1
|
Qing L, Qian X, Zhu H, Wang J, Sun J, Jin Z, Tang X, Zhao Y, Wang G, Zhao J, Chen W, Tian P. Maternal-infant probiotic transmission mitigates early-life stress-induced autism in mice. Gut Microbes 2025; 17:2456584. [PMID: 39931863 DOI: 10.1080/19490976.2025.2456584] [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: 11/09/2024] [Revised: 12/14/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025] Open
Abstract
Autism, a disorder influenced by both genetic and environmental factors, presents significant challenges for prevention and treatment. While maternal-infant gut microbiota has been a focus in autism research, preventive strategies targeting maternal gut microbiota remain underexplored. This study demonstrates that prenatal probiotic intake can effectively prevent maternal separation-induced autistic-like behaviors in offspring without altering the embryonic neurodevelopment in mice. Using specific PCR primers and cross-fostering experiments, we traced the vertical transmission of probiotics, primarily via fecal/vaginal contamination. Early probiotic colonization conferred resilience against stress-induced gut pathogenic microbes and Th17-mediated peripheral inflammation while significantly inhibiting hypermyelination and neuroinflammation linked to systemic inflammation. Microbial metabolites like tyrosol and xanthurenic acid alleviated neuroinflammation and hypermyelination in vitro, though the causal relationship among neuroinflammation, hypermyelination, and autism in vivo requires further validation. These findings underscore the importance of the maternal-infant microbiota transmission window in autism prevention and highlight the clinical potential of prenatal probiotic interventions.
Collapse
Affiliation(s)
- Li Qing
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Huiyue Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingyu Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingge Sun
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Zhiying Jin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xinyu Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Yingqi Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
2
|
Yu L, Long Q, Zhang Y, Liu Y, Guo Z, Cao X, Qin F, Xu Y, Qian Q, Gao B, Chen J, Liu J, Zeng Y, Teng Z. Bidirectional Mendelian randomization analysis of plasma lipidome and psychiatric disorders. J Affect Disord 2025; 379:871-883. [PMID: 39442703 DOI: 10.1016/j.jad.2024.10.063] [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: 06/20/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Evidence from observational studies and clinical experiments suggests a close association between plasma lipidome and psychiatric disorders. However, the causal relationship between plasma lipidome and psychiatric disorders remains insufficiently determined. Plasma lipidome are relatively easy to measure and regulate clinically, and they play a crucial role in neuronal signal transduction, making them a focus of interest as potential therapeutic targets for psychiatric disorders. METHODS In this study, we utilized the latest Finnish population-based genome-wide association study (GWAS) data on 179 lipid species. We downloaded data on five psychiatric disorders from the IEU database, including schizophrenia, bipolar disorder, depression, autism from the PGC consortium, and anxiety disorder from the Neale lab. Using two-sample bidirectional Mendelian randomization (MR) analysis, we assessed the relationship between these 179 lipid species and the risk of the five psychiatric disorders. To validate the assumptions of Mendelian randomization, we conducted tests for horizontal pleiotropy and heterogeneity. RESULTS After applying FDR correction to assess the relationship between 179 lipid species traits and the risk of five psychiatric disorders, our analysis provided evidence of a causal relationship specifically between genetic susceptibility in the plasma lipidome and bipolar disorder. This relationship notably involves eight phosphatidylcholines (PCs) and two sterols, with PCs displaying a dual and complex role in the disorder. Reverse Mendelian randomization (MR) analysis did not reveal a significant causal impact of psychiatric disorders on the plasma lipidome. LIMITATIONS Despite using two-sample bidirectional Mendelian randomization analysis, the complex biological pathways and potential confounding factors may still affect the accuracy of the causal relationships. The impact of genetic variations on the lipidome and psychiatric disorders may involve multiple mechanisms, which cannot be fully elucidated in this study. CONCLUSION This study identified a causal relationship between genetic susceptibility in plasma lipidome and bipolar disorder, indicating that plasma lipidome may influence the risk of psychiatric disorders and providing direction for exploring them as potential intervention targets. The findings not only deepen our understanding of the etiology of psychiatric disorders but also provide a critical theoretical foundation for future clinical interventions and prevention strategies, potentially contributing to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Ling Yu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qing Long
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunqiao Zhang
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China; Department of Psychiatry, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Yilin Liu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ziyi Guo
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiang Cao
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fuyi Qin
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yangyang Xu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qingqing Qian
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Biyao Gao
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jian Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jie Liu
- Department of Dermatology and Venereology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Yong Zeng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China; Department of Psychiatry, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| | - Zhaowei Teng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
3
|
Yan J, Han VX, Jones HF, Couttas TA, Jieu B, Leweke FM, Lee J, Loi C, Webster R, Kothur K, Menezes MP, Antony J, Kandula T, Cardamone M, Patel S, Bandodkar S, Dale RC. Cerebrospinal fluid metabolomics in autistic regression reveals dysregulation of sphingolipids and decreased β-hydroxybutyrate. EBioMedicine 2025; 114:105664. [PMID: 40138886 PMCID: PMC11986237 DOI: 10.1016/j.ebiom.2025.105664] [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: 11/20/2024] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Autism is highly heritable, however actionable genetic findings are only found in a minority of patients. Many people with autism suffer loss of neurodevelopmental skills, known as autistic regression. The cause of regression is poorly understood, and the diagnostic and therapeutic pathways are lacking. METHODS We used untargeted metabolomics using a UPLC-Q-Exactive-HFx Mass Spectrometry to examine cerebrospinal fluid (CSF) from twenty-two patients with autistic regression compared to sixteen controls with neurodevelopmental disorders (but not autistic regression) and thirty-four controls with other neurological disease (headache, encephalitis, epilepsy). The twenty-two patients with autistic regression consisted of two groups: early (infantile) autistic regression <2 years of age (n = 8), and later regression of skills >4 years of age, often in the context of pre-existing developmental concerns (n = 14). Metabolites of interest were then quantified and validated using targeted assays. FINDINGS Untargeted case-control studies revealed good separation of patients from controls using multivariate analysis. β-hydroxybutyrate was significantly decreased in the CSF of patients with autistic regression, and the findings were validated using a targeted β-hydroxybutyrate assay. The sphingolipid, sphingosine-1-phosphate was significantly elevated in the discovery case-control studies, and sphingolipid metabolism pathways were also significantly dysregulated. We therefore developed a targeted metabolite assay of forty sphingolipids. After FDR correction, 21 of the 40 sphingolipids were significantly dysregulated (pFDR < 0.05) (Benjamini-Hochberg correction) in autistic regression compared to the neurodevelopmental controls, and 26 of the 40 sphingolipids were significantly dysregulated in autistic regression compared to other neurological controls, with elevated ceramides, hexosylceramides, sphingosines (including sphingosine-1-phosphate), and sulfatides. By contrast, sphingomyelin levels were generally decreased in autistic regression. INTERPRETATION Our data shows the potential utility of CSF metabolomics in the context of autistic regression, a clinical syndrome which has historically lacked pathophysiological biomarkers and disease modifying therapies. FUNDING Financial support for the study was granted by Dale NHMRC Investigator grant APP1193648, Petre Foundation, Cerebral Palsy Alliance, and Ainsworth and SCHF Neuroscience grant scheme.
Collapse
Affiliation(s)
- Jingya Yan
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Velda X Han
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hannah F Jones
- Starship Hospital, Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Timothy A Couttas
- Neuroscience Research Australia, Randwick, NSW, Australia; Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - Beverly Jieu
- Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - F Markus Leweke
- Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - Jennifer Lee
- Department of Endocrinology, The Children's Hospital at Westmead, NSW, Australia
| | - Catherine Loi
- Department of Endocrinology, The Children's Hospital at Westmead, NSW, Australia
| | - Richard Webster
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Kavitha Kothur
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Manoj P Menezes
- Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Jayne Antony
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Tejaswi Kandula
- Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia
| | - Michael Cardamone
- Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia
| | - Shrujna Patel
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Sushil Bandodkar
- Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Department of Biochemistry, The Children's Hospital at Westmead, NSW, Australia
| | - Russell C Dale
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia.
| |
Collapse
|
4
|
Oraki Kohshour M, Papiol S, Tkachev A, Stekolshchikova E, Adorjan K, Budde M, Heilbronner U, Heilbronner M, Kalman JL, Reich-Erkelenz D, K Schaupp S, Senner F, Vogl T, Wiltfang J, Reininghaus EZ, Juckel G, Dannlowski U, Fallgatter AJ, Spitzer C, Schmauß M, von Hagen M, Falkai P, Khaitovich P, Schulze TG, Schulte EC. Investigating the association of the plasma lipidomic profile with cognitive performance and genetic risk in the PsyCourse study. Transl Psychiatry 2025; 15:105. [PMID: 40155381 PMCID: PMC11953450 DOI: 10.1038/s41398-025-03323-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 02/21/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025] Open
Abstract
Although lipid biology may play a key role in the pathophysiology of mental health disorders such as schizophrenia (SCZ) and bipolar disorder (BD), the nature of this interplay and how it could shape phenotypic presentation, including cognitive performance is still incompletely understood. To address this question, we analyzed the association of plasma level of different lipid species with cognitive performance in the transdiagnostic PsyCourse Study. Plasma lipidomic profiles of 623 individuals (188 SCZ, 243 BD, 192 healthy controls) belonging to the PsyCourse Study were assessed using liquid chromatography and untargeted mass spectrometry. The association between 364 annotated lipid species from 16 lipid classes and six cognitive tests was evaluated. Likewise, the association of polygenic risk scores (PRS) for SCZ, BD, executive function (EF), and educational attainment (EA) with lipid plasma levels were also investigated. In the regression analysis, three lipid species belonging to phosphatidylethanolamine plasmalogen and one belonging to ceramide class showed significant negative association with Digit-Symbol test scores. Lipid class-based enrichment analysis in LipidR replicated the significance of the phosphatidylethanolamines class for the Digit-Symbol test, which evaluates the processing speed in cognitive tasks. Polygenic load for SCZ, BD, EF, or EA was not associated with lipid levels. Our findings suggest a link between lipids and cognitive performance independent of mental health disorders. Still, independent replication is warranted to better understand if phosphatidylethanolamines could represent an actionable pharmacologic target to tackle cognitive dysfunction, an important unmet clinical need that affects long-term functional outcomes in individuals with severe mental health disorders.
Collapse
Affiliation(s)
- Mojtaba Oraki Kohshour
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sergi Papiol
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
- Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Anna Tkachev
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow, 121205, Russia
| | - Elena Stekolshchikova
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow, 121205, Russia
| | - Kristina Adorjan
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Monika Budde
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Maria Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Janos L Kalman
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Daniela Reich-Erkelenz
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Sabrina K Schaupp
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Fanny Senner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, 80336, Germany
- Centres for Psychiatry Suedwuerttemberg, Ravensburg, 88214, Germany
| | - Thomas Vogl
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, 37075, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, 37075, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Eva Z Reininghaus
- Division of Psychiatry and Psychotherapeutic Medicine, Research Unit for Bipolar Affective Disorder, Medical University of Graz, Graz, 8036, Austria
| | - Georg Juckel
- Department of Psychiatry, Ruhr University Bochum, LWL University Hospital, Bochum, 44791, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, 48149, Germany
| | - Andreas J Fallgatter
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, 72076, Germany
- German Center for Mental Health (DZPG), partner site Tübingen, Tübingen, 72076, Germany
| | - Carsten Spitzer
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Rostock, Rostock, 18147, Germany
| | - Max Schmauß
- Clinic for Psychiatry, Psychotherapy and Psychosomatics, Augsburg University, Medical Faculty, Bezirkskrankenhaus Augsburg, Augsburg, 86156, Germany
| | - Martin von Hagen
- Clinic for Psychiatry and Psychotherapy, Clinical Center Werra-Meißner, Eschwege, 37269, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, 80336, Germany
- German Center for Mental Health (DZPG), partner site Munich/Augsburg, 80336, Munich, Germany
| | - Philipp Khaitovich
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow, 121205, Russia
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
- German Center for Mental Health (DZPG), partner site Munich/Augsburg, 80336, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Norton College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eva C Schulte
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany.
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, 80336, Germany.
- German Center for Mental Health (DZPG), partner site Munich/Augsburg, 80336, Munich, Germany.
- Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Bonn, University of Bonn, Bonn, 53127, Germany.
- Institute of Human Genetics, Faculty of Medicine and University Hospital Bonn, University of Bonn, Bonn, 53127, Germany.
| |
Collapse
|
5
|
Kim S, Jeon B. Inequities in cardiovascular risk and lifestyle factors among individuals with developmental disabilities: evidence from Korea's national health screening program. Sci Rep 2025; 15:10463. [PMID: 40140661 PMCID: PMC11947128 DOI: 10.1038/s41598-025-94874-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Significant gaps persist in the awareness and understanding of the health challenges faced by individuals with developmental disabilities. This study aims to examine health disparities among individuals with developmental disabilities in South Korea, focusing on cardiovascular risk factors, including obesity, hypertension, diabetes, and dyslipidemia, and lifestyle factors like physical activity, smoking, and drinking behaviors. We analyzed data from the National General Health Screening Program for 2019 and 2020, including a sample of 17,012 persons with disabilities and 5,623,993 persons without disabilities. We utilized propensity score matching (1:1) and multivariable logistic regression estimated outcomes. After matching, each group included 13,863 individuals with balanced baseline characteristics. Individuals with disability had higher risks of overall and abdominal obesity and lower risks of smoking and drinking. No significant differences were found in blood pressure and fasting blood sugar levels post-matching. In addition, individuals with disability showed lower risks of abnormal cholesterol, low-density lipoprotein cholesterol and triglyceride levels, but higher risks for abnormal high-density lipoprotein cholesterol levels and lower physical activity levels. These findings highlight the urgent need for targeted interventions to address obesity and promote physical activity, while also acknowledging the lower risks of smoking and drinking in individuals with developmental disabilities.
Collapse
Affiliation(s)
- Sujin Kim
- Korea Institute for Health and Social Affairs, 370 Sicheong-daero, Sejong, 30147, Republic of Korea
| | - Boyoung Jeon
- Department of Health and Medical Information, Myongji College, 134 Gajwa-ro, Seodaemun-gu, Seoul, 03656, Republic of Korea.
| |
Collapse
|
6
|
Wang Z, Zhu H, Chen L, Gan C, Min W, Xiao J, Zou Z, He Y. Absence of Causal Relationship Between Levels of Unsaturated Fatty Acids and ADHD: Evidence From Mendelian Randomization Study. J Atten Disord 2024; 28:1716-1725. [PMID: 39082434 DOI: 10.1177/10870547241264660] [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] [Indexed: 10/29/2024]
Abstract
OBJECTIVE Previous research suggests a potential link between unsaturated fatty acids (UFAs) and ADHD, but the causal relationship remains uncertain. This study aims to investigate the causal association between ADHD and UFAs using Mendelian randomization (MR) analysis. METHODS Summary data from genome-wide association studies were used to estimate the concentration of circulating UFAs, including Monounsaturated Fatty Acids (MUFAs), Polyunsaturated Fatty Acids (PUFAs), Omega-3 PUFAs, Omega-6 PUFAs, Linoleic Acid (LA), and Docosahexaenoic Acid (DHA). Data from the Psychiatric Genomics Consortium, including both childhood and adult ADHD, were respectively used to examine the relationship between genetically predicted UFAs levels and ADHD. Various MR methods, including Inverse-variance weighted (IVW), MR Pleiotropy RESidual Sum and Outlier, MR-Egger, weighted median, and weighted mode, were employed to assess heterogeneity and pleiotropy. RESULTS The IVW revealed only nominal evidence suggesting a potential causal relationship between genetically predicted PUFAs (OR = 0.92, 95% CI [0.85, 0.99], p = .031), Omega-6 PUFAs (OR = 0.90, 95% CI [0.83, 0.98], p = .020), and LA levels (OR = 0.90, 95% CI [0.82, 0.98], p = .021) with childhood ADHD risk. However, after false discovery rate correction, the p-values for PUFAs, Omega-6 PUFAs, and LA levels all exceeded the threshold for significance. For adult ADHD, we did not find any significant associations between the six circulating UFA levels and adult ADHD. CONCLUSION Our findings do not support a causal relationship between UFAs levels and ADHD. This suggests that UFAs supplements may not be effective in improving ADHD symptoms and importantly, it appears that UFAs levels may not have a long-term effect on ADHD.
Collapse
Affiliation(s)
- Zuxing Wang
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
- Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Hongru Zhu
- West China Hospital of Sichuan University, Chengdu, China
| | - Lili Chen
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
- Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Chenyu Gan
- Jiaxiang Foreign Language Senior High School, Chengdu, China
| | - Wenjiao Min
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
- Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Jun Xiao
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
- Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Zhili Zou
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
- Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Ying He
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
- Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Yu XH, Lu HM, Li J, Su MZ, Li XM, Jin Y. Association between 25(OH) vitamin D and multiple sclerosis: cohort, shared genetics, and Causality. Nutr J 2024; 23:151. [PMID: 39616386 PMCID: PMC11608472 DOI: 10.1186/s12937-024-01059-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Multiple Sclerosis (MS), an autoimmune disorder causing demyelination and neurological damage, has been linked to 25-hydroxyvitamin D (25OHD) levels, suggesting its role in immune response and MS onset. This study used GWAS datasets to investigate genetic associations between 25OHD and MS. METHODS We utilized a large-scale prospective cohort to evaluate serum 25OHD levels and MS risk. Linkage Disequilibrium Score Regression (LDSC) assessed genetic correlations between 25OHD levels and MS. Cross-trait genome-wide pleiotropy analysis revealed shared genetic loci. MAGMA analysis identified pleiotropic genes, enriched tissues, and gene sets. Stratified LDSC estimated tissue-specific and cell-specific heritability enrichment, and multi-trait co-localization analysis identified shared immune cell subsets. Bidirectional Mendelian Randomization (MR) assessed the causal association between 25OHD and MS risk. RESULTS The observational study found a nonlinear relationship between 25OHD levels and MS risk, with the lowest quartile showing significant risk elevation. Our findings revealed shared genetic structure between 25OHD levels and MS, suggesting a common biological pathway involving immune function and CNS integrity. We found 24 independent loci shared between 25OHD levels and MS risk, enriched in brain tissues and involved in pathways like LDL, HDL, and TG metabolism. Four loci (6p24.3, 6p22.2, 12q14.1, and 19p13.2) had strong co-localization evidence, with mapped genes as potential drug targets. Bidirectional MR analysis supported a causal effect of 25OHD levels on MS risk, suggesting 25OHD supplementation could modulate MS risk. CONCLUSION This study reveals the complex relationship between 25OHD levels and MS, indicating that higher levels are not always advantageous and recommending moderation in supplementation. We identified SMARCA4 as a potential therapeutic target and detailed key pathways influencing this interaction.
Collapse
Affiliation(s)
- Xing-Hao Yu
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Hui-Min Lu
- Department of Outpatient and Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jun Li
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China
| | - Ming-Zhu Su
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
- Department of Good Clinical Practice, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China
| | - Xiao-Min Li
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China.
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China.
| | - Yi Jin
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China.
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China.
| |
Collapse
|
8
|
Liu K, Fu H, Chen Y, Li B, Huang H, Liao X. Relationship between residual cholesterol and cognitive performance: a study based on NHANES. Front Nutr 2024; 11:1458970. [PMID: 39323568 PMCID: PMC11423777 DOI: 10.3389/fnut.2024.1458970] [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: 07/03/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024] Open
Abstract
Background and aims Age-related cognitive impairment impacts a significant portion of the elderly population. Remnant cholesterol (RC) has attracted increased attention in relation to cardiovascular disease, diabetes, hypertension, and fatty liver disease. Nevertheless, its role in cognitive function is still enigmatic, prompting our exploration into the potential associations between them. Methods A total of 1,331 participants from the NHANES (2011-2014) database, all aged over 60, were included in this investigation. Cognitive function was assessed using four widely applied tests, including the Consortium to Establish a Registry for Alzheimer's Disease Word Learning (CERAD-WL), CERAD Delayed Recall (CERAD-DR), Animal Fluency Test (AFT), as well as Digit Symbol Substitution test (DSST). Z-score is calculated by scores from the above four tests. The association between RC, total cholesterol (TC) to RC and cognitive performance was assessed by logistic regression analyses. In addition, restricted cubic spline (RCS) regression was performed to assess non-linearity between RC and cognitive function. Subgroup analysis was performed to evaluate the robustness of the results in populations with relevant covariate variables. Results Those with Z-scores below the 25% quartile are defined as having cognitive impairment, totaling 498 individuals. Observationally, higher RC levels and a lower TC/RC were associated with an increased risk of cognitive impairment. After adjusting for confounding factors, the impact of RC levels on cognitive performance quartiles was consistent across various subgroups, except in individuals with trouble sleeping, no/unknown alcohol use, and no hypertension. Americans with high RC levels and trouble sleeping are more likely to develop cognitive impairment, with an odds ratio of 2.33 (95% CI: 1.18-4.59). Conclusion This study suggests that higher RC levels and lower levels of TC/RC are associated with an increased likelihood of cognitive impairment, suggesting that RC can serve as a novel and convenient indicator for predicting the risk of cognitive impairment in the US population.
Collapse
Affiliation(s)
- Kepeng Liu
- Department of Anesthesiology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Haishou Fu
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yong Chen
- Department of Anesthesiology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Binfei Li
- Department of Anesthesiology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Huaqing Huang
- Department of Pain Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xiaozu Liao
- Department of Anesthesiology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| |
Collapse
|
9
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
10
|
Wang T, Beyene HB, Yi C, Cinel M, Mellett NA, Olshansky G, Meikle TG, Wu J, Dakic A, Watts GF, Hung J, Hui J, Beilby J, Blangero J, Kaddurah-Daouk R, Salim A, Moses EK, Shaw JE, Magliano DJ, Huynh K, Giles C, Meikle PJ. A lipidomic based metabolic age score captures cardiometabolic risk independent of chronological age. EBioMedicine 2024; 105:105199. [PMID: 38905750 PMCID: PMC11246009 DOI: 10.1016/j.ebiom.2024.105199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Metabolic ageing biomarkers may capture the age-related shifts in metabolism, offering a precise representation of an individual's overall metabolic health. METHODS Utilising comprehensive lipidomic datasets from two large independent population cohorts in Australia (n = 14,833, including 6630 males, 8203 females), we employed different machine learning models, to predict age, and calculated metabolic age scores (mAge). Furthermore, we defined the difference between mAge and age, termed mAgeΔ, which allow us to identify individuals sharing similar age but differing in their metabolic health status. FINDINGS Upon stratification of the population into quintiles by mAgeΔ, we observed that participants in the top quintile group (Q5) were more likely to have cardiovascular disease (OR = 2.13, 95% CI = 1.62-2.83), had a 2.01-fold increased risk of 12-year incident cardiovascular events (HR = 2.01, 95% CI = 1.45-2.57), and a 1.56-fold increased risk of 17-year all-cause mortality (HR = 1.56, 95% CI = 1.34-1.79), relative to the individuals in the bottom quintile group (Q1). Survival analysis further revealed that men in the Q5 group faced the challenge of reaching a median survival rate due to cardiovascular events more than six years earlier and reaching a median survival rate due to all-cause mortality more than four years earlier than men in the Q1 group. INTERPRETATION Our findings demonstrate that the mAge score captures age-related metabolic changes, predicts health outcomes, and has the potential to identify individuals at increased risk of metabolic diseases. FUNDING The specific funding of this article is provided in the acknowledgements section.
Collapse
Affiliation(s)
- Tingting Wang
- Baker Heart and Diabetes Institute, Melbourne, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Australia
| | - Habtamu B Beyene
- Baker Heart and Diabetes Institute, Melbourne, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | - Changyu Yi
- Baker Heart and Diabetes Institute, Melbourne, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | - Michelle Cinel
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | | | | | - Thomas G Meikle
- Baker Heart and Diabetes Institute, Melbourne, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | - Jingqin Wu
- Baker Heart and Diabetes Institute, Melbourne, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | | | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | - Joseph Hung
- School of Medicine, University of Western Australia, Perth, Australia
| | - Jennie Hui
- PathWest Laboratory Medicine of Western Australia, Nedlands, Western Australia, Australia; School of Population and Global Health, University of Western Australia, Crawley, Western Australia, Australia; School of Biomedical Sciences, University of Western Australia, Australia
| | - John Beilby
- PathWest Laboratory Medicine of Western Australia, Nedlands, Western Australia, Australia; School of Biomedical Sciences, University of Western Australia, Australia
| | - John Blangero
- South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioural Sciences, Duke University, Durham, NC, USA; Duke Institute of Brain Sciences, Duke University, Durham, NC, USA; Department of Medicine, Duke University, Durham, NC, USA
| | - Agus Salim
- Baker Heart and Diabetes Institute, Melbourne, Australia; Melbourne School of Population and Global Health School of Mathematics and Statistics, The University of Melbourne, Australia
| | - Eric K Moses
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | | | | | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia.
| |
Collapse
|
11
|
Al-Beltagi M, Saeed NK, Bediwy AS, Elbeltagi R. Metabolomic changes in children with autism. World J Clin Pediatr 2024; 13:92737. [PMID: 38947988 PMCID: PMC11212761 DOI: 10.5409/wjcp.v13.i2.92737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficits in social communication and repetitive behaviors. Metabolomic profiling has emerged as a valuable tool for understanding the underlying metabolic dysregulations associated with ASD. AIM To comprehensively explore metabolomic changes in children with ASD, integrating findings from various research articles, reviews, systematic reviews, meta-analyses, case reports, editorials, and a book chapter. METHODS A systematic search was conducted in electronic databases, including PubMed, PubMed Central, Cochrane Library, Embase, Web of Science, CINAHL, Scopus, LISA, and NLM catalog up until January 2024. Inclusion criteria encompassed research articles (83), review articles (145), meta-analyses (6), systematic reviews (6), case reports (2), editorials (2), and a book chapter (1) related to metabolomic changes in children with ASD. Exclusion criteria were applied to ensure the relevance and quality of included studies. RESULTS The systematic review identified specific metabolites and metabolic pathways showing consistent differences in children with ASD compared to typically developing individuals. These metabolic biomarkers may serve as objective measures to support clinical assessments, improve diagnostic accuracy, and inform personalized treatment approaches. Metabolomic profiling also offers insights into the metabolic alterations associated with comorbid conditions commonly observed in individuals with ASD. CONCLUSION Integration of metabolomic changes in children with ASD holds promise for enhancing diagnostic accuracy, guiding personalized treatment approaches, monitoring treatment response, and improving outcomes. Further research is needed to validate findings, establish standardized protocols, and overcome technical challenges in metabolomic analysis. By advancing our understanding of metabolic dysregulations in ASD, clinicians can improve the lives of affected individuals and their families.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
- Department of Pediatric, University Medical Center, Dr. Sulaiman Al Habib Medical Group, Manama, Bahrain, Manama 26671, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 12, Bahrain
- Medical Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Bahrain, Busaiteen 15503, Muharraq, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonology, Faculty of Medicine, Tanta University, Tanta 31527, Alghrabia, Egypt
- Department of Chest Disease, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
- Department of Chest Disease, University Medical Center, Dr. Sulaiman Al Habib Medical Group, Manama, Manama 26671, Bahrain
| | - Reem Elbeltagi
- Department of Medicine, The Royal College of Surgeons in Ireland - Bahrain, Busiateen 15503, Muharraq, Bahrain
| |
Collapse
|
12
|
Deng K, Pan X, Voehler MW, Cai Q, Cai H, Shu X, Gupta DK, Lipworth L, Zheng W, Yu D. Blood Lipids, Lipoproteins, and Apolipoproteins With Risk of Coronary Heart Disease: A Prospective Study Among Racially Diverse Populations. J Am Heart Assoc 2024; 13:e034364. [PMID: 38726919 PMCID: PMC11179824 DOI: 10.1161/jaha.124.034364] [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: 01/08/2024] [Accepted: 04/16/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Comprehensive blood lipoprotein profiles and their association with incident coronary heart disease (CHD) among racially and geographically diverse populations remain understudied. METHODS AND RESULTS We conducted nested case-control studies of CHD among 3438 individuals (1719 pairs), including 1084 White Americans (542 pairs), 1244 Black Americans (622 pairs), and 1110 Chinese adults (555 pairs). We examined 36 plasma lipids, lipoproteins, and apolipoproteins, measured by nuclear magnetic resonance spectroscopy, with incident CHD among all participants and subgroups by demographics, lifestyle, and metabolic health status using conditional or unconditional logistic regression adjusted for potential confounders. Conventionally measured blood lipids, that is, total cholesterol, triglycerides, low-density lipoprotein-cholesterol, and high-density lipoprotein-cholesterol, were each associated with incident CHD, with odds ratios (ORs) being 1.33, 1.32, 1.24, and 0.79 per 1-SD increase among all participants. Seventeen lipoprotein biomarkers showed numerically stronger associations than conventional lipids, with ORs per 1-SD among all participants ranging from 1.35 to 1.57 and a negative OR of 0.78 (all false discovery rate <0.05), including apolipoprotein B100 to apolipoprotein A1 ratio (OR, 1.57 [95% CI, 1.45-1.7]), low-density lipoprotein-triglycerides (OR, 1.55 [95% CI, 1.43-1.69]), and apolipoprotein B (OR, 1.49 [95% CI, 1.37-1.62]). All these associations were significant and consistent across racial groups and other subgroups defined by age, sex, smoking, obesity, and metabolic health status, including individuals with normal levels of conventionally measured lipids. CONCLUSIONS Our study highlighted several lipoprotein biomarkers, including apolipoprotein B/ apolipoprotein A1 ratio, apolipoprotein B, and low-density lipoprotein-triglycerides, strongly and consistently associated with incident CHD. Our results suggest that comprehensive lipoprotein measures may complement the standard lipid panel to inform CHD risk among diverse populations.
Collapse
Affiliation(s)
- Kui Deng
- Vanderbilt Epidemiology Center and Division of EpidemiologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Xiong‐Fei Pan
- Vanderbilt Epidemiology Center and Division of EpidemiologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTNUSA
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & National Medical Products Administration Key Laboratory for Technical Research on Drug Products In Vitro and In Vivo Correlation, West China Second University HospitalSichuan UniversityChengduSichuanChina
| | - Markus W. Voehler
- Department of Chemistry and Center for Structural BiologyVanderbilt UniversityNashvilleTNUSA
| | - Qiuyin Cai
- Vanderbilt Epidemiology Center and Division of EpidemiologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Hui Cai
- Vanderbilt Epidemiology Center and Division of EpidemiologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Xiao‐Ou Shu
- Vanderbilt Epidemiology Center and Division of EpidemiologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Deepak K. Gupta
- Vanderbilt Translational and Clinical Cardiovascular Research Center and Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Loren Lipworth
- Vanderbilt Epidemiology Center and Division of EpidemiologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Wei Zheng
- Vanderbilt Epidemiology Center and Division of EpidemiologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Danxia Yu
- Vanderbilt Epidemiology Center and Division of EpidemiologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| |
Collapse
|
13
|
Mao Y, Lin X, Wu Y, Lu J, Shen J, Zhong S, Jin X, Ma J. Additive interaction between birth asphyxia and febrile seizures on autism spectrum disorder: a population-based study. Mol Autism 2024; 15:17. [PMID: 38600595 PMCID: PMC11007945 DOI: 10.1186/s13229-024-00596-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/21/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a pervasive neurodevelopmental disorder that can significantly impact an individual's ability to socially integrate and adapt. It's crucial to identify key factors associated with ASD. Recent studies link both birth asphyxia (BA) and febrile seizures (FS) separately to higher ASD prevalence. However, investigations into the interplay of BA and FS and its relationship with ASD are yet to be conducted. The present study mainly focuses on exploring the interactive effect between BA and FS in the context of ASD. METHODS Utilizing a multi-stage stratified cluster sampling, we initially recruited 84,934 Shanghai children aged 3-12 years old from June 2014 to June 2015, ultimately including 74,251 post-exclusion criteria. A logistic regression model was conducted to estimate the interaction effect after controlling for pertinent covariates. The attributable proportion (AP), the relative excess risk due to interaction (RERI), the synergy index (SI), and multiplicative-scale interaction were computed to determine the interaction effect. RESULTS Among a total of 74,251 children, 192 (0.26%) were diagnosed with ASD. The adjusted odds ratio for ASD in children with BA alone was 3.82 (95% confidence interval [CI] 2.42-6.02), for FS alone 3.06 (95%CI 1.48-6.31), and for comorbid BA and FS 21.18 (95%CI 9.10-49.30), versus children without BA or FS. The additive interaction between BA and FS showed statistical significance (P < 0.001), whereas the multiplicative interaction was statistically insignificant (P > 0.05). LIMITATIONS This study can only demonstrate the relationship between the interaction of BA and FS with ASD but cannot prove causation. Animal brain experimentation is necessary to unravel its neural mechanisms. A larger sample size, ongoing monitoring, and detailed FS classification are needed for confirming BA-FS interaction in ASD. CONCLUSION In this extensive cross-sectional study, both BA and FS were significantly linked to ASD. The coexistence of these factors was associated with an additive increase in ASD prevalence, surpassing the cumulative risk of each individual factor.
Collapse
Affiliation(s)
- Yi Mao
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xindi Lin
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yuhan Wu
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jiayi Lu
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jiayao Shen
- Department of Nephrology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shaogen Zhong
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xingming Jin
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jun Ma
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
14
|
Chen Y, Fan C, Liu J. Investigating the shared genetic architecture between COVID-19 and obesity: a large-scale genome wide cross-trait analysis. Front Endocrinol (Lausanne) 2024; 15:1325939. [PMID: 38352709 PMCID: PMC10862482 DOI: 10.3389/fendo.2024.1325939] [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: 10/22/2023] [Accepted: 01/11/2024] [Indexed: 02/16/2024] Open
Abstract
Observational studies have reported high comorbidity between obesity and severe COVID-19. The aim of this study is to explore whether genetic factors are involved in the co-occurrence of the two traits. Based on the available genome-wide association studies (GWAS) summary statistics, we explored the genetic correlation and performed cross-trait meta-analysis (CPASSOC) and colocalization analysis (COLOC) to detect pleiotropic single nucleotide polymorphisms (SNPs). At the genetic level, we obtained genes detected by Functional mapping and annotation (FUMA) and the Multi-marker Analysis of GenoMic Annotation (MAGMA). Potential functional genes were further investigated by summary-data-based Mendelian randomization (SMR). Finally, the casualty was identiied using the latent causal variable model (LCV). A significant positive genetic correlation was revealed between obesity and COVID-19. We found 331 shared genetic SNPs by CPASSOC and 13 shared risk loci by COLOC. At the genetic level, We obtained 3546 pleiotropic genes, among which 107 genes were found to be significantly expressed by SMR. Lastly, we observed these genes were mainly enriched in immune pathways and signaling transduction. These indings could provide new insights into the etiology of comorbidity and have implications for future therapeutic trial.
Collapse
Affiliation(s)
- Yanjing Chen
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunhua Fan
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan, China
| |
Collapse
|
15
|
Ljungdahl A, Sanders SJ. Neuropsychiatric biomarker discovery: go big or go home. Trends Mol Med 2023; 29:878-879. [PMID: 37714797 PMCID: PMC10879988 DOI: 10.1016/j.molmed.2023.09.002] [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: 08/21/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Technological advances have enabled high-throughput omics assays, such as parallelized screening of lipids across plasma samples. Pioneering a new paradigm for neuropsychiatric biomarker discovery, Yap et al. describe a large-scale systematic analysis of comprehensive phenotypic, genotypic, environmental, and lipidomic data to unravel the intricate interplay between these and autism-associated traits.
Collapse
Affiliation(s)
- Alicia Ljungdahl
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, OX3 7TY, UK; Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Stephan J Sanders
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, OX3 7TY, UK; Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
16
|
Wang Q, Yang Q, Liu X. The microbiota-gut-brain axis and neurodevelopmental disorders. Protein Cell 2023; 14:762-775. [PMID: 37166201 PMCID: PMC10599644 DOI: 10.1093/procel/pwad026] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 04/27/2023] [Indexed: 05/12/2023] Open
Abstract
The gut microbiota has been found to interact with the brain through the microbiota-gut-brain axis, regulating various physiological processes. In recent years, the impacts of the gut microbiota on neurodevelopment through this axis have been increasingly appreciated. The gut microbiota is commonly considered to regulate neurodevelopment through three pathways, the immune pathway, the neuronal pathway, and the endocrine/systemic pathway, with overlaps and crosstalks in between. Accumulating studies have identified the role of the microbiota-gut-brain axis in neurodevelopmental disorders including autism spectrum disorder, attention deficit hyperactivity disorder, and Rett Syndrome. Numerous researchers have examined the physiological and pathophysiological mechanisms influenced by the gut microbiota in neurodevelopmental disorders (NDDs). This review aims to provide a comprehensive overview of advancements in research pertaining to the microbiota-gut-brain axis in NDDs. Furthermore, we analyzed both the current state of research progress and discuss future perspectives in this field.
Collapse
Affiliation(s)
- Qinwen Wang
- State Key Laboratory of Reproductive Medicine and offspring Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Department of Pathogen Biology-Microbiology Division, Key Laboratory of Pathogen of Jiangsu Province Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Qianyue Yang
- State Key Laboratory of Reproductive Medicine and offspring Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Department of Pathogen Biology-Microbiology Division, Key Laboratory of Pathogen of Jiangsu Province Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Xingyin Liu
- State Key Laboratory of Reproductive Medicine and offspring Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Department of Pathogen Biology-Microbiology Division, Key Laboratory of Pathogen of Jiangsu Province Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
- Department of Microbiota Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
17
|
Arora A, Becker M, Marques C, Oksanen M, Li D, Mastropasqua F, Watts ME, Arora M, Falk A, Daub CO, Lanekoff I, Tammimies K. Screening autism-associated environmental factors in differentiating human neural progenitors with fractional factorial design-based transcriptomics. Sci Rep 2023; 13:10519. [PMID: 37386098 PMCID: PMC10310850 DOI: 10.1038/s41598-023-37488-0] [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: 02/03/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023] Open
Abstract
Research continues to identify genetic variation, environmental exposures, and their mixtures underlying different diseases and conditions. There is a need for screening methods to understand the molecular outcomes of such factors. Here, we investigate a highly efficient and multiplexable, fractional factorial experimental design (FFED) to study six environmental factors (lead, valproic acid, bisphenol A, ethanol, fluoxetine hydrochloride and zinc deficiency) and four human induced pluripotent stem cell line derived differentiating human neural progenitors. We showcase the FFED coupled with RNA-sequencing to identify the effects of low-grade exposures to these environmental factors and analyse the results in the context of autism spectrum disorder (ASD). We performed this after 5-day exposures on differentiating human neural progenitors accompanied by a layered analytical approach and detected several convergent and divergent, gene and pathway level responses. We revealed significant upregulation of pathways related to synaptic function and lipid metabolism following lead and fluoxetine exposure, respectively. Moreover, fluoxetine exposure elevated several fatty acids when validated using mass spectrometry-based metabolomics. Our study demonstrates that the FFED can be used for multiplexed transcriptomic analyses to detect relevant pathway-level changes in human neural development caused by low-grade environmental risk factors. Future studies will require multiple cell lines with different genetic backgrounds for characterising the effects of environmental exposures in ASD.
Collapse
Affiliation(s)
- Abishek Arora
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Martin Becker
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Cátia Marques
- Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Marika Oksanen
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Danyang Li
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Francesca Mastropasqua
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Michelle Evelyn Watts
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Lund Stem Cell Center, Division of Neurobiology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Carsten Oliver Daub
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Stockholm, Sweden
| | - Ingela Lanekoff
- Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Kristiina Tammimies
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden.
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden.
| |
Collapse
|