1
|
Quach TT, Duchemin AM. Intelligence, brain structure, dendrites, and genes: Genetic, epigenetic and the underlying of the quadruple helix complexity. Neurosci Biobehav Rev 2025; 175:106212. [PMID: 40389043 DOI: 10.1016/j.neubiorev.2025.106212] [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: 01/20/2025] [Revised: 05/01/2025] [Accepted: 05/12/2025] [Indexed: 05/21/2025]
Abstract
Intelligence can be referred to as the mental ability to learn, comprehend abstract concepts, and solve complex problems. Twin and adoption studies have provided insights into the influence of the familial environment and highlighted the importance of heritability in the development of cognition. Detecting the relative contribution of brain areas, neuronal structures, and connectomes has brought some understanding on how various brain areas, white/gray matter structures and neuronal connectivity process information and contribute to intelligence. Using histological, anatomical, electrophysiological, neuropsychological, neuro-imaging and molecular biology methods, several key concepts have emerged: 1) the parietofrontal-hippocampal integrations probably constitute a substrate for smart behavior, 2) neuronal activity results in structural plasticity of dendritic branches responsible for information transfer, critical for learning and memory, 3) intelligent people process information efficiently, 4) the environment triggers mnemonic epigenomic programs (via dynamic regulation of chromatin accessibility, DNA methylation, loop interruption/formation and histone modification) conferring cognitive phenotypes throughout life, and 5) single/double DNA breaks are prominent in human brain disorders associated with cognitive impairment including Alzheimer's disease and schizophrenia. Along with these observations, molecular/cellular/biological studies have identified sets of specific genes associated with higher scores on intelligence tests. Interestingly, many of these genes are associated with dendritogenesis. Because dendrite structure/function is involved in cognition, the control of dendrite genesis/maintenance may be critical for understanding the landscape of general/specific cognitive ability and new pathways for therapeutic approaches.
Collapse
Affiliation(s)
- Tam T Quach
- Department of Neuroscience. The Ohio State University, Columbus, OH 43210, USA.
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
2
|
Askeland-Gjerde DE, Westlye LT, Andersson P, Korbmacher M, de Lange AM, van der Meer D, Smeland OB, Halvorsen S, Andreassen OA, Gurholt TP. Mediation Analyses Link Cardiometabolic Factors and Liver Fat With White Matter Hyperintensities and Cognitive Performance: A UK Biobank Study. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100488. [PMID: 40330223 PMCID: PMC12052680 DOI: 10.1016/j.bpsgos.2025.100488] [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: 12/20/2024] [Revised: 01/19/2025] [Accepted: 03/10/2025] [Indexed: 05/08/2025] Open
Abstract
Background Liver fat is associated with cardiometabolic disease, cerebrovascular disease, and dementia. Cerebrovascular disease, most often cerebral small vessel disease, identified by magnetic resonance imaging as white matter hyperintensities (WMHs) often contributes to dementia. However, liver fat's role in the relationship between cardiometabolic risk, WMHs, and cognitive performance is unclear. Methods In the UK Biobank cohort (N = 32,461, 52.6% female; mean age 64.2 ± 7.7 years; n = 23,354 in the cognitive performance subsample), we used linear regression to investigate associations between cardiometabolic factors measured at baseline and liver fat, WMHs, and cognitive performance measured at follow-up, which was 9.3 ± 2.0 years later on average. We used structural equation modeling to investigate whether liver fat mediated associations between cardiometabolic factors and WMHs and whether WMHs mediated associations between liver fat and cognitive performance. Results Nearly all cardiometabolic factors were significantly associated with liver fat (|r| range = 0.03-0.41, p = 3.4 × 10-8 to 0) and WMHs (|r| = 0.04-0.15, p = 5.8 × 10-13 to 7.0 × 10-159) in regression models. Liver fat was associated with WMHs (r = 0.11, p = 4.3 × 10-82) and cognitive performance (r = -0.03, p = 1.6 × 10-7). Liver fat mediated the associations between cardiometabolic factors and WMHs (|βmediation| = 0.003-0.027, p mediation = 1.9 × 10-8 to 0), and WMHs mediated the associations between liver fat and cognitive performance (βmediation = -0.01, p mediation = 0). Conclusions Our findings indicate that liver fat mediates associations between cardiometabolic factors and WMHs and that WMHs mediate the association between liver fat and cognitive performance. This suggests that liver fat may be important for understanding the effects of cardiometabolic factors on cerebrovascular disease and cognitive function. Experimental studies are warranted to determine relevant targets for preventing vascular-driven cognitive impairment.
Collapse
Affiliation(s)
- Daniel E. Askeland-Gjerde
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars T. Westlye
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | | | - Max Korbmacher
- Neuro-SysMed Center of Excellence for Clinical Research in Neurological Diseases, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Department of Health and Functioning, Western Norway University of Applied Sciences, Bergen, Norway
| | - Ann-Marie de Lange
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Dennis van der Meer
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Olav B. Smeland
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sigrun Halvorsen
- Department of Cardiology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Tiril P. Gurholt
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
3
|
Jeon S, Kang JE, Hwang J, Calhoun VD, Lee JH. Abnormal association between neural activity and genetic expressions of impulsivity in attention deficit hyperactivity disorder: an Adolescent Brain Cognitive Development study. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2025:S2451-9022(25)00195-8. [PMID: 40514009 DOI: 10.1016/j.bpsc.2025.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 05/29/2025] [Accepted: 06/03/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND Impulsivity in highly heritable attention deficit hyperactivity disorder (ADHD) has been studied using neural activity via fMRI or genetic data, but rarely with multivariate methods linking both. We investigated coupled neural activity and gene expression signatures, using parallel independent component analysis (pICA) and Adolescent Brain Cognitive Development data. METHODS Children with ADHD (n = 394; 63% males) and healthy controls (n = 1,000; 47% males) of European ancestry were included. The subjects were randomly divided into 80% discovery and 20% replication datasets with demographic stratification. We analyzed neural activity and gene expressions from the discovery datasets using pICA and extracted paired independent components (pICs). The loading coefficients of the pICs were utilized to predict behavioral and cognitive data for stop signal task (SST) in replication datasets. RESULTS We identified three pICs estimated from gene expression in the cortex, cerebellum, and nucleus accumbens. Significant neural activity was mainly localized to the orbital/inferior/middle frontal gyri, rectal gyrus, precuneus, inferior temporal gyrus, inferior parietal lobule, and cerebellum. Significant gene components were associated with immunoglobulin, taste receptor, and immunity-related terms and were overlapped with ADHD-related genes. The extracted fMRI-/Gene-ICs were significantly correlated with mean reaction time, stop signal reaction time of SST, and behavioral inhibition with a large boost in sensitivity when both the paired fMRI-/Gene-ICs and their interaction were used in a multimodal regression analysis. CONCLUSION We reported biologically plausible pairs of neural activity and gene sets using pICA, which were significantly associated with ADHD impulsivity-related behavioral and cognitive data.
Collapse
Affiliation(s)
- Soohyun Jeon
- Department of Brain and Cognitive Engineering, Korea University, Seoul, South Korea
| | - Jae-Eon Kang
- Department of Brain and Cognitive Engineering, Korea University, Seoul, South Korea
| | - Jundong Hwang
- Department of Brain and Cognitive Engineering, Korea University, Seoul, South Korea
| | - Vince D Calhoun
- Center for Translational Research in Neuroimaging and Data Science (TReNDS): Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, USA; Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, USA
| | - Jong-Hwan Lee
- Department of Brain and Cognitive Engineering, Korea University, Seoul, South Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul, South Korea; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Boston, MA.
| |
Collapse
|
4
|
Chen HW, Wang SA, Xu ZY, Shao ZH, Zhong Q, Wei YF, Cao BF, Liu K, Wu XB. Association of Predicted Visceral Fat Percentage With Dementia Risk in Older Adults: The Role of Genetic Risk and Lifestyle. Neurology 2025; 104:e213630. [PMID: 40373249 DOI: 10.1212/wnl.0000000000213630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/05/2025] [Indexed: 05/17/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Obesity is a modifiable dementia risk factor, but body mass index does not account for fat distribution, particularly visceral fat, which is more strongly linked to metabolic and cardiovascular health. Despite its relevance, research on visceral fat and dementia is limited, especially in large-scale prospective studies. The aim of this study was to assess the association between visceral fat and dementia risk, its interaction with genetic predisposition, and the impact of healthy lifestyle adherence. METHODS Using data from the UK Biobank (UKB) cohort, we computed baseline, sex-specific visceral fat percentage (VFP), defined as the ratio of visceral fat mass to total body fat mass. Nonlinear associations between VFP and incident dementia were initially assessed using restricted cubic splines. The relationship between VFP and incident dementia was further examined using Cox proportional hazard models. In addition, stratified and interaction analyses were conducted to assess dementia incidence across VFP levels, lifestyle factors, and genetic risk. RESULTS The study included 63,042 women (mean age: 63.96 years) and 74,001 men (64.20 years) aged 60 years and older from the UKB, with a median follow-up of 14.07 years for men and 14.09 years for women. During follow-up, 2,805 men and 1,893 women developed dementia. A U-shaped association between VFP and dementia risk was observed in both sexes. In men, each SD increase in VFP below the median value of 8.1% was associated with a reduced risk of dementia (HR: 0.90, 95% CI 0.85-0.96), whereas above the median, the risk increased (1.06, 1.00-1.11). Similarly, in women, below the median VFP value of 3.1%, each SD increase was linked to a decreased dementia risk (0.89, 0.83-0.96), and above the median, the risk increased (1.14, 1.07-1.22). No significant interactions were found between VFP and genetic risk or lifestyle factors. DISCUSSION Among nondemented, community-dwelling older Britons, atypical VFP was associated with higher dementia risk in both sexes. The lack of interaction between VFP and genetic risk highlights the complexity of dementia pathogenesis. In addition, a healthy lifestyle may mitigate the dementia risk associated with atypical VFP levels.
Collapse
Affiliation(s)
- Hao-Wen Chen
- Department of Epidemiology, School of Public Health, Southern Medical University GuangZhou City, GuangDong Province, China
| | - Shi-Ao Wang
- Department of Epidemiology, School of Public Health, Southern Medical University GuangZhou City, GuangDong Province, China
| | - Zheng-Yun Xu
- Department of Epidemiology, School of Public Health, Southern Medical University GuangZhou City, GuangDong Province, China
| | - Zhan-Hui Shao
- Department of Epidemiology, School of Public Health, Southern Medical University GuangZhou City, GuangDong Province, China
| | - Qi Zhong
- Department of Epidemiology, School of Public Health, Southern Medical University GuangZhou City, GuangDong Province, China
| | - Yan-Fei Wei
- Department of Epidemiology, School of Public Health, Southern Medical University GuangZhou City, GuangDong Province, China
| | - Bi-Fei Cao
- Department of Epidemiology, School of Public Health, Southern Medical University GuangZhou City, GuangDong Province, China
| | - Kuan Liu
- Department of Epidemiology, School of Public Health, Southern Medical University GuangZhou City, GuangDong Province, China
| | - Xian-Bo Wu
- Department of Epidemiology, School of Public Health, Southern Medical University GuangZhou City, GuangDong Province, China
| |
Collapse
|
5
|
Zhang L, Ivleva EI, Parker DA, Hill SK, Lizano PL, Keefe RSE, Keedy SK, McDowell JE, Pearlson GD, Clementz BA, Keshavan MS, Gershon ES, Tamminga CA, Sweeney JA, Bishop JR. Impact of Polygenic Interactions With Anticholinergic Burden on Cognition and Brain Structure in Psychosis Spectrum Disorders. Am J Psychiatry 2025:appiajp20240709. [PMID: 40432343 DOI: 10.1176/appi.ajp.20240709] [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] [Indexed: 05/29/2025]
Abstract
OBJECTIVE The authors sought to determine whether genetic predispositions to cognitive ability or psychiatric conditions interact with anticholinergic burden (AChB) to impact cognition and brain structure in individuals with psychotic disorders. METHODS Participants with psychosis spectrum disorders (N=1,704) from the Bipolar-Schizophrenia Network for Intermediate Phenotypes (B-SNIP) consortium, 18-65 years of age and representing diverse ancestries, underwent cognitive assessments, structural neuroimaging, genotyping, and a comprehensive medication review. The primary cognitive outcome was the Brief Assessment of Cognition in Schizophrenia (BACS) composite score, and the primary brain structural phenotype was total gray matter volume. AChB scores for scheduled medications were quantified using the CRIDECO Anticholinergic Load Scale. Polygenic scores (PGSs) for cognition, schizophrenia, bipolar disorder, and depression were constructed, and a composite psychiatric PGS was subsequently generated. Linear regression models were used to examine AChB-PGS interactions and their associations with cognitive and brain structure outcomes, adjusting for clinical covariates and multiple testing with false discovery rate. Hypothesis-driven moderated mediation models were used to explore potential association pathways. RESULTS Higher AChB was significantly associated with lower BACS performance and reduced gray matter volume. Individuals with higher cognitive PGS values exhibited greater adverse effects of AChB on BACS, while those with lower composite psychiatric PGS values showed more pronounced gray matter volume reductions from AChB. AChB associations with cognitive impairment were partially mediated by reduced gray matter volume and were moderated by composite psychiatric PGS. CONCLUSIONS Anticholinergic-polygenic interactions significantly impact cognition and brain structure in individuals with psychotic disorders, highlighting a novel gene-by-environment interaction that advances our mechanistic understanding of cognitive impairments in this population.
Collapse
Affiliation(s)
- Lusi Zhang
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Elena I Ivleva
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - David A Parker
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Scot K Hill
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Paulo L Lizano
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Richard S E Keefe
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Sarah K Keedy
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Jennifer E McDowell
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Godfrey D Pearlson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Brett A Clementz
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Matcheri S Keshavan
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Elliot S Gershon
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Carol A Tamminga
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - John A Sweeney
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| | - Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis (Zhang, Bishop); Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas (Ivleva, Tamminga); Department of Psychology and Neuroscience, University of Georgia, Athens (Parker, McDowell, Clementz); Department of Human Genetics, Emory University School of Medicine, Atlanta (Parker); Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago (Hill); Department of Psychiatry (Lizano, Keshavan) and Division of Translational Neuroscience (Lizano), Beth Israel Deaconess Medical Center, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lizano, Keshavan); Departments of Psychiatry, Neuroscience, and Psychology, Duke University, Durham, NC (Keefe); Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago (Keedy, Gershon); Departments of Psychiatry and Neurobiology, School of Medicine, Yale University, New Haven, CT (Pearlson); Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati (Sweeney); Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis (Bishop)
| |
Collapse
|
6
|
Gustavson DE, Borriello GA, Karhadkar MA, Rhee SH, Corley RP, Rhea SA, DiLalla LF, Wadsworth SJ, Friedman NP, Reynolds CA. Stability of general cognitive ability from infancy to adulthood: A combined twin and genomic investigation. Proc Natl Acad Sci U S A 2025; 122:e2426531122. [PMID: 40388623 DOI: 10.1073/pnas.2426531122] [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: 12/18/2024] [Accepted: 04/15/2025] [Indexed: 05/21/2025] Open
Abstract
Measures of general cognitive ability (GCA) are highly stable from adolescence onward, particularly at the level of genetic influences. In contrast, measurement of GCA in early life (before 3 y old) is less reliable and less is known about the stability of GCA across this period, including its relation to adult GCA. Using data from the Colorado Longitudinal Twin study (N = 1,098), we examined the stability of GCA measures across 5 time-points (years 1 to 2, 3, 7, 16, and 29), including how an array of cognitive measures given at 7 and 9 mo relate to later GCA. We then examined the genetic and environmental stability of GCA across the first 30 y of life using complementary methods: twin analyses and polygenic scores (PGSs). Two infant cognition measures, object novelty and tester-rated task orientation, predicted GCA in adulthood (r = 0.16 and 0.18, respectively). Correlational analyses were consistent with a pattern of increasing stability across development for GCA measures between year 1 to 2 and adulthood (r = 0.39 to 0.85). Subsequent twin analyses revealed that 22% of variance in adulthood GCA was captured by genetic influences on GCA from year 3 or earlier, with an additional 10% explained by shared environmental influences on GCA at year 1 to 2. PGSs for adulthood GCA and educational attainment predicted GCA from 1 to 2 y onward (βs = 0.09 to 0.44) but not infant cognition. Findings suggest that genetic and environmental influences on GCA demonstrate considerable stability as early as age 3 y, but that measures of infant cognition are less predictive of later cognitive ability.
Collapse
Affiliation(s)
- Daniel E Gustavson
- Institute for Behavioral Genetics, University of Colorado Boulder, CO 80309
- Department of Psychology and Neuroscience, University of Colorado Boulder, CO 80309
| | | | - Mohini A Karhadkar
- Institute for Behavioral Genetics, University of Colorado Boulder, CO 80309
- Department of Psychology and Neuroscience, University of Colorado Boulder, CO 80309
| | - Soo Hyun Rhee
- Institute for Behavioral Genetics, University of Colorado Boulder, CO 80309
- Department of Psychology and Neuroscience, University of Colorado Boulder, CO 80309
| | - Robin P Corley
- Institute for Behavioral Genetics, University of Colorado Boulder, CO 80309
| | - Sally-Ann Rhea
- Institute for Behavioral Genetics, University of Colorado Boulder, CO 80309
| | - Lisabeth F DiLalla
- Department of Family and Community Medicine, Southern Illinois University School of Medicine, Carbondale, IL 62901
| | - Sally J Wadsworth
- Institute for Behavioral Genetics, University of Colorado Boulder, CO 80309
| | - Naomi P Friedman
- Institute for Behavioral Genetics, University of Colorado Boulder, CO 80309
- Department of Psychology and Neuroscience, University of Colorado Boulder, CO 80309
| | - Chandra A Reynolds
- Institute for Behavioral Genetics, University of Colorado Boulder, CO 80309
- Department of Psychology and Neuroscience, University of Colorado Boulder, CO 80309
| |
Collapse
|
7
|
Fanelli G, Robinson J, Fabbri C, Bralten J, Mota NR, Arenella M, Rovný M, Sprooten E, Franke B, Kas M, Andlauer TFM, Serretti A. Shared genetics and causal relationship between sociability and the brain's default mode network. Psychol Med 2025; 55:e157. [PMID: 40400235 DOI: 10.1017/s0033291725000832] [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: 05/23/2025]
Abstract
BACKGROUND The brain's default mode network (DMN) plays a role in social cognition, with altered DMN function being associated with social impairments across various neuropsychiatric disorders. However, the genetic basis linking sociability with DMN function remains underexplored. This study aimed to elucidate the shared genetics and causal relationship between sociability and DMN-related resting-state functional MRI (rs-fMRI) traits. METHODS We conducted a comprehensive genomic analysis using large-scale genome-wide association study (GWAS) summary statistics for sociability and 31 activity and 64 connectivity DMN-related rs-fMRI traits (N = 34,691-342,461). We performed global and local genetic correlations analyses and bi-directional Mendelian randomization (MR) to assess shared and causal effects. We prioritized genes influencing both sociability and rs-fMRI traits by combining expression quantitative trait loci MR analyses, the CELLECT framework - integrating single-nucleus RNA sequencing (snRNA-seq) data with GWAS - and network propagation within a protein-protein interaction network. RESULTS Significant local genetic correlations were identified between sociability and two rs-fMRI traits, one representing spontaneous activity within the temporal cortex, the other representing connectivity between the cingulate and angular/temporal cortices. MR analyses suggested potential causal effects of sociability on 12 rs-fMRI traits. Seventeen genes were highly prioritized, with LINGO1, ELAVL2, and CTNND1 emerging as top candidates. Among these, DRD2 was also identified, serving as a robust internal validation of our approach. CONCLUSIONS By combining genomic and transcriptomic data, our gene prioritization strategy may serve as a blueprint for future studies. Our findings can guide further research into the biological mechanisms underlying sociability and its role in the development, prognosis, and treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Giuseppe Fanelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jamie Robinson
- Global Computational Biology and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Janita Bralten
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nina Roth Mota
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martina Arenella
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Forensic and Neurodevelopmental Science, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Maroš Rovný
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Emma Sprooten
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martien Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Till F M Andlauer
- Global Computational Biology and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
- Oasi Research Institute-IRCCS, Troina, Italy
| |
Collapse
|
8
|
Lesch KP, Gorbunov N. Antisocial personality disorder:Failure to balance excitation/inhibition? Neuropharmacology 2025; 268:110321. [PMID: 39855295 DOI: 10.1016/j.neuropharm.2025.110321] [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: 08/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
While healthy brain function relies on a dynamic but tightly regulated interaction between excitation (E) and inhibition (I), a spectrum of social cognition disorders, including antisocial behavior and antisocial personality disorder (ASPD), frequently ensuing from irregular neurodevelopment, may be associated with E/I imbalance and concomitant alterations in neural connectivity. Technological advances in the evaluation of structural and functional E/I balance proxies in clinical settings and in human cell culture models provide a general basis for identification of biomarkers providing a powerful concept for prevention and intervention across different dimensions of mental health and disease. In this perspective we outline a framework for research to characterize neurodevelopmental pathways to antisocial behavior and ASPD driven by (epi)genetic factors across life, and to identify molecular targets for preventing the detrimental effects of cognitive dysfunction and maladaptive social behavior, considering psychosocial experience; to validate signatures of E/I imbalance and altered myelination proxies as biomarkers of pathogenic neural circuitry mechanisms to determine etiological processes in the transition from mental health to antisocial behavior and ASPD and in the switch from prevention to treatment; to develop a neurobiologically-grounded integrative model of antisocial behavior and ASPD resultant of disrupted E/I balance, allowing to establish objective diagnoses and monitoring tools, to personalize prevention and therapeutic decisions, to predict treatment response, and thus counteract relapse; and finally, to promote transformation of dimensional disorder taxonomy and to enhance societal awareness and reception of the neurobiological basis of antisocial behavior and ASPD.
Collapse
Affiliation(s)
- Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Child- and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
| | - Nikita Gorbunov
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
9
|
Noble AJ, Adams AT, Satsangi J, Boden JM, Osborne AJ. Prenatal cannabis exposure is associated with alterations in offspring DNA methylation at genes involved in neurodevelopment, across the life course. Mol Psychiatry 2025; 30:1418-1429. [PMID: 39277688 PMCID: PMC11919715 DOI: 10.1038/s41380-024-02752-w] [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: 12/03/2023] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
Prenatal cannabis exposure (PCE) is of increasing concern globally, due to the potential impact on offspring neurodevelopment, and its association with childhood and adolescent brain development and cognitive function. However, there is currently a lack of research addressing the molecular impact of PCE, that may help to clarify the association between PCE and neurodevelopment. To address this knowledge gap, here we present epigenome-wide association study data across multiple time points, examining the effect of PCE and co-exposure with tobacco using two longitudinal studies, the Avon Longitudinal Study of Parents and Children (ALSPAC) and the Christchurch Health and Development Study (CHDS) at birth (0 y), 7 y and 15-17 y (ALSPAC), and ~27 y (CHDS). Our findings reveal genome-wide significant DNA methylation differences in offspring at 0 y, 7 y, 15-17 y, and 27 y associated with PCE alone, and co-exposure with tobacco. Importantly, we identified significantly differentially methylated CpG sites within the genes LZTS2, NPSR1, NT5E, CRIP2, DOCK8, COQ5, and LRP5 that are shared between different time points throughout development in offspring. Notably, functional pathway analysis showed enrichment for differential DNA methylation in neurodevelopment, neurotransmission, and neuronal structure pathways, and this was consistent across all timepoints in both cohorts. Given the increasing volume of epidemiological evidence that suggests a link between PCE and adverse neurodevelopmental outcomes in exposed offspring, this work highlights the need for further investigation into PCE, particularly in larger cohorts.
Collapse
Affiliation(s)
- Alexandra J Noble
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK.
| | - Alex T Adams
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
- Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
- Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Joseph M Boden
- Christchurch Health and Development Study, Department of Psychological Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Amy J Osborne
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand.
| |
Collapse
|
10
|
Cheng M, Meng Y, Zhang J, Wang X, Zhang D, Li S. The Association of Wnt Signaling Pathway Gene Variants, Blood Lipoproteins and Cognitive Function in Elderly People. Mol Neurobiol 2025:10.1007/s12035-025-04847-z. [PMID: 40140223 DOI: 10.1007/s12035-025-04847-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025]
Abstract
Wnt signaling is believed to play an important role in the nervous system. However, few studies have examined the association between gene variants of the Wnt signaling pathway and mild cognitive impairment (MCI). Additionally, the potential modulation of this association by blood lipoproteins remains poorly understood. We aimed to investigate these associations in the present analysis. The cross-sectional study comprised 459 participants from 17 villages in Jimo District, Qingdao, Shandong Province. A total of 46 single nucleotide polymorphisms (SNPs) in nine Wnt signaling pathway genes were included. Cognitive function was measured using Montreal Cognitive Assessment (MOCA). Polygenic risk scores (PRS) were used to summarize the effect of each gene. Ordered logistic regression and Poisson regression with robust variance were applied to examine the associations of SNPs with MCI and the dimension score of MOCA. Interaction analysis was conducted to verify the interaction with lipoproteins. A random forest classifier was used to develop a predictive model for MCI. The SNP PRKCA-rs2286674 was associated with MCI across three models. The risk of MCI increased by 31% and 2% for each unit increase of PRS of PRKCA and WNT7B respectively. Based on the multiplicative interaction model, the effects of certain PRSs on the risk of MCI were modified by blood lipoproteins. Integrating total PRS into the prediction model significantly improved the ability to predict MCI. Genetic variations in Wnt signaling pathway were associated with MCI in older adults. Interaction effects between gene variants and blood lipoproteins on MCI were observed.
Collapse
Affiliation(s)
- Murong Cheng
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, No. 308 Ningxia Road, 266071, Qingdao, China
| | - Yuchi Meng
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, No. 308 Ningxia Road, 266071, Qingdao, China
| | - Jiesong Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, No. 308 Ningxia Road, 266071, Qingdao, China
| | - Xueyan Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, No. 308 Ningxia Road, 266071, Qingdao, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, No. 308 Ningxia Road, 266071, Qingdao, China.
| | - Suyun Li
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, No. 308 Ningxia Road, 266071, Qingdao, China.
| |
Collapse
|
11
|
Chen Z, Wu X, Yang Q, Zhao H, Ying H, Liu H, Wang C, Zheng R, Lin H, Wang S, Li M, Wang T, Zhao Z, Xu M, Chen Y, Xu Y, Lu J, Ning G, Wang W, Luo S, Au Yeung SL, Bi Y, Zheng J. The Effect of SGLT2 Inhibition on Brain-related Phenotypes and Aging: A Drug Target Mendelian Randomization Study. J Clin Endocrinol Metab 2025; 110:1096-1104. [PMID: 39270733 PMCID: PMC11913115 DOI: 10.1210/clinem/dgae635] [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: 06/15/2024] [Revised: 08/06/2024] [Accepted: 09/12/2024] [Indexed: 09/15/2024]
Abstract
INTRODUCTION An observational study suggested sodium-glucose cotransporter 2 (SGLT2) inhibitors might promote healthy aging. However, whether brain-related phenotypes mediate this association is still a question. We applied Mendelian randomization (MR) to investigate the effect of SGLT2 inhibition on chronological age, biological age, and cognition and explore the mediation effects of brain imaging-derived phenotypes (IDPs). METHODS We selected genetic variants associated with both expression levels of SLC5A2 (Genotype-Tissue Expression and eQTLGen data; n = 129 to 31 684) and hemoglobin A1c (HbA1c) levels (UK Biobank; n = 344 182) and used them to proxy the effect of SGLT2 inhibition. Aging-related outcomes, including parental longevity (n = 389 166) and epigenetic clocks (n = 34 710), and cognitive phenotypes, including cognitive function (n = 300 486) and intelligence (n = 269 867) were derived from genome-wide association studies. Two-step MR was conducted to explore the associations between SGLT2 inhibition, IDPs, and aging outcomes and cognition. RESULTS SGLT2 inhibition was associated with longer father's attained age [years of life increase per SD (6.75 mmol/mol) reduction in HbA1c levels = 6.21, 95% confidence interval (CI) 1.27-11.15], better cognitive function (beta = .17, 95% CI 0.03-0.31), and higher intelligence (beta = .47, 95% CI 0.19-0.75). Two-step MR identified 2 IDPs as mediators linking SGLT2 inhibition with chronological age (total proportion of mediation = 22.6%), where 4 and 5 IDPs were mediators for SGLT2 inhibition on cognitive function and intelligence, respectively (total proportion of mediation = 61.6% and 68.6%, respectively). CONCLUSION Our study supported that SGLT2 inhibition increases father's attained age, cognitive function, and intelligence, which was mediated through brain images of different brain regions. Future studies are needed to investigate whether a similar effect could be observed for users of SGLT2 inhibitors.
Collapse
Affiliation(s)
- Zhihe Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qianqian Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huiling Zhao
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol BS8 2BN, UK
| | - Hui Ying
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haoyu Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chaoyue Wang
- SJTU-Ruijin-UIH Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shan Luo
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region 999077, China
| | - Shiu Lun Au Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region 999077, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol BS8 2BN, UK
| |
Collapse
|
12
|
Chaar DL, Li Z, Shang L, Ratliff SM, Mosley TH, Kardia SLR, Zhao W, Zhou X, Smith JA. Multi-Ancestry Transcriptome-Wide Association Studies of Cognitive Function, White Matter Hyperintensity, and Alzheimer's Disease. Int J Mol Sci 2025; 26:2443. [PMID: 40141087 PMCID: PMC11942532 DOI: 10.3390/ijms26062443] [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: 01/09/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Genetic variants increase the risk of neurocognitive disorders in later life, including vascular dementia (VaD) and Alzheimer's disease (AD), but the precise relationships between genetic risk factors and underlying disease etiologies are not well understood. Transcriptome-wide association studies (TWASs) can be leveraged to better characterize the genes and biological pathways underlying genetic influences on disease. To date, almost all existing TWASs on VaD and AD have been conducted using expression studies from individuals of a single genetic ancestry, primarily European. Using the joint likelihood-based inference framework in Multi-ancEstry TRanscriptOme-wide analysis (METRO), we leveraged gene expression data from European ancestry (EA) and African ancestry (AA) samples to identify genes associated with general cognitive function, white matter hyperintensity (WMH), and AD. Regions were fine-mapped using Fine-mapping Of CaUsal gene Sets (FOCUS). We identified 266, 23, 69, and 2 genes associated with general cognitive function, WMH, AD (using EA GWAS summary statistics), and AD (using AA GWAS), respectively (Bonferroni-corrected alpha = p < 2.9 × 10-6), some of which had been previously identified. Enrichment analysis showed that many of the identified genes were in pathways related to innate immunity, vascular dysfunction, and neuroinflammation. Further, the downregulation of ICA1L was associated with a higher WMH and with AD, indicating its potential contribution to overlapping AD and VaD neuropathology. To our knowledge, our study is the first TWAS on cognitive function and neurocognitive disorders that used expression mapping studies for multiple ancestries. This work may expand the benefits of TWASs beyond a single ancestry group and help to identify gene targets for pharmaceuticals or preventative treatments for dementia.
Collapse
Affiliation(s)
- Dima L. Chaar
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
| | - Zheng Li
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (Z.L.); (X.Z.)
| | - Lulu Shang
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Scott M. Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
| | - Thomas H. Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Xiang Zhou
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (Z.L.); (X.Z.)
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| |
Collapse
|
13
|
Feng T, Xiao W, Li Y, Zhao X. Exploring the neural mechanisms linking healthy aging and cognitive maintenance: insights from Mendelian randomization and mediation analyses. Cereb Cortex 2025; 35:bhaf006. [PMID: 40089937 DOI: 10.1093/cercor/bhaf006] [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: 09/15/2024] [Revised: 12/02/2024] [Accepted: 01/02/2025] [Indexed: 03/17/2025] Open
Abstract
As global population ages, maintaining cognitive health in elderly is crucial. Previous studies suggest a positive link between healthy aging and cognition, but the neural mechanisms remain unclear. This study used genome-wide association studydata to investigate neural mechanisms between healthy aging and cognition. We employed 2-sample Mendelian randomization to evaluate causal relationship between healthy aging (indexed by a multivariate genetic predictor, mvAge) and 6 cognitive measurements. We then used a 2-step Mendelian randomization approach and mediation analysis to identify brain imaging-derived phenotypes potentially mediating this relationship. Mendelian randomization analysis indicated that healthy aging had a positive causal relationship with various cognitive functions (common executive function, intelligence, cognitive performance, and fluid intelligence score). Two-step Mendelian randomization analysis identified 27 brain imaging-derived phenotypes having robust causal relationships with healthy aging and various cognitive measurements. Mediation analysis suggested that volume of subcallosal cortex might mediate effects of healthy aging on all 4 cognitive functions. Volume of cerebellum's VIIb could mediate effects on common executive functions, while fractional anisotropy in the anterior thalamic radiation might mediate effects on intelligence and cognitive performance. These findings suggest that specific brain regions may play a potential mediating role in the relationship between healthy aging and cognitive maintenance.
Collapse
Affiliation(s)
- Tianyuyi Feng
- Department of Radiology, Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China
| | - Weizhong Xiao
- Center of Vascular and interventional Surgery, Department of General Surgery, The Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, 82 Qinglong Street, Qingyang District, Chengdu, Sichuan 610031, China
| | - Yunfei Li
- Department of Radiology, Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China
| | - Xiaohu Zhao
- Department of Radiology, Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China
| |
Collapse
|
14
|
Hamlin V, Ansaf H, Heffern R, Williams-Simon PA, King EG. Multiple methods for assessing learning and memory in Drosophila melanogaster demonstrates the highly complex, context-dependent genetic underpinnings of cognitive traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640179. [PMID: 40060392 PMCID: PMC11888412 DOI: 10.1101/2025.02.26.640179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Learning and memory are fundamental for an individual to be able to respond to changing stimuli in their environment. Between individuals we see variation in their ability to perform learning and memory tasks, however, it is still largely unknown what genetic factors may impact this variability. To gain better insight to the genetic components impacting variation in learning and memory, we use recombinant inbred lines (RILs) from the Drosophila synthetic population resource (DSPR), a multiparent mapping population exhibiting natural variation in many traits. Using a reward based associative learning and memory assay, we trained flies to associate an odor with a sucrose reward under starvation condition and measured olfactory learning and memory ability in y-mazes for 50 DSPR RILs. While we do not find significant QTLs for olfactory learning or memory, we found suggestive regions that may be contributing to variability in performance when trained to different odors. We provide evidence that performance with specific odors should be considered different phenotypes and introduce new methods for analysis for olfactory y-maze assays with multiple decision points. Additionally, we compare our data to previously collected place learning and memory data to show there is limited correlation in performance outcomes.
Collapse
Affiliation(s)
- Victoria Hamlin
- Division of Biological Sciences, University of Missouri, Columbia, Missouri
| | - Huda Ansaf
- Division of Biological Sciences, University of Missouri, Columbia, Missouri
| | - Reiley Heffern
- Division of Biological Sciences, University of Missouri, Columbia, Missouri
| | | | - Elizabeth G King
- Division of Biological Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
15
|
Wu XR, Wu BS, Kang JJ, Chen LM, Deng YT, Chen SD, Dong Q, Feng JF, Cheng W, Yu JT. Contribution of copy number variations to education, socioeconomic status and cognition from a genome-wide study of 305,401 subjects. Mol Psychiatry 2025; 30:889-898. [PMID: 39215183 DOI: 10.1038/s41380-024-02717-z] [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: 02/04/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Educational attainment (EA), socioeconomic status (SES) and cognition are phenotypically and genetically linked to health outcomes. However, the role of copy number variations (CNVs) in influencing EA/SES/cognition remains unclear. Using a large-scale (n = 305,401) genome-wide CNV-level association analysis, we discovered 33 CNV loci significantly associated with EA/SES/cognition, 20 of which were novel (deletions at 2p22.2, 2p16.2, 2p12, 3p25.3, 4p15.2, 5p15.33, 5q21.1, 8p21.3, 9p21.1, 11p14.3, 13q12.13, 17q21.31, and 20q13.33, as well as duplications at 3q12.2, 3q23, 7p22.3, 8p23.1, 8p23.2, 17q12 (105 kb), and 19q13.32). The genes identified in gene-level tests were enriched in biological pathways such as neurodegeneration, telomere maintenance and axon guidance. Phenome-wide association studies further identified novel associations of EA/SES/cognition-associated CNVs with mental and physical diseases, such as 6q27 duplication with upper respiratory disease and 17q12 (105 kb) duplication with mood disorders. Our findings provide a genome-wide CNV profile for EA/SES/cognition and bridge their connections to health. The expanded candidate CNVs database and the residing genes would be a valuable resource for future studies aimed at uncovering the biological mechanisms underlying cognitive function and related clinical phenotypes.
Collapse
Affiliation(s)
- Xin-Rui Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Li-Min Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, UK
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Zheng Q, van Alten S, Lyngstad TH, Ciscato E, Sun Z, Miao J, Wu Y, Dorn S, Zheng B, Havdahl A, Corfield EC, Nivard M, Galama TJ, Turley P, Chiappori PA, Fletcher JM, Lu Q. Genetic basis of partner choice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636375. [PMID: 39975039 PMCID: PMC11838572 DOI: 10.1101/2025.02.03.636375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Previous genetic studies of human assortative mating have primarily focused on searching for its genomic footprint but have revealed limited insights into its biological and social mechanisms. Combining insights from the economics of the marriage market with advanced tools in statistical genetics, we perform the first genome-wide association study (GWAS) on a latent index for partner choice. Using 206,617 individuals from four global cohorts, we uncover phenotypic characteristics and social processes underlying assortative mating. We identify a broadly robust genetic component of the partner choice index between sexes and several countries and identify its genetic correlates. We also provide solutions to reduce assortative mating-driven biases in genetic studies of complex traits by conditioning GWAS summary statistics on the genetic associations with the latent partner choice index.
Collapse
Affiliation(s)
- Qinwen Zheng
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, WI, USA 53706
| | - Sjoerd van Alten
- School of Business and Economics, Vrije Universiteit Amsterdam, Amsterdam, NL
| | | | - Edoardo Ciscato
- Department of Economics of KU Leuven, Katholieke Universiteit te Leuven, Leuven, Belgium
| | - Zhongxuan Sun
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, WI, USA 53706
| | - Jiacheng Miao
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, WI, USA 53706
| | - Yuchang Wu
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, WI, USA 53706
| | - Stephen Dorn
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, WI, USA 53706
| | - Boyan Zheng
- Department of Sociology, University of Wisconsin–Madison, Madison, WI, USA 53706
| | - Alexandra Havdahl
- PROMENTA Research Centre, Department of Psychology, University of Oslo, Oslo, Norway 0349
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway 0473
- Psychiatric Genetic Epidemiology Group, Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway 0456
| | - Elizabeth C. Corfield
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway 0473
- Psychiatric Genetic Epidemiology Group, Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway 0456
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Michel Nivard
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Titus J. Galama
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA 90089
| | - Patrick Turley
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA 90089
- Department of Economics, University of Southern California, Los Angeles, CA, USA 90089
| | | | - Jason M. Fletcher
- La Follette School of Public Affairs, University of Wisconsin–Madison, Madison, WI 53706
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, WI, USA 53706
| |
Collapse
|
17
|
Patel PC. Look before you leap: Earnings gaps and elderly self-employment. JOURNAL OF BUSINESS RESEARCH 2025; 189:115081. [DOI: 10.1016/j.jbusres.2024.115081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
18
|
Taylor H, Lewins M, Foody MGB, Gray O, Bešević J, Conroy MC, Collins R, Lacey B, Allen N, Burkitt-Gray L. UK Biobank-A Unique Resource for Discovery and Translation Research on Genetics and Neurologic Disease. Neurol Genet 2025; 11:e200226. [PMID: 39911793 PMCID: PMC11796045 DOI: 10.1212/nxg.0000000000200226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/09/2024] [Indexed: 02/07/2025]
Abstract
UK Biobank is a large-scale prospective study with extensive genetic and phenotypic data from half a million adults. Participants, aged 40 to 69, were recruited from the general UK population between 2006 and 2010. During recruitment, participants completed questionnaires covering lifestyle and medical history, underwent physical measurements, and provided biological samples for long-term storage. Whole-cohort assays have been conducted, including biochemical markers, genotyping, whole-exome and whole-genome sequencing, as well as proteomics and metabolomics in large subsets of the cohort, with potential for additional assays in the future. Participants consented to link their data to electronic health records, enabling the identification of health outcomes over time. Research studies using UK Biobank data have already enhanced our understanding of the role of genetic variation in neurologic disease, offering insights into potential therapeutic approaches. The integration of genetic and imaging data has led to significant discoveries regarding the relationship between genetic variants and brain structure and function, particularly in Alzheimer disease and Parkinson disease. Genetic data have also allowed Mendelian randomization analyses to be performed, enabling further investigation into the causality of associations between behavioral and physiologic factors-such as diet and blood pressure-and neurologic outcomes. Furthermore, genetic and proteomic data have been particularly useful in identifying new drug targets for neurologic disease and in enhancing risk prediction algorithms that are increasingly applied in clinical practice to identify those at higher risk. As UK Biobank continues to be enhanced, and the cases of neurologic disease accrue over time, the study will become increasingly valuable for both discovery and translational research on genetics and neurologic disease.
Collapse
Affiliation(s)
- Hannah Taylor
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford, United Kingdom; and
- UK Biobank, Stockport, Greater Manchester, United Kingdom
| | - Melissa Lewins
- UK Biobank, Stockport, Greater Manchester, United Kingdom
| | | | - Oliver Gray
- UK Biobank, Stockport, Greater Manchester, United Kingdom
| | - Jelena Bešević
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford, United Kingdom; and
- UK Biobank, Stockport, Greater Manchester, United Kingdom
| | - Megan C Conroy
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford, United Kingdom; and
- UK Biobank, Stockport, Greater Manchester, United Kingdom
| | - Rory Collins
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford, United Kingdom; and
- UK Biobank, Stockport, Greater Manchester, United Kingdom
| | - Ben Lacey
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford, United Kingdom; and
- UK Biobank, Stockport, Greater Manchester, United Kingdom
| | - Naomi Allen
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford, United Kingdom; and
- UK Biobank, Stockport, Greater Manchester, United Kingdom
| | | |
Collapse
|
19
|
Ma H, Cong Z, Liang L, Su Z, Zhang J, Yang H, Wang M. Association of Stmn1 Polymorphism and Cognitive Function: An Observational Study in the Chinese Adults. ALPHA PSYCHIATRY 2025; 26:38719. [PMID: 40110371 PMCID: PMC11915711 DOI: 10.31083/ap38719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 03/22/2025]
Abstract
Background Stathmin1 (Stmn1) is a protein highly expressed during the development of the central nervous system. The phosphorylation of Stmn1 involves microtubule dynamics, so Stmn1 plays a vital part in neurite outgrowth and synaptic plasticity. Previous studies reported that Stmn1 genetic variants influence fear and anxiety as well as cognitive-affective processing. However, no study reported on the relationship between Stmn1 gene polymorphism and cognition in Chinese. Thus, this association was investigated in the present study. Methods A total of 129 healthy Han Chinese were genotyped for Stmn1 rs182455 polymorphism by polymerase chain reaction and restriction fragment length polymorphism analyses. Cognitive function was assessed using the Stroop Color-Word Test (SCWT) and Hopkins Verbal Learning Test-Revised (HVLT-R). Results In the present sample, rs182455 CC, CT, and TT genotypes were found in 56 (43.41%), 65 (50.39%) and 8 (6.20%) cases, respectively. The genotype distribution did not deviate from Hardy-Weinberg equilibrium (χ2 = 3.715, p = 0.054). Significant differences were found between the three rs182455 genotypes and between the CC and (CT+TT) genotype groups in the Stroop Color (SC) scores of the SCWT (F = 3.322, 2.377; p = 0.039, 0.019, respectively) and the total recall (TR) scores on the HVLT-R (F = 3.118, 2.225; p = 0.048, 0.028, respectively). There was a female-specific difference in SC scores between the three rs182455 genotypes (F = 2.318, p = 0.023). The rs182455 genotype distribution showed no significant difference between two sexes (χ2 = 1.313, p = 0.519), whereas significant differences were seen in SC and TR scores between two sexes (t = -2.294, -2.490; p = 0.023, 0.014, respectively). Conclusions The findings suggest that rs182455 Stmn1 polymorphism might affect cognitive flexibility and immediate free recall in healthy Chinese individuals, especially females.
Collapse
Affiliation(s)
- Hui Ma
- Psychological Counseling and Treatment Center, Hainan Provincial Anning Hospital, 570207 Haikou, Hainan, China
| | - Zhengtu Cong
- Psychological Counseling and Treatment Center, Hainan Provincial Anning Hospital, 570207 Haikou, Hainan, China
| | - Lijuan Liang
- Department of Clinical Psychology, The First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Zhaoxia Su
- Department of Clinical Psychology, Hainan Pingshan Hospital, 572299 Wuzhishan, Hainan, China
| | - Jing Zhang
- Psychological Counseling and Treatment Center, Hainan Provincial Anning Hospital, 570207 Haikou, Hainan, China
| | - Hua Yang
- The Seventh Department of Psychiatry, Hainan Provincial Anning Hospital, 570207 Haikou, Hainan, China
| | - Man Wang
- Department of Clinical Psychology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, 518020 Shenzhen, Guangdong, China
| |
Collapse
|
20
|
Piffer D. Directional Selection and Evolution of Polygenic Traits in Eastern Eurasia: Insights from Ancient DNA. Twin Res Hum Genet 2025:1-20. [PMID: 39881595 DOI: 10.1017/thg.2024.49] [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: 01/31/2025]
Abstract
This study explores directional selection on physical and psychosocial phenotypes in Eastern Eurasian populations, utilizing a dataset of 1245 ancient genomes. By analyzing polygenic scores (PGS) for traits including height, educational attainment (EA), IQ, autism, schizophrenia, and others, we observed significant temporal trends spanning the Holocene era. The results suggest positive selection for cognitive-related traits such as IQ, EA and autism spectrum disorder (ASD), alongside negative selection for anxiety and depression. The results for height were mixed and showed nonlinear relationships with Years Before Present (BP). These trends were partially mediated by genetic components linked to distinct ancestral populations. Regression models incorporating admixture, geography, and temporal variables were used to account for biases in population composition over time. Latitude showed a positive effect on ASD PGS, EA and height, while it had a negative effect on skin pigmentation scores. Additionally, latitude exhibited significant nonlinear effects on multiple phenotypes. The observed patterns highlight the influence of climate-mediated selection pressures on trait evolution. Spline regression revealed that several polygenic scores had nonlinear relationships with years BP. The findings provide evidence for complex evolutionary dynamics, with distinct selective pressures shaping phenotypic diversity across different timescales and environments.
Collapse
|
21
|
Gao Y, Wang D, Wang Q, Wang J, Li S, Wang T, Hu X, Wan C. Causal Impacts of Psychiatric Disorders on Cognition and the Mediating Effect of Oxidative Stress: A Mendelian Randomization Study. Antioxidants (Basel) 2025; 14:162. [PMID: 40002349 PMCID: PMC11852177 DOI: 10.3390/antiox14020162] [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: 12/31/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Many psychiatric disorders are associated with major cognitive deficits. However, it is uncertain whether these deficits develop as a result of psychiatric disorders and what shared risk factors might mediate this relationship. Here, we utilized the Mendelian randomization (MR) analysis to investigate the complex causal relationship between nine major psychiatric disorders and three cognitive phenotypes, while also examining the potential mediating role of oxidative stress as a shared biological underpinning. Schizophrenia (SZ), major depressive disorder (MDD), and attention deficit hyperactivity disorder (ADHD) showed a decreasing effect on cognitive performance, intelligence, and education, while bipolar disorder (BPD) increased educational attainment. MR-Clust results exhibit the shared genetic basis between SZ and other psychiatric disorders in relation to cognitive function. Furthermore, when oxidative stress was considered as a potential mediating factor, the associations between SZ and the three dimensions of cognition, as well as between MDD and intelligence and ADHD and intelligence, exhibited larger effect sizes than the overall. Mediation MR analysis also supported the causal effects between psychiatric disorders and cognition via oxidative stress traits, including carotene, vitamin E, bilirubin, and uric acid. Finally, summary-based MR identified 29 potential causal associations of oxidative stress genes with both cognitive performance and psychiatric disorders. Our findings highlight the importance of considering oxidative stress in understanding and potentially treating cognitive impairments associated with psychiatric conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaowen Hu
- Bio-X Institutes, Key Laboratory for The Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai 200030, China; (Y.G.); (D.W.); (Q.W.); (J.W.); (S.L.); (T.W.)
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for The Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai 200030, China; (Y.G.); (D.W.); (Q.W.); (J.W.); (S.L.); (T.W.)
| |
Collapse
|
22
|
Bjornson KJ, Kermath BA, Cahill ME. Identification of ARHGEF11 (PDZ-RhoGEF) as an in vivo regulator of synapses and cognition. Proc Natl Acad Sci U S A 2025; 122:e2415316122. [PMID: 39835891 PMCID: PMC11789018 DOI: 10.1073/pnas.2415316122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Given the influence of cognitive abilities on life outcomes, there is inherent value in identifying genes involved in controlling learning and memory. Further, cognitive dysfunction is a core feature of many neuropsychiatric disorders. Here, we use a combinatory in silico approach to identify human gene targets that will have an especially high likelihood of individually and directly impacting cognition. This broad and unbiased screen led to the specific identification of ARHGEF11, which encodes PDZ-RhoGEF. PDZ-RhoGEF is a largely RhoA-specific activator that is highly enriched in dendritic spines, and recent work identified hyperexpression of PDZ-RhoGEF in the prefrontal cortex of bipolar disorder subjects, a disease characterized by an early emergence and persistence of broad scope cognitive dysfunction. Here, we characterize the effects of PDZ-RhoGEF on synaptic and behavioral phenotypes, and we identify molecular and biochemical mechanisms that control PDZ-RhoGEF's expression, synaptic spatial localization, and enzymatic activity. Importantly, our identified direct regulators of PDZ-RhoGEF (miR-132 and DISC1) have themselves been repeatedly implicated in controlling cognitive phenotypes in humans, including those caused by several neuropsychiatric disorders. Taken together, our findings indicate that PDZ-RhoGEF is a key convergence point among multiple synaptic and cognition-relevant signaling cascades with potential translational significance.
Collapse
Affiliation(s)
- Kathryn J. Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI53706
| | - Bailey A. Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI53706
| | - Michael E. Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI53706
| |
Collapse
|
23
|
Jiang X, Zai C, Mio M, Dimick MK, Sultan AA, Young LT, Goldstein BI. Neurocognitive correlates of polygenic risk for bipolar disorder among youth with and without bipolar disorder. J Affect Disord 2025; 369:845-853. [PMID: 39426505 DOI: 10.1016/j.jad.2024.10.047] [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: 05/16/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
INTRODUCTION There is well-established evidence of reduced neurocognitive performance in adults and youth with bipolar disorder (BD). However, little is known about the polygenic underpinnings of neurocognition in individuals with BD, particularly in youth. The current study aimed to examine the association between polygenic risk score for BD (BD-PRS) and neurocognition among youth with BD and healthy controls (HC). METHODS 129 youth of European ancestry (72 BD, 57 HC), ages 13-20 years, were included. Six neurocognitive tasks within the Cambridge Neuropsychological Test Automated Battery were assessed. General linear models were used to examine the association between BD-PRS and neurocognitive composite scores, controlling for age, sex, IQ, and two genetic principal components. RESULTS In the overall sample, higher BD-PRS was associated with significantly poorer affective processing (β = -0.25, p = 0.01), decision-making (β = -0.23, p = 0.02), and sustained attention (β = -0.28, p = 0.002). Secondary analyses revealed that higher BD-PRS was associated with significantly poorer sustained attention within the BD group (β = -0.27, p = 0.04), and with significantly poorer affective processing within the HC group (β = -0.29, p = 0.04). LIMITATIONS Cross-sectional design. Modest sample size may have reduced power to detect smaller effect sizes. CONCLUSION The current study found that higher BD-PRS generated based on adult GWAS was associated with poorer neurocognitive performance in youth with BD and HC. Future longitudinal studies incorporating repeated neurocognitive assessments would further inform whether the associations of BD-PRS with neurocognition vary from youth to adulthood, and whether BD-PRS is associated with differential neurodevelopmental trajectories in individuals with and without BD.
Collapse
Affiliation(s)
- Xinyue Jiang
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Clement Zai
- Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Megan Mio
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada
| | - Mikaela K Dimick
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada
| | - Alysha A Sultan
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada
| | - L Trevor Young
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada.
| |
Collapse
|
24
|
Huang SY, Ge YJ, Ren P, Wu BS, Gong W, Du J, Chen SD, Kang JJ, Ma Q, Bokde ALW, Desrivières S, Garavan H, Grigis A, Lemaitre H, Smolka MN, Hohmann S, Feng JF, Zhang YR, Cheng W, Yu JT. Genome-wide association study unravels mechanisms of brain glymphatic activity. Nat Commun 2025; 16:626. [PMID: 39805841 PMCID: PMC11730627 DOI: 10.1038/s41467-024-55706-9] [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: 05/02/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Brain glymphatic activity, as indicated by diffusion-tensor imaging analysis along the perivascular space (ALPS) index, is involved in developmental neuropsychiatric and neurodegenerative diseases, but its genetic architecture is poorly understood. Here, we identified 17 unique genome-wide significant loci and 161 candidate genes linked to the ALPS-indexes in a discovery sample of 31,021 individuals from the UK Biobank. Seven loci were replicated in two independent datasets. Genetic signals located at the 2p23.3 locus yielded significantly concordant effects in both young and aging cohorts. Genetic correlation and polygenic overlap analyses indicate a common underlying genetic mechanism between the ALPS-index, ventricular volumes, and cerebrospinal fluid tau levels, with GMNC (3q28) and C16orf95 (16q24.2) as the shared genetic basis. Our findings enhance the understanding of the genetics of the ALPS-index and provide insight for further research into the neurobiological mechanisms of glymphatic clearance activity across the lifespan and its relation to neuropsychiatric phenotypes.
Collapse
Affiliation(s)
- Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Ren
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weikang Gong
- School of Data Science, Fudan University, Shanghai, China
| | - Jing Du
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, UNSW, Sydney, Australia
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Qing Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College, London, UK
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, 05405, Burlington, VT, USA
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Herve Lemaitre
- NeuroSpin, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, 33076, Bordeaux, France
| | - Michael N Smolka
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China.
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer 79 Center, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Pan DN, Xie H, Zeng Y, Zhou Y, Lin C, Ma X, Ren J, Jiao Y, Wu Y, Wei W, Xue G. The development and validation of a tablet-based assessment battery of general cognitive ability. BMC Psychol 2024; 12:778. [PMID: 39719650 DOI: 10.1186/s40359-024-02283-7] [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: 12/11/2023] [Accepted: 12/10/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Traditional cognitive assessments, often reliant on paper-and-pencil tests and professional evaluators, suffer from subjectivity and limited result discrimination. This study introduces the Baguan Online Cognitive Assessment System (BOCAS), a tablet-based system that evaluates both general cognitive ability (GCA) and domain-specific functions across six domains: sensory-motor skills, processing speed, sustained attention, working memory, cognitive flexibility, and spatial ability. METHODS BOCAS was validated with 151 healthy Chinese adults aged 18-40. Reliability was assessed through internal consistency and test-retest reliability. Factor analysis and confirmatory factor analysis (CFA) were used to validate the model. The GCA score was correlated with the Raven IQ test and self-reported cognitive flexibility, and its relationship with negative emotions (depression and anxiety) was examined. RESULTS BOCAS showed satisfactory reliability, with internal consistency ranging from 0.712 to 0.846 and test-retest reliability from 0.56 to 0.71. Factor analysis revealed a common factor explaining 40% of the variance, and CFA indicated a good model fit (χ²/df = 1.81; CFI = 0.932). The GCA score strongly correlated with the Raven IQ test (r = 0.58) and was related to self-reported cognitive flexibility and negative emotions. CONCLUSION BOCAS offers a digital solution for cognitive assessment, providing automated, remote, and precise evaluations. It demonstrates reliability, validity, and potential for use in clinical and research settings.
Collapse
Affiliation(s)
- Dong-Ni Pan
- School of Psychology, Beijing Language and Culture University, Beijing, 100083, China
| | - Hailun Xie
- Beijing Infinite Brain Technology Co., Ltd, Beijing, 100022, PR China
| | - Yanjia Zeng
- School of Psychology, Beijing Language and Culture University, Beijing, 100083, China
| | - Yixiang Zhou
- School of Psychology, Beijing Language and Culture University, Beijing, 100083, China
| | - Cuizhu Lin
- School of Psychology, Beijing Language and Culture University, Beijing, 100083, China
| | - Xin Ma
- School of Psychology, Beijing Language and Culture University, Beijing, 100083, China
| | - Juejing Ren
- Beijing Infinite Brain Technology Co., Ltd, Beijing, 100022, PR China
| | - Yuanyun Jiao
- Beijing Infinite Brain Technology Co., Ltd, Beijing, 100022, PR China
| | - Yingying Wu
- Beijing Infinite Brain Technology Co., Ltd, Beijing, 100022, PR China
| | - Wei Wei
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, PR China
| | - Gui Xue
- Beijing Infinite Brain Technology Co., Ltd, Beijing, 100022, PR China.
- State Key Laboratory Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
26
|
Qin L, Huang T, Zhang D, Wei L, Li G, Zhu Q, Tong Q, Ding G, Liu J. The mitochondrial function of peripheral blood cells in cognitive frailty patients. Front Aging Neurosci 2024; 16:1503246. [PMID: 39723155 PMCID: PMC11669044 DOI: 10.3389/fnagi.2024.1503246] [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: 09/28/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background Cognitive frailty (CF), characterized by the coexistence of physical frailty and cognitive impairment, is linked to increased morbidity and mortality in older adults. While CF has been linked to multiple physiological and lifestyle factors, the underlying biological mechanisms remain poorly understood. This study investigated the risk factors for CF and explored the relationship between mitochondrial function and CF in hospitalized patients. Methods A total of 279 hospitalized individuals were recruited from December 2020 to August 2022, conducted comprehensive clinical assessments, and collected peripheral blood samples. CF was evaluated using the Physical Frailty Phenotype and Montreal Cognitive Assessment scales. Nutritional status was assessed with the Mini Nutritional Assessment, and depression was measured using the Geriatric Depression Scale. DNA was obtained from the peripheral blood and interrogated for mitochondrial DNA copy number (mtDNAcn). Peripheral blood mononuclear cells isolated from peripheral blood were examined for respiratory function and reactive oxygen species (ROS) levels. Additionally, plasma samples were analyzed for inflammatory markers and Carnitine Palmitoyltransferase II (CPT2). Results Among the participants, 90 were classified as CF and 46 as non-CF. Logistic regression analysis revealed that increased age (OR 1.156, 95% CI 1.064-1.255), lower educational attainment (OR 0.115, 95% CI 0.024-0.550), malnutrition (OR 0.713, 95% CI 0.522-0.973), and higher depression scores (OR 1.345, 95% CI 1.065-1.699) were significantly associated with CF. The independent t tests and Mann-Whitney U tests showed the CF group exhibited impaired mitochondrial function, characterized by reduced mtDNAcn and respiratory activity, coupled with elevated ROS, interleukin-6, and CPT2 levels compared with the non-CF group. After adjusted for age, sex, and BMI, compared with non-CF group, the OR values for the CF group of mtDNAcn and ROS were 0.234 (95% CI = 0.065-0.849) (p = 0.027) and 1.203 (95% CI = 1.075-1.347) (p = 0.001), respectively. The Sensitive analysis showed that the area under curve values for mtDNAcn and ROS were 0.653 and 0.925. Conclusion Age, lower educational attainment, malnutrition, and depression are significant risk factors for CF. Moreover, mitochondrial dysfunction, characterized by decreased mtDNAcn, impaired respiratory function and increased ROS levels appears to be a critical phenotype of CF.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Guoxian Ding
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Liu
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
27
|
Jiang W, Zhang J, Wang M, Zou Y, Liu Q, Song Y, Sun G, Gong Y, Zhang F, Jiang B. The X-linked intellectual disability gene CUL4B is critical for memory and synaptic function. Acta Neuropathol Commun 2024; 12:188. [PMID: 39633474 PMCID: PMC11619648 DOI: 10.1186/s40478-024-01903-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] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Cullin 4B (CUL4B) is the scaffold protein in the CUL4B-RING E3 ubiquitin ligase (CRL4B) complex. Loss-of-function mutations in the human CUL4B gene lead to syndromic X-linked intellectual disability (XLID). Till now, the mechanism of intellectual disability caused by CUL4B mutation still needs to be elucidated. In this study, we used single-nucleus RNA sequencing (snRNA-seq) to investigate the impact of CUL4B deficiency on the transcriptional programs of diverse cell types. The results revealed that depletion of CUL4B resulted in impaired intercellular communication and elicited cell type-specific transcriptional changes relevant to synapse dysfunction. Golgi-Cox staining of brain slices and immunostaining of in vitro cultured neurons revealed remarkable synapse loss in CUL4B-deficient mice. Ultrastructural analysis via transmission electron microscopy (TEM) showed that the width of the synaptic cleft was significantly greater in CUL4B-deficient mice. Electrophysiological experiments found a decrease in the amplitude of AMPA receptor-mediated EPSCs in the hippocampal CA1 pyramidal neurons of CUL4B-deficient mice. These results indicate that depletion of CUL4B in mice results in morphological and functional abnormalities in synapses. Furthermore, behavioral tests revealed that depletion of CUL4B in the mouse nervous system results in impaired spatial learning and memory. Taken together, the findings of this study reveal the pathogenesis of neurological disorders associated with CUL4B mutations and promote the identification of therapeutic targets that can halt synaptic abnormalities and preserve memory in individuals.
Collapse
Affiliation(s)
- Wei Jiang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jian Zhang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Molin Wang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qiao Liu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yu Song
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Gongping Sun
- The Key Laboratory of Experimental Teratology of the Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fan Zhang
- Medical Morphology Teaching Laboratory, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
28
|
Huang QQ, Wigdor EM, Malawsky DS, Campbell P, Samocha KE, Chundru VK, Danecek P, Lindsay S, Marchant T, Koko M, Amanat S, Bonfanti D, Sheridan E, Radford EJ, Barrett JC, Wright CF, Firth HV, Warrier V, Strudwick Young A, Hurles ME, Martin HC. Examining the role of common variants in rare neurodevelopmental conditions. Nature 2024; 636:404-411. [PMID: 39567701 PMCID: PMC11634775 DOI: 10.1038/s41586-024-08217-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 10/15/2024] [Indexed: 11/22/2024]
Abstract
Although rare neurodevelopmental conditions have a large Mendelian component1, common genetic variants also contribute to risk2,3. However, little is known about how this polygenic risk is distributed among patients with these conditions and their parents nor its interplay with rare variants. It is also unclear whether polygenic background affects risk directly through alleles transmitted from parents to children, or whether indirect genetic effects mediated through the family environment4 also play a role. Here we addressed these questions using genetic data from 11,573 patients with rare neurodevelopmental conditions, 9,128 of their parents and 26,869 controls. Common variants explained around 10% of variance in risk. Patients with a monogenic diagnosis had significantly less polygenic risk than those without, supporting a liability threshold model5. A polygenic score for neurodevelopmental conditions showed only a direct genetic effect. By contrast, polygenic scores for educational attainment and cognitive performance showed no direct genetic effect, but the non-transmitted alleles in the parents were correlated with the child's risk, potentially due to indirect genetic effects and/or parental assortment for these traits4. Indeed, as expected under parental assortment, we show that common variant predisposition for neurodevelopmental conditions is correlated with the rare variant component of risk. These findings indicate that future studies should investigate the possible role and nature of indirect genetic effects on rare neurodevelopmental conditions, and consider the contribution of common and rare variants simultaneously when studying cognition-related phenotypes.
Collapse
Affiliation(s)
| | | | | | - Patrick Campbell
- Wellcome Sanger Institute, Hinxton, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Kaitlin E Samocha
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - V Kartik Chundru
- Wellcome Sanger Institute, Hinxton, UK
- Institute of Biomedical and Clinical Science, University of Exeter, Exeter, UK
| | | | | | | | | | | | | | - Eamonn Sheridan
- Wellcome Sanger Institute, Hinxton, UK
- Leeds Institute of Medical Research, University of Leeds, St. James's University Hospital, Leeds, UK
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds, UK
| | - Elizabeth J Radford
- Wellcome Sanger Institute, Hinxton, UK
- Department of Paediatrics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Caroline F Wright
- Institute of Biomedical and Clinical Science, University of Exeter, Exeter, UK
| | - Helen V Firth
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Varun Warrier
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Alexander Strudwick Young
- University of California Los Angeles Anderson School of Management, Los Angeles, CA, USA
- Human Genetics Department, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | | | | |
Collapse
|
29
|
Jaholkowski P, Bahrami S, Fominykh V, Hindley GFL, Tesfaye M, Parekh P, Parker N, Filiz TT, Nordengen K, Hagen E, Koch E, Bakken NR, Frei E, Birkenæs V, Rahman Z, Frei O, Haavik J, Djurovic S, Dale AM, Smeland OB, O'Connell KS, Shadrin AA, Andreassen OA. Charting the shared genetic architecture of Alzheimer's disease, cognition, and educational attainment, and associations with brain development. Neurobiol Dis 2024; 203:106750. [PMID: 39608471 DOI: 10.1016/j.nbd.2024.106750] [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: 06/23/2024] [Revised: 10/09/2024] [Accepted: 11/23/2024] [Indexed: 11/30/2024] Open
Abstract
The observation that the risk of developing Alzheimer's disease is reduced in individuals with high premorbid cognitive functioning, higher educational attainment, and occupational status has led to the 'cognitive reserve' hypothesis. This hypothesis suggests that individuals with greater cognitive reserve can tolerate a more significant burden of neuropathological changes before the onset of cognitive decline. The underpinnings of cognitive reserve remain poorly understood, although a shared genetic basis between measures of cognitive reserve and Alzheimer's disease has been suggested. Using the largest samples to date and novel statistical tools, we aimed to investigate shared genetic variants between Alzheimer's disease, and measures of cognitive reserve; cognition and educational attainment to identify molecular and neurobiological foundations. We applied the causal mixture model (MiXeR) to estimate the number of trait-influencing variants shared between Alzheimer's disease, cognition, and educational attainment, and condFDR/conjFDR to identify shared loci. To provide biological insights loci were functionally characterized. Subsequently, we constructed a Structural Equation Model (SEM) to determine if the polygenic foundation of cognition has a direct impact on Alzheimer's disease risk, or if its effect is mediated through established risk factors for the disease, using a case-control sample from the UK Biobank. Univariate MiXeR analysis (after excluding chromosome 19) revealed that Alzheimer's disease was substantially less polygenic (450 trait-influencing variants) compared to cognition (11,100 trait-influencing variants), and educational attainment (12,700 trait-influencing variants). Bivariate MiXeR analysis estimated that Alzheimer's disease shared approximately 70 % of trait-influencing variants with cognition, and approximately 40 % with educational attainment, with mixed effect directions. Using condFDR analysis, we identified 18 loci jointly associated with Alzheimer's disease and cognition and 6 loci jointly associated with Alzheimer's disease and educational attainment. Genes mapped to shared loci were associated with neurodevelopment, expressed in early life, and implicated the dendritic tree and phosphatidylinositol phosphate binding mechanisms. Spatiotemporal gene expression analysis of the identified genes showed that mapped genes were highly expressed during the mid-fetal period, further suggesting early neurodevelopmental stages as critical periods for establishing cognitive reserve which affect the risk of Alzheimer's disease in old age. Furthermore, our SEM analysis showed that genetic variants influencing cognition had a direct effect on the risk of developing Alzheimer's disease, providing evidence in support of the neurodevelopmental hypothesis of the disease.
Collapse
Affiliation(s)
- Piotr Jaholkowski
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Shahram Bahrami
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vera Fominykh
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Guy F L Hindley
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Markos Tesfaye
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pravesh Parekh
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nadine Parker
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tahir T Filiz
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kaja Nordengen
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Espen Hagen
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Elise Koch
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nora R Bakken
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Evgeniia Frei
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Viktoria Birkenæs
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Zillur Rahman
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Oleksandr Frei
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Jan Haavik
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway; Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Srdjan Djurovic
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Anders M Dale
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA; Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA 92093, USA; Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Olav B Smeland
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kevin S O'Connell
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexey A Shadrin
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
30
|
Miyoshi E, Morabito S, Henningfield CM, Das S, Rahimzadeh N, Shabestari SK, Michael N, Emerson N, Reese F, Shi Z, Cao Z, Srinivasan SS, Scarfone VM, Arreola MA, Lu J, Wright S, Silva J, Leavy K, Lott IT, Doran E, Yong WH, Shahin S, Perez-Rosendahl M, Head E, Green KN, Swarup V. Spatial and single-nucleus transcriptomic analysis of genetic and sporadic forms of Alzheimer's disease. Nat Genet 2024; 56:2704-2717. [PMID: 39578645 PMCID: PMC11631771 DOI: 10.1038/s41588-024-01961-x] [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] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/26/2024] [Indexed: 11/24/2024]
Abstract
The pathogenesis of Alzheimer's disease (AD) depends on environmental and heritable factors, with its molecular etiology still unclear. Here we present a spatial transcriptomic (ST) and single-nucleus transcriptomic survey of late-onset sporadic AD and AD in Down syndrome (DSAD). Studying DSAD provides an opportunity to enhance our understanding of the AD transcriptome, potentially bridging the gap between genetic mouse models and sporadic AD. We identified transcriptomic changes that may underlie cortical layer-preferential pathology accumulation. Spatial co-expression network analyses revealed transient and regionally restricted disease processes, including a glial inflammatory program dysregulated in upper cortical layers and implicated in AD genetic risk and amyloid-associated processes. Cell-cell communication analysis further contextualized this gene program in dysregulated signaling networks. Finally, we generated ST data from an amyloid AD mouse model to identify cross-species amyloid-proximal transcriptomic changes with conformational context.
Collapse
Affiliation(s)
- Emily Miyoshi
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Samuel Morabito
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
- Mathematical, Computational, and Systems Biology (MCSB) Program, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems (CCBS), University of California, Irvine, Irvine, CA, USA
| | - Caden M Henningfield
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Sudeshna Das
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Negin Rahimzadeh
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
- Mathematical, Computational, and Systems Biology (MCSB) Program, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems (CCBS), University of California, Irvine, Irvine, CA, USA
| | - Sepideh Kiani Shabestari
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Neethu Michael
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Nora Emerson
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Fairlie Reese
- Center for Complex Biological Systems (CCBS), University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Zechuan Shi
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Zhenkun Cao
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Shushrruth Sai Srinivasan
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
- Mathematical, Computational, and Systems Biology (MCSB) Program, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems (CCBS), University of California, Irvine, Irvine, CA, USA
- Department of Computer Science, University of California, Irvine, Irvine, CA, USA
| | - Vanessa M Scarfone
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Miguel A Arreola
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Jackie Lu
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Sierra Wright
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Justine Silva
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Kelsey Leavy
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Ira T Lott
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California, Irvine, School of Medicine, Orange, CA, USA
| | - Eric Doran
- Department of Pediatrics, University of California, Irvine, School of Medicine, Orange, CA, USA
| | - William H Yong
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Saba Shahin
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Mari Perez-Rosendahl
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Elizabeth Head
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA.
- Center for Complex Biological Systems (CCBS), University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
31
|
Murphy AE, Beardall W, Rei M, Phuycharoen M, Skene NG. Predicting cell type-specific epigenomic profiles accounting for distal genetic effects. Nat Commun 2024; 15:9951. [PMID: 39550354 PMCID: PMC11569248 DOI: 10.1038/s41467-024-54441-5] [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: 03/20/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024] Open
Abstract
Understanding how genetic variants affect the epigenome is key to interpreting GWAS, yet profiling these effects across the non-coding genome remains challenging due to experimental scalability. This necessitates accurate computational models. Existing machine learning approaches, while progressively improving, are confined to the cell types they were trained on, limiting their applicability. Here, we introduce Enformer Celltyping, a deep learning model which incorporates distal effects of DNA interactions, up to 100,000 base-pairs away, to predict epigenetic signals in previously unseen cell types. Using DNA and chromatin accessibility data for epigenetic imputation, Enformer Celltyping outperforms current best-in-class approaches and generalises across cell types and biological regions. Moreover, we propose a framework for evaluating models on genetic variant effect prediction using regulatory quantitative trait loci mapping studies, highlighting current limitations in genomic deep learning models. Despite this, Enformer Celltyping can also be used to study cell type-specific genetic enrichment of complex traits.
Collapse
Affiliation(s)
- Alan E Murphy
- UK Dementia Research Institute at Imperial College London, London, W12 0BZ, UK.
- Department of Brain Sciences, Imperial College London, London, W12 0BZ, UK.
| | - William Beardall
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Marek Rei
- Department of Computing, Imperial College London, London, SW7 2RH, UK
| | - Mike Phuycharoen
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Nathan G Skene
- UK Dementia Research Institute at Imperial College London, London, W12 0BZ, UK.
- Department of Brain Sciences, Imperial College London, London, W12 0BZ, UK.
| |
Collapse
|
32
|
Talaei M, Waters S, Portas L, Jacobs BM, Dodd JW, Marshall CR, Minelli C, Shaheen SO. Lung development genes, adult lung function and cognitive traits. Brain Commun 2024; 6:fcae380. [PMID: 39544701 PMCID: PMC11562126 DOI: 10.1093/braincomms/fcae380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/18/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
Lower lung function is associated with lower cognitive function and an increased risk of dementia. This has not been adequately explained and may partly reflect shared developmental pathways. In UK Biobank participants of European ancestry, we tested the association between lung function measures (forced vital capacity and forced expiratory volume in 1 s to forced vital capacity ratio; n = 306 476) and cognitive traits including nine cognitive function test scores (n = 32 321-428 609), all-cause dementia, Alzheimer's disease and vascular dementia (6805, 2859 and 1544 cases, respectively, and ∼421 241 controls). In the same population, we derived summary statistics for associations between common genetic variants in 55 lung development genes and lung function measures and cognitive traits using adjusted linear/logistic regression models. Using a hypothesis-driven Bayesian co-localization analysis, we finally investigated the presence of shared genetic signals between lung function measures and cognitive traits at each of these 55 genes. Higher lung function measures were generally associated with higher scores of cognitive function tests as well as lower risk of dementia. The strongest association was between forced vital capacity and vascular dementia (adjusted hazard ratio 0.74 per standard deviation increase, 95% confidence interval 0.67-0.83). Of the 55 genes of interest, we found shared variants in four genes, namely: CSNK2B rs9267531 (forced vital capacity and forced expiratory volume in 1 s to forced vital capacity ratio with fluid intelligence and pairs matching), NFATC3 rs548092276 & rs11275011 (forced expiratory volume in 1 s to forced vital capacity ratio with fluid intelligence), PTCH1 rs2297086 & rs539078574 (forced expiratory volume in 1 s to forced vital capacity ratio with reaction time) and KAT8 rs138259061 (forced vital capacity with pairs matching). However, the direction of effects was not in keeping with our hypothesis, i.e. variants associated with lower lung function were associated with better cognitive function or vice versa. We also found distinct variants associated with lung function and cognitive function in KAT8 (forced vital capacity and Alzheimer's disease) and PTCH1 (forced vital capacity and forced expiratory volume in 1 s to forced vital capacity ratio with fluid intelligence and reaction time). The links between CSNK2B and NFATC3 and cognitive traits have not been previously reported by genome-wide association studies. Despite shared genes and variants, our findings do not support the hypothesis that shared developmental signalling pathways explain the association of lower adult lung function with poorer cognitive function.
Collapse
Affiliation(s)
- Mohammad Talaei
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London EC1M 6BQ, UK
| | - Sheena Waters
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London EC1M 6BQ, UK
| | - Laura Portas
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, UK
| | - Benjamin M Jacobs
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London EC1M 6BQ, UK
| | - James W Dodd
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 2BN, UK
- Academic Respiratory Unit, Southmead Hospital, University of Bristol, Bristol BS10 5NB, UK
| | - Charles R Marshall
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London EC1M 6BQ, UK
| | - Cosetta Minelli
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Seif O Shaheen
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London EC1M 6BQ, UK
- Allergy and Lung Health Unit, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
33
|
Zhang S, Zhao L, Liao A, Li D, Li H, Ouyang L, Chen X, Li Z. Investigating the Shared Genetic Architecture Between Psychiatric Disorders and Executive Function. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100392. [PMID: 39829962 PMCID: PMC11740799 DOI: 10.1016/j.bpsgos.2024.100392] [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: 07/12/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 01/03/2025] Open
Abstract
Background Evidence for widespread comorbidity of executive dysfunctions with psychiatric disorders suggests common mechanisms underlying their pathophysiology. However, the shared genetic architectures between psychiatric disorders and executive function (EF) remain poorly understood. Methods Leveraging large genome-wide association study datasets of European ancestry on bipolar disorder (N = 353,899), major depressive disorder (N = 674,452), and schizophrenia (N = 130,644) from the Psychiatric Genomics Consortium and iPSYCH and a common factor of EF (N = 427,037) from UK Biobank, we systematically investigated the shared genomic architectures between psychiatric disorders and EF with a set of statistical genetic, functional genomic, and gene-level analyses. Results Our study demonstrated substantial genetic overlaps and significant genetic correlations between psychiatric disorders and EF. EF showed an estimated 95.9%, 98.1%, and 99.2% of phenotype-influencing variants, as well as 50, 23, and 130 genomic loci shared with bipolar disorder, major depressive disorder, and schizophrenia, respectively. Single nucleotide polymorphism heritability enrichment suggests that the genetic architecture of psychiatric disorders and EF involves the brain's frontal cortex and prefrontal glutamatergic neurons 1 and 2. Functional genomic analysis of shared variants identified 12 functional regulatory variants that regulate gene expression by affecting the binding affinities of 5 transcription factors. In addition, functional characterization analyses of shared genes revealed potential common biological mechanisms related to synaptic processes and fetal brain development. Conclusions Our findings provide evidence for extensive shared genetic architectures between psychiatric disorders and EF and have valuable implications for future mechanistic investigations and drug development efforts.
Collapse
Affiliation(s)
- Sijie Zhang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Linlin Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Aijun Liao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - David Li
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hong Li
- Department of Psychiatry, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lijun Ouyang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaogang Chen
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- China National Technology Institute on Mental Disorders & Hunan Key Laboratory of Psychiatry and Mental Health, Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zongchang Li
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- China National Technology Institute on Mental Disorders & Hunan Key Laboratory of Psychiatry and Mental Health, Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
34
|
Morey RA, Zheng Y, Bayly H, Sun D, Garrett ME, Gasperi M, Maihofer AX, Baird CL, Grasby KL, Huggins AA, Haswell CC, Thompson PM, Medland S, Gustavson DE, Panizzon MS, Kremen WS, Nievergelt CM, Ashley-Koch AE, Logue MW. Genomic structural equation modeling reveals latent phenotypes in the human cortex with distinct genetic architecture. Transl Psychiatry 2024; 14:451. [PMID: 39448598 PMCID: PMC11502831 DOI: 10.1038/s41398-024-03152-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: 08/10/2023] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024] Open
Abstract
Genetic contributions to human cortical structure manifest pervasive pleiotropy. This pleiotropy may be harnessed to identify unique genetically-informed parcellations of the cortex that are neurobiologically distinct from functional, cytoarchitectural, or other cortical parcellation schemes. We investigated genetic pleiotropy by applying genomic structural equation modeling (SEM) to map the genetic architecture of cortical surface area (SA) and cortical thickness (CT) for 34 brain regions recently reported in the ENIGMA cortical GWAS. Genomic SEM uses the empirical genetic covariance estimated from GWAS summary statistics with LD score regression (LDSC) to discover factors underlying genetic covariance, which we are denoting genetically informed brain networks (GIBNs). Genomic SEM can fit a multivariate GWAS from summary statistics for each of the GIBNs, which can subsequently be used for LD score regression (LDSC). We found the best-fitting model of cortical SA identified 6 GIBNs and CT identified 4 GIBNs, although sensitivity analyses indicated that other structures were plausible. The multivariate GWASs of the GIBNs identified 74 genome-wide significant (GWS) loci (p < 5 × 10-8), including many previously implicated in neuroimaging phenotypes, behavioral traits, and psychiatric conditions. LDSC of GIBN GWASs found that SA-derived GIBNs had a positive genetic correlation with bipolar disorder (BPD), and cannabis use disorder, indicating genetic predisposition to a larger SA in the specific GIBN is associated with greater genetic risk of these disorders. A negative genetic correlation was observed between attention deficit hyperactivity disorder (ADHD) and major depressive disorder (MDD). CT GIBNs displayed a negative genetic correlation with alcohol dependence. Even though we observed model instability in our application of genomic SEM to high-dimensional data, jointly modeling the genetic architecture of complex traits and investigating multivariate genetic links across neuroimaging phenotypes offers new insights into the genetics of cortical structure and relationships to psychopathology.
Collapse
Affiliation(s)
- Rajendra A Morey
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Yuanchao Zheng
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Henry Bayly
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Delin Sun
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Melanie E Garrett
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
- Department of Medicine, Duke Molecular Physiology Institute, Carmichael Building, Duke University Medical Center, Durham, NC, 27701, USA
| | - Marianna Gasperi
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Adam X Maihofer
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - C Lexi Baird
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
| | - Katrina L Grasby
- Psychiatric Genetics, QIMR, Berghofer Medical Research Institute, 4006, Brisbane, QLD, Australia
| | - Ashley A Huggins
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Courtney C Haswell
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute Keck School of Medicine University of Southern California, Los Angeles, CA, 90033, USA
| | - Sarah Medland
- Queensland Institute for Medical Research, Berghofer Medical Research Institute, 4006, Brisbane, QLD, Australia
| | - Daniel E Gustavson
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Matthew S Panizzon
- Stein Institute for Research on Aging, University of California San Diego, La Jolla, CA, 92093, USA
| | - William S Kremen
- Stein Institute for Research on Aging, University of California San Diego, La Jolla, CA, 92093, USA
| | - Caroline M Nievergelt
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Allison E Ashley-Koch
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
- Department of Medicine, Duke Molecular Physiology Institute, Carmichael Building, Duke University Medical Center, Durham, NC, 27701, USA
| | - Mark W Logue
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA.
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, 02118, USA.
- Biomedical Genetics, Boston University School of Medicine, Boston, MA, 02118-2526, USA.
| |
Collapse
|
35
|
Ostermann PN, Wu Y, Bowler SA, Siddiqui MA, Herrera A, Sidharta M, Ramnarine K, Martínez-Meza S, St. Bernard LA, Nixon DF, Jones RB, Yamashita M, Ndhlovu LC, Zhou T, Evering TH. A Transcriptional Signature of Induced Neurons Differentiates Virologically Suppressed People Living With HIV from People Without HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619617. [PMID: 39484396 PMCID: PMC11526917 DOI: 10.1101/2024.10.22.619617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Neurocognitive impairment is a prevalent and important co-morbidity in virologically suppressed people living with HIV (PLWH), yet the underlying mechanisms remain elusive and treatments lacking. Here, we explored for the first time, use of participant-derived directly induced neurons (iNs) to model neuronal biology and injury in PLWH. iNs retain age- and disease-related features of the donors, providing unique opportunities to reveal novel aspects of neurological disorders. We obtained primary dermal fibroblasts from six virologically suppressed PLWH (range: 27 - 64 years, median: 53); 83% Male; 50% White) and seven matched people without HIV (PWOH) (range: 27 - 66, median: 55); 71% Male; 57% White). iNs were generated using transcription factors NGN2 and ASCL1, and validated by immunocytochemistry and single-cell-RNAseq. Transcriptomic analysis using bulk-RNAseq identified 29 significantly differentially expressed genes between iNs from PLWH and PWOH. Of these, 16 genes were downregulated and 13 upregulated in PLWH iNs. Protein-protein interaction network mapping indicates that iNs from PLWH exhibit differences in extracellular matrix organization and synaptic transmission. IFI27 was upregulated in iNs from PLWH, which complements independent post-mortem studies demonstrating elevated IFI27 expression in PLWH-derived brain tissue, indicating that iN generation reconstitutes this pathway. Finally, we observed that expression of the FOXL2NB-FOXL2-LINC01391 genome locus is reduced in iNs from PLWH and negatively correlates with neurocognitive impairment. Thus, we have identified an iN gene signature of HIV through direct reprogramming of skin fibroblasts into neurons revealing novel mechanisms of neurocognitive impairment in PLWH.
Collapse
Affiliation(s)
- Philipp N. Ostermann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Youjun Wu
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Scott A. Bowler
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mohammad Adnan Siddiqui
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY. 10032, USA
| | - Alberto Herrera
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mega Sidharta
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Kiran Ramnarine
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Samuel Martínez-Meza
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Leslie Ann St. Bernard
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Douglas F. Nixon
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - R. Brad Jones
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Masahiro Yamashita
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY. 10032, USA
| | - Lishomwa C. Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ting Zhou
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Teresa H. Evering
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
36
|
Jia Z, Zhang H, Lv Y, Yu L, Cui Y, Zhang L, Yang C, Liu H, Zheng T, Xia W, Xu S, Li Y. Intrauterine chromium exposure and cognitive developmental delay: The modifying effect of genetic predisposition. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174350. [PMID: 38960203 DOI: 10.1016/j.scitotenv.2024.174350] [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: 03/06/2024] [Revised: 06/04/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
There is limited evidence on the effects of intrauterine chromium (Cr) exposure on children's cognitive developmental delay (CDD). Further, little is known about the genetic factors in modifying the association between intrauterine Cr exposure and CDD. The present study involved 2361 mother-child pairs, in which maternal plasma Cr concentrations were assessed, a polygenic risk score for the child was constructed, and the child's cognitive development was evaluated using the Bayley Scales of Infant Development. The risks of CDD conferred by intrauterine Cr exposure in children with different genetic backgrounds were evaluated by logistic regression. The additive interaction between intrauterine Cr exposure and genetic factors was evaluated by calculating the relative excess risk due to interaction (RERI), attributable proportion due to interaction (AP), and synergy index (SI). According to present study, higher intrauterine Cr exposure was significantly associated with increased CDD risk [each unit increase in ln-transformed maternal plasma Cr concentration (ln-Cr): adjusted OR (95 % CI), 1.18 (1.04-1.35); highest vs lowest quartile: adjusted OR (95 % CI), 1.57 (1.10-2.23)]. The dose-response relationship of intrauterine Cr exposure and CDD for children with high genetic risk was more prominent [each unit increased ln-Cr: adjusted OR (95 % CI), 1.36 (1.09-1.70)]. Joint effects between intrauterine Cr exposure and genetic factors were found. Specifically, for high genetic risk carriers, the association between intrauterine Cr exposure and CDD was more evident [highest vs lowest quartile: adjusted OR (95 % CI), 2.33 (1.43-3.80)]. For those children with high intrauterine Cr exposure and high genetic risk, the adjusted AP was 0.39 (95 % CI, 0.07-0.72). Conclusively, intrauterine Cr exposure was a high-risk factor for CDD in children, particularly for those with high genetic risk. Intrauterine Cr exposure and one's adverse genetic background jointly contribute to an increased risk of CDD in children.
Collapse
Affiliation(s)
- Zhenxian Jia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hongling Zhang
- Wuchang University of Technology, Wuhan, Hubei, People's Republic of China
| | - Yiqing Lv
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ling Yu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yuan Cui
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Liping Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Chenhui Yang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hongxiu Liu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Tongzhang Zheng
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI 02912, United States
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Shunqing Xu
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, People's Republic of China.
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
37
|
Dinneen TJ, Ní Ghrálaigh F, Ormond C, Heron EA, Kirov G, Lopez LM, Gallagher L. Polygenic scores stratify neurodevelopmental copy number variant carrier cognitive outcomes in the UK Biobank. NPJ Genom Med 2024; 9:43. [PMID: 39341812 PMCID: PMC11438881 DOI: 10.1038/s41525-024-00426-8] [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] [Received: 01/03/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024] Open
Abstract
Rare copy-number variants associated with neurodevelopmental conditions (ND-CNVs) exhibit variable expressivity of clinical, physical, behavioural outcomes. Findings from clinically ascertained cohorts suggest this variability may be partly due to additional genetic variation. Here, we assessed the impact of polygenic scores (PGS) and rare variants on ND-CNV carrier fluid intelligence (FI) scores in the UK Biobank. Greater PGS for cognition (PSCog) and educational attainment (PSEA) is associated with increased FI scores in all ND-CNVs (n = 1317), 15q11.2 del. (n = 543), and 16p13.11 dup. carriers (n = 275). No association of rare variants associated with intellectual disability, autism, or putatively loss-of-function, brain-expressed genes was found. Positive predictive values in the first deciles of PScog and PSEA showed a two- to five-fold increase in the rate of low FI scores compared to baseline rates. These findings demonstrate that PGS can stratify ND-CNV carrier cognitive outcomes in a population-based cohort.
Collapse
Affiliation(s)
- Thomas J Dinneen
- Trinity College Dublin, Department of Psychiatry, School of Medicine, Trinity Centre for Health Sciences, St. James' Hospital, Dublin 8, Ireland.
- The Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St., Toronto, ON, M5G 0A4, Canada.
| | - Fiana Ní Ghrálaigh
- Department of Biology, Maynooth University, Maynooth, Co, Kildare, Ireland
| | - Cathal Ormond
- Trinity College Dublin, Department of Psychiatry, School of Medicine, Trinity Centre for Health Sciences, St. James' Hospital, Dublin 8, Ireland
| | - Elizabeth A Heron
- Trinity College Dublin, Department of Psychiatry, School of Medicine, Trinity Centre for Health Sciences, St. James' Hospital, Dublin 8, Ireland
| | - George Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Lorna M Lopez
- Department of Biology, Maynooth University, Maynooth, Co, Kildare, Ireland
| | - Louise Gallagher
- Trinity College Dublin, Department of Psychiatry, School of Medicine, Trinity Centre for Health Sciences, St. James' Hospital, Dublin 8, Ireland
- The Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St., Toronto, ON, M5G 0A4, Canada
- Centre for Addiction and Mental Health, 80 Workman Way, Toronto, ON, M6J 1H4, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A1, Canada
| |
Collapse
|
38
|
Vicuña L, Barrientos E, Leiva-Yamaguchi V, Alvares D, Mericq V, Pereira A, Eyheramendy S. Joint models reveal genetic architecture of pubertal stage transitions and their association with BMI in admixed Chilean population. Hum Mol Genet 2024; 33:1660-1670. [PMID: 38981621 DOI: 10.1093/hmg/ddae098] [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: 11/28/2023] [Revised: 05/01/2024] [Accepted: 06/04/2024] [Indexed: 07/11/2024] Open
Abstract
Early or late pubertal onset can lead to disease in adulthood, including cancer, obesity, type 2 diabetes, metabolic disorders, bone fractures, and psychopathologies. Thus, knowing the age at which puberty is attained is crucial as it can serve as a risk factor for future diseases. Pubertal development is divided into five stages of sexual maturation in boys and girls according to the standardized Tanner scale. We performed genome-wide association studies (GWAS) on the "Growth and Obesity Chilean Cohort Study" cohort composed of admixed children with mainly European and Native American ancestry. Using joint models that integrate time-to-event data with longitudinal trajectories of body mass index (BMI), we identified genetic variants associated with phenotypic transitions between pairs of Tanner stages. We identified $42$ novel significant associations, most of them in boys. The GWAS on Tanner $3\rightarrow 4$ transition in boys captured an association peak around the growth-related genes LARS2 and LIMD1 genes, the former of which causes ovarian dysfunction when mutated. The associated variants are expression and splicing Quantitative Trait Loci regulating gene expression and alternative splicing in multiple tissues. Further, higher individual Native American genetic ancestry proportions predicted a significantly earlier puberty onset in boys but not in girls. Finally, the joint models identified a longitudinal BMI parameter significantly associated with several Tanner stages' transitions, confirming the association of BMI with pubertal timing.
Collapse
Affiliation(s)
- Lucas Vicuña
- Department of Medicine, Genetics Section, University of Chicago, Chicago, IL 60637, United States
| | - Esteban Barrientos
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Santiago, Chile
| | | | - Danilo Alvares
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
| | - Veronica Mericq
- Institute of Maternal and Child Research, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Anita Pereira
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Susana Eyheramendy
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Santiago, Chile
- Data Observatory Foundation, ANID Technology Center No. DO210001, Chile
- Instituto Milenio Fundamentos de los Datos, Chile
| |
Collapse
|
39
|
Gracheva AS, Kashatnikova DA, Redkin IV, Zakharchenko VE, Kuzovlev AN, Salnikova LE. Genetics and Traumatic Brain Injury: Findings from an Exome-Based Study of a 50-Patient Case Series. Curr Issues Mol Biol 2024; 46:10351-10368. [PMID: 39329968 PMCID: PMC11430351 DOI: 10.3390/cimb46090616] [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: 08/21/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of global mortality and morbidity. Because TBI is accident-related, the role of genetics in predisposing to TBI has been largely unexplored. However, the likelihood of injury may not be entirely random and may be associated with certain physical and mental characteristics. In this study, we analyzed the exomes of 50 patients undergoing rehabilitation after TBI. Patients were divided into three groups according to rehabilitation outcome: improvement, no change, and deterioration/death. We focused on rare, potentially functional missense and high-impact variants in genes intolerant to these variants. The concordant results from the three independent groups of patients allowed for the suggestion of the existence of a genetic predisposition to TBI, associated with rare functional variations in intolerant genes, with a prevalent dominant mode of inheritance and neurological manifestations in the genetic phenotypes according to the OMIM database. Forty-four of the 50 patients had one or more rare, potentially deleterious variants in one or more neurological genes. Comparison of these results with those of a 50-sampled matched non-TBI cohort revealed significant differences: P = 2.6 × 10-3, OR = 4.89 (1.77-13.47). There were no differences in the distribution of the genes of interest between the TBI patient groups. Our exploratory study provides new insights into the impact of genetics on TBI risk and is the first to address potential genetic susceptibility to TBI.
Collapse
Affiliation(s)
- Alesya S Gracheva
- The Department of Population Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Darya A Kashatnikova
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- The Laboratory of Molecular Pathophysiology, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Ivan V Redkin
- The Laboratory of Organoprotection in Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Vladislav E Zakharchenko
- The Department of Clinical Laboratory Diagnostics, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Artem N Kuzovlev
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Lyubov E Salnikova
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- The Laboratory of Molecular Immunology, National Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia
| |
Collapse
|
40
|
Guo J, Yang P, Wang JH, Tang SH, Han JZ, Yao S, Yu K, Liu CC, Dong SS, Zhang K, Duan YY, Yang TL, Guo Y. Blood metabolites, neurocognition and psychiatric disorders: a Mendelian randomization analysis to investigate causal pathways. Transl Psychiatry 2024; 14:376. [PMID: 39285197 PMCID: PMC11405529 DOI: 10.1038/s41398-024-03095-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Neurocognitive dysfunction is observationally associated with the risk of psychiatric disorders. Blood metabolites, which are readily accessible, may become highly promising biomarkers for brain disorders. However, the causal role of blood metabolites in neurocognitive function, and the biological pathways underlying their association with psychiatric disorders remain unclear. METHODS To explore their putative causalities, we conducted bidirectional two-sample Mendelian randomization (MR) using genetic variants associated with 317 human blood metabolites (nmax = 215,551), g-Factor (an integrated index of multiple neurocognitive tests with nmax = 332,050), and 10 different psychiatric disorders (n = 9,725 to 807,553) from the large-scale genome-wide association studies of European ancestry. Mediation analysis was used to assess the potential causal pathway among the candidate metabolite, neurocognitive trait and corresponding psychiatric disorder. RESULTS MR evidence indicated that genetically predicted acetylornithine was positively associated with g-Factor (0.035 standard deviation units increase in g-Factor per one standard deviation increase in acetylornithine level; 95% confidence interval, 0.021 to 0.049; P = 1.15 × 10-6). Genetically predicted butyrylcarnitine was negatively associated with g-Factor (0.028 standard deviation units decrease in g-Factor per one standard deviation increase in genetically proxied butyrylcarnitine; 95% confidence interval, -0.041 to -0.015; P = 1.31 × 10-5). There was no evidence of associations between genetically proxied g-Factor and metabolites. Furthermore, the mediation analysis via two-step MR revealed that the causal pathway from acetylornithine to bipolar disorder was partly mediated by g-Factor, with a mediated proportion of 37.1%. Besides, g-Factor mediated the causal pathway from butyrylcarnitine to schizophrenia, with a mediated proportion of 37.5%. Other neurocognitive traits from different sources provided consistent findings. CONCLUSION Our results provide genetic evidence that acetylornithine protects against bipolar disorder through neurocognitive abilities, while butyrylcarnitine has an adverse effect on schizophrenia through neurocognition. These findings may provide insight into interventions at the metabolic level for risk of neurocognitive and related disorders.
Collapse
Affiliation(s)
- Jing Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Ping Yang
- Hunan Brain Hospital, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, P. R. China
| | - Jia-Hao Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Shi-Hao Tang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Ji-Zhou Han
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Shi Yao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, China
| | - Ke Yu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Cong-Cong Liu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Kun Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Yuan-Yuan Duan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China.
| |
Collapse
|
41
|
Li M, Dang X, Chen Y, Chen Z, Xu X, Zhao Z, Wu D. Cognitive processing speed and accuracy are intrinsically different in genetic architecture and brain phenotypes. Nat Commun 2024; 15:7786. [PMID: 39242605 PMCID: PMC11379965 DOI: 10.1038/s41467-024-52222-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
Since the birth of cognitive science, researchers have used reaction time and accuracy to measure cognitive ability. Although recognition of these two measures is often based on empirical observations, the underlying consensus is that most cognitive behaviors may be along two fundamental dimensions: cognitive processing speed (CPS) and cognitive processing accuracy (CPA). In this study, we used genomic-wide association studies (GWAS) data from 14 cognitive traits to show the presence of those two factors and revealed the specific neurobiological basis underlying them. We identified that CPS and CPA had distinct brain phenotypes (e.g. white matter microstructure), neurobiological bases (e.g. postsynaptic membrane), and developmental periods (i.e. late infancy). Moreover, those two factors showed differential associations with other health-related traits such as screen exposure and sleep status, and a significant causal relationship with psychiatric disorders such as major depressive disorder and schizophrenia. Utilizing an independent cohort from the Adolescent Brain Cognitive Development (ABCD) study, we also uncovered the distinct contributions of those two factors on the cognitive development of young adolescents. These findings reveal two fundamental factors underlying various cognitive abilities, elucidate the distinct brain structural fingerprint and genetic architecture of CPS and CPA, and hint at the complex interrelationship between cognitive ability, lifestyle, and mental health.
Collapse
Affiliation(s)
- Mingyang Li
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Xixi Dang
- Department of Psychology, Hangzhou Normal University, Hangzhou, China
| | - Yiwei Chen
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Zhifan Chen
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Xinyi Xu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Zhiyong Zhao
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China.
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
- Binjiang Institute, Zhejiang University, Hangzhou, China.
| |
Collapse
|
42
|
Hu T, Parrish RL, Dai Q, Buchman AS, Tasaki S, Bennett DA, Seyfried NT, Epstein MP, Yang J. Omnibus proteome-wide association study identifies 43 risk genes for Alzheimer disease dementia. Am J Hum Genet 2024; 111:1848-1863. [PMID: 39079537 PMCID: PMC11393696 DOI: 10.1016/j.ajhg.2024.07.001] [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: 01/18/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 09/08/2024] Open
Abstract
Transcriptome-wide association study (TWAS) tools have been applied to conduct proteome-wide association studies (PWASs) by integrating proteomics data with genome-wide association study (GWAS) summary data. The genetic effects of PWAS-identified significant genes are potentially mediated through genetically regulated protein abundance, thus informing the underlying disease mechanisms better than GWAS loci. However, existing TWAS/PWAS tools are limited by considering only one statistical model. We propose an omnibus PWAS pipeline to account for multiple statistical models and demonstrate improved performance by simulation and application studies of Alzheimer disease (AD) dementia. We employ the Aggregated Cauchy Association Test to derive omnibus PWAS (PWAS-O) p values from PWAS p values obtained by three existing tools assuming complementary statistical models-TIGAR, PrediXcan, and FUSION. Our simulation studies demonstrated improved power, with well-calibrated type I error, for PWAS-O over all three individual tools. We applied PWAS-O to studying AD dementia with reference proteomic data profiled from dorsolateral prefrontal cortex of postmortem brains from individuals of European ancestry. We identified 43 risk genes, including 5 not identified by previous studies, which are interconnected through a protein-protein interaction network that includes the well-known AD risk genes TOMM40, APOC1, and APOC2. We also validated causal genetic effects mediated through the proteome for 27 (63%) PWAS-O risk genes, providing insights into the underlying biological mechanisms of AD dementia and highlighting promising targets for therapeutic development. PWAS-O can be easily applied to studying other complex diseases.
Collapse
Affiliation(s)
- Tingyang Hu
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Division of Biostatistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Randy L Parrish
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA 30322, USA
| | - Qile Dai
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA 30322, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael P Epstein
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jingjing Yang
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
43
|
Xia R, Jian X, Rodrigue AL, Bressler J, Boerwinkle E, Cui B, Daviglus ML, DeCarli C, Gallo LC, Glahn DC, Knowles EEM, Moon J, Mosley TH, Satizabal CL, Sofer T, Tarraf W, Testai F, Blangero J, Seshadri S, González HM, Fornage M. Admixture mapping of cognitive function in diverse Hispanic and Latino adults: Results from the Hispanic Community Health Study/Study of Latinos. Alzheimers Dement 2024; 20:6070-6081. [PMID: 38946675 PMCID: PMC11497725 DOI: 10.1002/alz.14082] [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: 04/11/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION We conducted admixture mapping and fine-mapping analyses to identify ancestry-of-origin loci influencing cognitive abilities. METHODS We estimated the association of local ancestry intervals across the genome with five neurocognitive measures in 7140 diverse Hispanic and Latino adults (mean age 55 years). We prioritized genetic variants in associated loci and tested them for replication in four independent cohorts. RESULTS We identified nine local ancestry-associated regions for the five neurocognitive measures. There was strong biological support for the observed associations to cognitive function at all loci and there was statistical evidence of independent replication at 4q12, 9p22.1, and 13q12.13. DISCUSSION Our study identified multiple novel loci harboring genes implicated in cognitive functioning and dementia, and uncovered ancestry-relevant genetic variants. It adds to our understanding of the genetic architecture of cognitive function in Hispanic and Latino adults and demonstrates the power of admixture mapping to discover unique haplotypes influencing cognitive function, complementing genome-wide association studies. HIGHLIGHTS We identified nine ancestry-of-origin chromosomal regions associated with five neurocognitive traits. In each associated region, we identified single nucleotide polymorphisms (SNPs) that explained, at least in part, the admixture signal and were tested for replication in independent samples of Black, non-Hispanic White, and Hispanic/Latino adults with the same or similar neurocognitive tests. Statistical evidence of independent replication of the prioritized SNPs was observed for three of the nine associations, at chr4q12, chr9p22.1, and chr13q12.13. At all loci, there was strong biological support for the observed associations to cognitive function and dementia, prioritizing genes such as KIT, implicated in autophagic clearance of neurotoxic proteins and on mast cell and microglial-mediated inflammation; SLC24A2, implicated in synaptic plasticity associated with learning and memory; and MTMR6, implicated in phosphoinositide lipids metabolism.
Collapse
Affiliation(s)
- Rui Xia
- Institute of Molecular Medicine, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Xueqiu Jian
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Amanda L. Rodrigue
- Department of Psychiatry, Harvard Medical SchoolBoston Children's HospitalBostonMassachusettsUSA
| | - Jan Bressler
- Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Biqi Cui
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Martha L. Daviglus
- Institute for Minority Health ResearchUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - Charles DeCarli
- Department of NeurologyUniversity of California DavisSacramentoCaliforniaUSA
| | - Linda C. Gallo
- Department of PsychologySan Diego State UniversitySan DiegoCaliforniaUSA
| | - David C. Glahn
- Department of Psychiatry, Harvard Medical SchoolBoston Children's HospitalBostonMassachusettsUSA
| | - Emma E. M. Knowles
- Department of Psychiatry, Harvard Medical SchoolBoston Children's HospitalBostonMassachusettsUSA
| | - Jee‐Young Moon
- Department of Epidemiology & Population HealthAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Thomas H. Mosley
- The MIND CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Department of Population Health SciencesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Tamar Sofer
- Department of MedicineHarvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
- CardioVascular InstituteBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Wassim Tarraf
- Institute of Gerontology & Department of Healthcare SciencesWayne State UniversityDetroitMichiganUSA
| | - Fernando Testai
- Department of Neurology and RehabilitationUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - John Blangero
- Department of Human Genetics, South Texas Diabetes and Obesity InstituteUniversity of Texas Rio Grande ValleyBrownsvilleTexasUSA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Hector M. González
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Myriam Fornage
- Institute of Molecular Medicine, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
- Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| |
Collapse
|
44
|
Gustavson DE, Morrison CL, Mallard TT, Jennings MV, Fontanillas P, Elson SL, Palmer AA, Friedman NP, Sanchez-Roige S. Executive Function and Impulsivity Predict Distinct Genetic Variance in Internalizing Problems, Externalizing Problems, Thought Disorders, and Compulsive Disorders: A Genomic Structural Equation Modeling Study. Clin Psychol Sci 2024; 12:865-881. [PMID: 39323941 PMCID: PMC11423426 DOI: 10.1177/21677026231207845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Individual differences in self-control predict many health and life outcomes. Building on twin literature, we used genomic structural equation modeling to test the hypothesis that genetic influences on executive function and impulsivity predict independent variance in mental health and other outcomes. The impulsivity factor (comprising urgency, lack of premeditation, and other facets) was only modestly genetically correlated with low executive function (rg =.13). Controlling for impulsivity, low executive function was genetically associated with increased internalizing (βg =.15), externalizing (βg =.13), thought disorders (βg =.38), compulsive disorders (βg =.22), and chronotype (βg =.11). Controlling for executive function, impulsivity was positively genetically associated with internalizing (βg =.36), externalizing (βg =.55), body mass index (βg =.26), and insomnia (βg =.35), and negatively genetically associated with compulsive disorders (βg = -.17). Executive function and impulsivity were both genetically correlated with general cognitive ability and educational attainment. This work suggests that executive function and impulsivity are genetically separable and show independent associations with mental health.
Collapse
Affiliation(s)
- Daniel E Gustavson
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO
| | - Claire L Morrison
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Travis T Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| | - Mariela V Jennings
- Department of Psychiatry, University of California San Diego, La Jolla CA, USA
| | | | | | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Naomi P Friedman
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
45
|
Camerota M, Lester BM, McGowan EC, Carter BS, Check J, Dansereau LM, DellaGrotta SA, Helderman JB, Hofheimer JA, Loncar CM, Neal CR, O’Shea TM, Pastyrnak SL, Smith LM, Abrishamcar S, Hüls A, Marsit CJ, Everson TM. Contributions of prenatal risk factors and neonatal epigenetics to cognitive outcome in children born very preterm. Dev Psychol 2024; 60:1606-1619. [PMID: 38358663 PMCID: PMC11618652 DOI: 10.1037/dev0001709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Children born less than 30 weeks gestational age (GA) are at high risk for neurodevelopmental delay compared to term peers. Prenatal risk factors and neonatal epigenetics could help identify preterm children at highest risk for poor cognitive outcomes. We aimed to understand the associations among cumulative prenatal risk, neonatal DNA methylation, and child cognitive ability at age 3 years, including whether DNA methylation mediates the association between prenatal risk and cognitive ability. We studied 379 neonates (54% male) born less than 30 weeks GA who had DNA methylation measured at neonatal intensive care unit discharge along with 3-year follow-up data. Cumulative prenatal risk was calculated from 24 risk factors obtained from maternal report and medical record and epigenome-wide neonatal DNA methylation was assayed from buccal swabs. At 3-year follow-up, child cognitive ability was assessed using the Bayley Scales of Infant and Toddler Development (third edition). Cumulative prenatal risk and DNA methylation at two cytosine-phosphate-guanines (CpGs) were uniquely associated with child cognitive ability. Using high-dimensional mediation analysis, we also identified differential methylation of 309 CpGs that mediated the association between cumulative prenatal risk and child cognitive ability. Many of the associated CpGs were located in genes (TNS3, TRAPPC4, MAD1L1, APBB2, DIP2C, TRAPPC9, DRD2) that have previously been associated with prenatal exposures and/or neurodevelopmental phenotypes. Our findings suggest a role for both prenatal risk factors and DNA methylation in explaining outcomes for children born preterm and suggest we should further study DNA methylation as a potential mechanism underlying the association between prenatal risk and child neurodevelopment. (PsycInfo Database Record (c) 2024 APA, all rights reserved).
Collapse
Affiliation(s)
- Marie Camerota
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI
- Center for the Study of Children at Risk, Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI
| | - Barry M. Lester
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI
- Center for the Study of Children at Risk, Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI
- Department of Pediatrics, Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI
| | - Elisabeth C. McGowan
- Center for the Study of Children at Risk, Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI
- Department of Pediatrics, Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI
| | - Brian S. Carter
- Department of Pediatrics-Neonatology, Children’s Mercy Hospital, Kansas City, MO
| | - Jennifer Check
- Department of Pediatrics, Wake Forest School of Medicine, Winston-Salem, NC
| | - Lynne M. Dansereau
- Department of Pediatrics, Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI
| | - Sheri A. DellaGrotta
- Department of Pediatrics, Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI
| | | | - Julie A. Hofheimer
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Cynthia M. Loncar
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI
- Center for the Study of Children at Risk, Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI
- Department of Pediatrics, Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI
| | - Charles R. Neal
- Department of Pediatrics, University of Hawaii John A. Burns School of Medicine, Honolulu, HI
| | - T. Michael O’Shea
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Steven L. Pastyrnak
- Department of Pediatrics, Spectrum Health-Helen DeVos Hospital, Grand Rapids, MI
| | - Lynne M. Smith
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Sarina Abrishamcar
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
| | - Anke Hüls
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA
| | - Carmen J. Marsit
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA
| | - Todd M. Everson
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA
| |
Collapse
|
46
|
Leger BS, Meredith JJ, Ideker T, Sanchez-Roige S, Palmer AA. Rare and common variants associated with alcohol consumption identify a conserved molecular network. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1704-1715. [PMID: 39031522 PMCID: PMC11576244 DOI: 10.1111/acer.15399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified hundreds of common variants associated with alcohol consumption. In contrast, genetic studies of alcohol consumption that use rare variants are still in their early stages. No prior studies of alcohol consumption have examined whether common and rare variants implicate the same genes and molecular networks, leaving open the possibility that the two approaches might identify distinct biology. METHODS To address this knowledge gap, we used publicly available alcohol consumption GWAS summary statistics (GSCAN, N = 666,978) and whole exome sequencing data (Genebass, N = 393,099) to identify a set of common and rare variants for alcohol consumption. We used gene-based analysis to implicate genes from common and rare variant analyses, which we then propagated onto a shared molecular network using a network colocalization procedure. RESULTS Gene-based analysis of each dataset implicated 294 (common variants) and 35 (rare variants) genes, including ethanol metabolizing genes ADH1B and ADH1C, which were identified by both analyses, and ANKRD12, GIGYF1, KIF21B, and STK31, which were identified in only the rare variant analysis, but have been associated with other neuropsychiatric traits. Network colocalization revealed significant network overlap between the genes identified via common and rare variants. The shared network identified gene families that function in alcohol metabolism, including ADH, ALDH, CYP, and UGT. Seventy-one of the genes in the shared network were previously implicated in neuropsychiatric or substance use disorders but not alcohol-related behaviors (e.g. EXOC2, EPM2A, and CACNG4). Differential gene expression analysis showed enrichment in the liver and several brain regions. CONCLUSIONS Genes implicated by network colocalization identify shared biology relevant to alcohol consumption, which also underlie neuropsychiatric traits and substance use disorders that are comorbid with alcohol use, providing a more holistic understanding of two disparate sources of genetic information.
Collapse
Affiliation(s)
- Brittany S Leger
- Program in Biomedical Sciences, University of California San Diego, La Jolla, California, USA
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - John J Meredith
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, California, USA
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, California, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
47
|
Wang Q, Liu Y, Xu S, Liu F, Huang L, Xu F, Liu Y. Development and validation of the eMCI-CHD tool: A multivariable prediction model for the risk of mild cognitive impairment in patients with coronary heart disease. J Evid Based Med 2024; 17:535-549. [PMID: 39107928 DOI: 10.1111/jebm.12632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 07/23/2024] [Indexed: 09/30/2024]
Abstract
OBJECTIVE This study aimed to develop and validate an eMCI-CHD tool based on clinical data to predict mild cognitive impairment (MCI) risk in patients with coronary heart disease (CHD). METHODS This cross-sectional study prospectively collected data from 400 patients with coronary heart disease (aged 55-90 years, 62% men) from July 2022 to September 2023 and randomized (7:3 ratio) them into training and validation sets. After determining the modeling variables through least absolute shrinkage and selection operator regression analysis, four ML classifiers were developed: logistic regression, extreme gradient boosting (XGBoost), support vector machine, and random forest. The performance of the models was evaluated using area under the ROC curve, accuracy, sensitivity, specificity, and F1 score. Decision curve analysis was used to assess the clinical performance of the established models. The SHapley Additive exPlanations (SHAP) method was applied to determine the significance of the features, the predictive model was visualized with a nomogram, and an online web-based calculator for predicting CHD-MCI risk scores was developed. RESULTS Of 400 CHD patients (average age 70.86 ± 8.74 years), 220 (55%) had MCI. The XGBoost model demonstrated superior performance (AUC: 0.86, accuracy: 78.57%, sensitivity: 0.74, specificity: 0.84, F1: 0.79) and underwent validation. An online tool (https://mr.cscps.com.cn/mci/index.html) with seven predictive variables (APOE gene typing, age, education, TyG index, NT-proBNP, C-reactive protein, and occupation) assessed MCI risk in CHD patients. CONCLUSION This study highlights the potential for predicting MCI risk among CHD patients using an ML model-driven nomogram and risk scoring tool based on clinical data.
Collapse
Affiliation(s)
- Qing Wang
- The Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanfei Liu
- The Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Xu
- The Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fenglan Liu
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Luqi Huang
- China Evidence-Based Medicine Center of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- The Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yue Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
48
|
Balqis-Ali NZ, Ahmad N, Minhat HS, Fattah Azman AZ. Biopsychosocial factors of depression among community-dwelling geriatric population with low perceived social support; a population-based study. BMC Geriatr 2024; 24:685. [PMID: 39143517 PMCID: PMC11323693 DOI: 10.1186/s12877-024-05211-x] [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: 04/19/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Although significant and disabling consequences are presented due to geriatric population-related depression, an insufficient comprehension of various biological, psychological, and social factors affecting this issue has been observed. Notably, these factors can contribute to geriatric population-related depression with low social support. This study aimed to identify factors associated with depression among the community-dwelling geriatric population with low social support in Malaysia. METHODS This study used secondary data from a population-based health survey in Malaysia, namely the National Health Morbidity Survey (NHMS) 2018: Elderly Health. The analysis included 926 community-dwelling geriatric population aged 60 and above with low social support. The primary data collection was from August to October 2018, using face-to-face interviews. This paper reported the analysis of depression as the dependent variable, while various biological, psychological and social factors, guided by established biopsychosocial models, were the independent variables. Multiple logistic regression was applied to identify the factors. Analysis was performed using the complex sampling module in the IBM SPSS version 29. RESULTS The weighted prevalence of depression among the community-dwelling geriatric population aged 60 and above with low social support was 22.5% (95% CI: 17.3-28.7). This was significantly higher than depression among the general geriatric Malaysian population. The factors associated with depression were being single, as compared to those married (aOR 2.010, 95% CI: 1.063-3.803, p: 0.031), having dementia, as opposed to the absence of the disease (aOR 3.717, 95% CI: 1.544-8.888, p: 0.003), and having a visual disability, as compared to regular visions (aOR 3.462, 95% CI: 1.504-7.972, p: 0.004). The analysis also revealed that a one-unit increase in control in life and self-realisation scores were associated with a 32.6% (aOR: 0.674, 95% CI: 0.599-0.759, p < 0.001) and 24.7% (aOR: 0.753, 95% CI: 0.671-0.846, p < 0.001) decrease in the likelihood of developing depression, respectively. CONCLUSION This study suggested that conducting depression screenings for the geriatric population with low social support could potentially prevent or improve the management of depression. The outcome could be achieved by considering the identified risk factors while implementing social activities, which enhanced control and self-fulfilment.
Collapse
Affiliation(s)
- Nur Zahirah Balqis-Ali
- Institute for Health Systems Research, National Institutes of Health, Ministry of Health, Selangor, Malaysia
- Department of Community Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Norliza Ahmad
- Department of Community Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| | - Halimatus Sakdiah Minhat
- Department of Community Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Ahmad Zaid Fattah Azman
- Department of Community Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
49
|
Voglewede MM, Ozsen EN, Ivak N, Bernabucci M, Tang R, Sun M, Pang ZP, Zhang H. Loss of the polarity protein Par3 promotes dendritic spine neoteny and enhances learning and memory. iScience 2024; 27:110308. [PMID: 39045101 PMCID: PMC11263792 DOI: 10.1016/j.isci.2024.110308] [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: 09/12/2023] [Revised: 03/25/2024] [Accepted: 06/17/2024] [Indexed: 07/25/2024] Open
Abstract
The Par3 polarity protein is critical for subcellular compartmentalization in different developmental processes. Variants of PARD3, encoding PAR3, are associated with intelligence and neurodevelopmental disorders. However, the role of Par3 in glutamatergic synapse formation and cognitive functions in vivo remains unknown. Here, we show that forebrain-specific Par3 conditional knockout leads to increased long, thin dendritic spines in vivo. In addition, we observed a decrease in the amplitude of miniature excitatory postsynaptic currents. Surprisingly, loss of Par3 enhances hippocampal-dependent spatial learning and memory and repetitive behavior. Phosphoproteomic analysis revealed proteins regulating cytoskeletal dynamics are significantly dysregulated downstream of Par3. Mechanistically, we found Par3 deletion causes increased Rac1 activation and dysregulated microtubule dynamics through CAMSAP2. Together, our data reveal an unexpected role for Par3 as a molecular gatekeeper in regulating the pool of immature dendritic spines, a rate-limiting step of learning and memory, through modulating Rac1 activation and microtubule dynamics in vivo.
Collapse
Affiliation(s)
- Mikayla M. Voglewede
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Elif Naz Ozsen
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Noah Ivak
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Matteo Bernabucci
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Ruizhe Tang
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Miao Sun
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Zhiping P. Pang
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
50
|
Zhao B, Li Y, Fan Z, Wu Z, Shu J, Yang X, Yang Y, Wang X, Li B, Wang X, Copana C, Yang Y, Lin J, Li Y, Stein JL, O'Brien JM, Li T, Zhu H. Eye-brain connections revealed by multimodal retinal and brain imaging genetics. Nat Commun 2024; 15:6064. [PMID: 39025851 PMCID: PMC11258354 DOI: 10.1038/s41467-024-50309-w] [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: 06/23/2023] [Accepted: 07/02/2024] [Indexed: 07/20/2024] Open
Abstract
The retina, an anatomical extension of the brain, forms physiological connections with the visual cortex of the brain. Although retinal structures offer a unique opportunity to assess brain disorders, their relationship to brain structure and function is not well understood. In this study, we conducted a systematic cross-organ genetic architecture analysis of eye-brain connections using retinal and brain imaging endophenotypes. We identified novel phenotypic and genetic links between retinal imaging biomarkers and brain structure and function measures from multimodal magnetic resonance imaging (MRI), with many associations involving the primary visual cortex and visual pathways. Retinal imaging biomarkers shared genetic influences with brain diseases and complex traits in 65 genomic regions, with 18 showing genetic overlap with brain MRI traits. Mendelian randomization suggests bidirectional genetic causal links between retinal structures and neurological and neuropsychiatric disorders, such as Alzheimer's disease. Overall, our findings reveal the genetic basis for eye-brain connections, suggesting that retinal images can help uncover genetic risk factors for brain disorders and disease-related changes in intracranial structure and function.
Collapse
Affiliation(s)
- Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Statistics, Purdue University, West Lafayette, IN, 47907, USA.
- Applied Mathematics and Computational Science Graduate Group, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Population Aging Research Center, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Yujue Li
- Department of Statistics, Purdue University, West Lafayette, IN, 47907, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zhenyi Wu
- Department of Statistics, Purdue University, West Lafayette, IN, 47907, USA
| | - Juan Shu
- Department of Statistics, Purdue University, West Lafayette, IN, 47907, USA
| | - Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN, 47907, USA
| | - Yilin Yang
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Bingxuan Li
- Department of Computer Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Xiyao Wang
- Department of Computer Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Carlos Copana
- Department of Statistics, Purdue University, West Lafayette, IN, 47907, USA
| | - Yue Yang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jinjie Lin
- Yale School of Management, Yale University, New Haven, CT, 06511, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joan M O'Brien
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Diseases, Philadelphia, PA, 19104, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|