remained unclear based on current evidence.
Collapse
Affiliation(s)
- Xiao-Xuan Ou
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xin Wang
- Key Laboratory of Brain, Cognition and Education Science, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou, China
| | - Xiao-Ling Zhan
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Shu-Li Shen
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Shamshad Karatela
- Faculty of Health and Behavioural Sciences, Pharmacy Australia Centre of Excellence, University of Queensland, Woolloongabba, QLD, Australia; Institute of Tropical Health and Medicine (AITHM), James Cook University, Townsville, QLD, Australia
| | - Jin Jing
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Li Cai
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ru-Qing Liu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, 510080 Guangzhou, China
| | - Li-Zi Lin
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, 510080 Guangzhou, China.
| | - Guang-Hui Dong
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, 510080 Guangzhou, China.
| |
Collapse
|
46
|
Takada R, Toritsuka M, Yamauchi T, Ishida R, Kayashima Y, Nishi Y, Ishikawa M, Yamamuro K, Ikehara M, Komori T, Noriyama Y, Kamikawa K, Saito Y, Okano H, Makinodan M. Granulocyte macrophage colony-stimulating factor-induced macrophages of individuals with autism spectrum disorder adversely affect neuronal dendrites through the secretion of pro-inflammatory cytokines. Mol Autism 2024; 15:10. [PMID: 38383466 PMCID: PMC10882766 DOI: 10.1186/s13229-024-00589-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND A growing body of evidence suggests that immune dysfunction and inflammation in the peripheral tissues as well as the central nervous system are associated with the neurodevelopmental deficits observed in autism spectrum disorder (ASD). Elevated expression of pro-inflammatory cytokines in the plasma, serum, and peripheral blood mononuclear cells of ASD has been reported. These cytokine expression levels are associated with the severity of behavioral impairments and symptoms in ASD. In a prior study, our group reported that tumor necrosis factor-α (TNF-α) expression in granulocyte-macrophage colony-stimulating factor-induced macrophages (GM-CSF MΦ) and the TNF-α expression ratio in GM-CSF MΦ/M-CSF MΦ (macrophage colony-stimulating factor-induced macrophages) was markedly higher in individuals with ASD than in typically developed (TD) individuals. However, the mechanisms of how the macrophages and the highly expressed cytokines affect neurons remain to be addressed. METHODS To elucidate the effect of macrophages on human neurons, we used a co-culture system of control human-induced pluripotent stem cell-derived neurons and differentiated macrophages obtained from the peripheral blood mononuclear cells of five TD individuals and five individuals with ASD. All participants were male and ethnically Japanese. RESULTS Our results of co-culture experiments showed that GM-CSF MΦ affect the dendritic outgrowth of neurons through the secretion of pro-inflammatory cytokines, interleukin-1α and TNF-α. Macrophages derived from individuals with ASD exerted more severe effects than those derived from TD individuals. LIMITATIONS The main limitations of our study were the small sample size with a gender bias toward males, the use of artificially polarized macrophages, and the inability to directly observe the interaction between neurons and macrophages from the same individuals. CONCLUSIONS Our co-culture system revealed the non-cell autonomous adverse effects of GM-CSF MΦ in individuals with ASD on neurons, mediated by interleukin-1α and TNF-α. These results may support the immune dysfunction hypothesis of ASD, providing new insights into its pathology.
Collapse
Affiliation(s)
- Ryohei Takada
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan.
| | - Takahira Yamauchi
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Rio Ishida
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Yoshinori Kayashima
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Yuki Nishi
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Minobu Ikehara
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Takashi Komori
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Yuki Noriyama
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Kohei Kamikawa
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Yasuhiko Saito
- Department of Neurophysiology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara City, Nara, 634-8522, Japan
- Osaka Psychiatric Research Center, 3-16-21 Miyanosaka, Hirakata City, Osaka, 573-0022, Japan
| |
Collapse
|
47
|
Ou J, Dong H, Dai S, Hou Y, Wang Y, Lu X, Xun G, Xia K, Zhao J, Shen Y. Development and validation of a risk score model for predicting autism based on pre- and perinatal factors. Front Psychiatry 2024; 15:1291356. [PMID: 38435974 PMCID: PMC10904522 DOI: 10.3389/fpsyt.2024.1291356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024] Open
Abstract
Background The use of pre- and perinatal risk factors as predictive factors may lower the age limit for reliable autism prediction. The objective of this study was to develop a clinical model based on these risk factors to predict autism. Methods A stepwise logistic regression analysis was conducted to explore the relationships between 28 candidate risk factors and autism risk among 615 Han Chinese children with autism and 615 unrelated typically developing children. The significant factors were subsequently used to create a clinical risk score model. A chi-square automatic interaction detector (CHAID) decision tree was used to validate the selected predictors included in the model. The predictive performance of the model was evaluated by an independent cohort. Results Five factors (pregnancy influenza-like illness, pregnancy stressors, maternal allergic/autoimmune disease, cesarean section, and hypoxia) were found to be significantly associated with autism risk. A receiver operating characteristic (ROC) curve indicated that the risk score model had good discrimination ability for autism, with an area under the curve (AUC) of 0.711 (95% CI=0.679-0.744); in the external validation cohort, the model showed slightly worse but overall similar predictive performance. Further subgroup analysis indicated that a higher risk score was associated with more behavioral problems. The risk score also exhibited robustness in a subgroup analysis of patients with mild autism. Conclusion This risk score model could lower the age limit for autism prediction with good discrimination performance, and it has unique advantages in clinical application.
Collapse
Affiliation(s)
- Jianjun Ou
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huixi Dong
- Mental Health Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Si Dai
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yanting Hou
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ying Wang
- Mental Health Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaozi Lu
- Qingdao Mental Health Center, Qingdao, Shandong, China
| | - Guanglei Xun
- Shandong Mental Health Center, Jinan, Shandong, China
| | - Kun Xia
- Center for Medical Genetics and School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jingping Zhao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yidong Shen
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
48
|
Jin T, Pang Q, Huang W, Xing D, He Z, Cao Z, Zhang T. Particulate matter 2.5 causally increased genetic risk of autism spectrum disorder. BMC Psychiatry 2024; 24:129. [PMID: 38365642 PMCID: PMC10870670 DOI: 10.1186/s12888-024-05564-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: 11/16/2023] [Accepted: 01/28/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Growing evidence suggested that particulate matter (PM) exhibit an increased risk of autism spectrum disorder (ASD). However, the causal association between PM and ASD risk remains unclear. METHODS We performed two-sample Mendelian randomization (MR) analyses, using instrumental variables (IVs) sourced from the largest genome-wide association studies (GWAS) databases. We employed three MR methods: inverse-variance weighted (IVW), weighted median (WM), and MR-Egger, with IVW method serving as our primary MR method. Sensitivity analyses were performed to ensure the stability of these findings. RESULTS The MR results suggested that PM2.5 increased the genetic risk of ASD (β = 2.41, OR = 11.13, 95% CI: 2.54-48.76, P < 0.01), and similar result was found for PM2.5 absorbance (β = 1.54, OR = 4.67, 95% CI: 1.21-18.01, P = 0.03). However, no such association was found in PM10 (β = 0.27, OR = 1.30, 95% CI: 0.72-2.36, P = 0.38). After adjusting for the false discovery rate (FDR) correction, our MR results remain consistent. Sensitivity analyses did not find significant heterogeneity or horizontal pleiotropy. CONCLUSIONS Our findings indicate that PM2.5 is a potential risk factor for ASD. Effective strategies to mitigate air pollutants might lead to a reduced incidence of ASD.
Collapse
Affiliation(s)
- Tianyu Jin
- Department of Rehabilitation Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Neurological rehabilitation, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Qiongyi Pang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Neurological rehabilitation, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Wei Huang
- Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China
- Department of Medicine and Health, University of Sydney, Sydney, Australia
| | - Dalin Xing
- Department of Rehabilitation Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Neurological rehabilitation, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Zitian He
- Department of Rehabilitation Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Neurological rehabilitation, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Zheng Cao
- The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Tong Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Department of Neurological rehabilitation, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China.
| |
Collapse
|
49
|
Goutman SA, Boss J, Jang DG, Mukherjee B, Richardson RJ, Batterman S, Feldman EL. Environmental risk scores of persistent organic pollutants associate with higher ALS risk and shorter survival in a new Michigan case/control cohort. J Neurol Neurosurg Psychiatry 2024; 95:241-248. [PMID: 37758454 PMCID: PMC11060633 DOI: 10.1136/jnnp-2023-332121] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/05/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal, progressive neurogenerative disease caused by combined genetic susceptibilities and environmental exposures. Identifying and validating these exposures are of paramount importance to modify disease risk. We previously reported that persistent organic pollutants (POPs) associate with ALS risk and survival and aimed to replicate these findings in a new cohort. METHOD Participants with and without ALS recruited in Michigan provided plasma samples for POPs analysis by isotope dilution with triple quadrupole mass spectrometry. ORs for risk models and hazard ratios for survival models were calculated for individual POPs. POP mixtures were represented by environmental risk scores (ERS), a summation of total exposures, to evaluate the association with risk (ERSrisk) and survival (ERSsurvival). RESULTS Samples from 164 ALS and 105 control participants were analysed. Several individual POPs significantly associated with ALS, including 8 of 22 polychlorinated biphenyls and 7 of 10 organochlorine pesticides (OCPs). ALS risk was most strongly represented by the mixture effects of OCPs alpha-hexachlorocyclohexane, hexachlorobenzene, trans-nonachlor and cis-nonachlor and an interquartile increase in ERSrisk enhanced ALS risk 2.58 times (p<0.001). ALS survival was represented by the combined mixture of all POPs and an interquartile increase in ERSsurvival enhanced ALS mortality rate 1.65 times (p=0.008). CONCLUSIONS These data continue to support POPs as important factors for ALS risk and progression and replicate findings in a new cohort. The assessments of POPs in non-Michigan ALS cohorts are encouraged to better understand the global effect and the need for targeted disease risk reduction strategies.
Collapse
Affiliation(s)
- Stephen A Goutman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Jonathan Boss
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Dae-Gyu Jang
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Rudy J Richardson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Stuart Batterman
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
50
|
Li H, Guo W, Li S, Sun B, Li N, Xie D, Dong Z, Luo D, Chen W, Fu W, Zheng J, Zhu J. Alteration of the gut microbiota profile in children with autism spectrum disorder in China. Front Microbiol 2024; 14:1326870. [PMID: 38420215 PMCID: PMC10899803 DOI: 10.3389/fmicb.2023.1326870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/18/2023] [Indexed: 03/02/2024] Open
Abstract
Background Autism spectrum disorder (ASD) is associated with alterations in the gut microbiome. However, there are few studies on gut microbiota of children with ASD in China, and there is a lack of consensus on the changes of bacterial species. Purpose Autism spectrum disorder (ASD) is associated with alterations in the gut microbiome. However, there are few studies on gut microbiota of children with ASD in China, and there is a lack of consensus on the changes of bacterial species. Methods We used 16S rRNA sequencing to analyze ASD children (2 to 12 years), HC (2 to 12 years). Results Our findings showed that the α-diversity, composition, and relative abundance of gut microbiota in the ASD group were significantly different from those in the HC groups. Compared with the HC group, the α-diversity in the ASD group was significantly decreased. At the genus level, the relative abundance of g_Faecalibacterium, g_Blautia, g_Eubacterium_eligens_group, g_Parasutterella, g_Lachnospiraceae_NK4A136_group and g_Veillonella in ASD group was significantly increased than that in HC groups, while the relative abundance of g_Prevotella 9 and g_Agathobacter was significantly decreased than that in HC groups. In addition, KEGG pathway analysis showed that the microbial functional abnormalities in ASD patients were mainly concentrated in metabolic pathways related to fatty acid, amino acid metabolism and aromatic compound metabolism, and were partially involved in neurotransmitter metabolism. Conclusion This study revealed the characteristics of gut microbiota of Chinese children with ASD and provided further evidence of gut microbial dysbiosis in ASD.
Collapse
Affiliation(s)
- Hui Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wei Guo
- Stroke Center, Puyang People's Hospital, Puyang, China
| | - Sijie Li
- Department of Pediatrics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Bishao Sun
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ningshan Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Dongjing Xie
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zongming Dong
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Dan Luo
- Department of Neurology, Yunyang People's Hospital, Yunyang, China
| | - Wei Chen
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Weihua Fu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jingzhen Zhu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|