151
|
Liu X, Shi L, Li E, Jia S. Associations of hearing loss and structural changes in specific cortical regions: a Mendelian randomization study. Cereb Cortex 2024; 34:bhae084. [PMID: 38494888 DOI: 10.1093/cercor/bhae084] [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: 12/04/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/19/2024] Open
Abstract
INTRODUCTION Previous studies have suggested a correlation between hearing loss (HL) and cortical alterations, but the specific brain regions that may be affected are unknown. METHODS Genome-wide association study (GWAS) data for 3 subtypes of HL phenotypes, sensorineural hearing loss (SNHL), conductive hearing loss, and mixed hearing loss, were selected as exposures, and GWAS data for brain structure-related traits were selected as outcomes. The inverse variance weighted method was used as the main estimation method. RESULTS Negative associations were identified between genetically predicted SNHL and brain morphometric indicators (cortical surface area, cortical thickness, or volume of subcortical structures) in specific brain regions, including the bankssts (β = -0.006 mm, P = 0.016), entorhinal cortex (β = -4.856 mm2, P = 0.029), and hippocampus (β = -24.819 cm3, P = 0.045), as well as in brain regions functionally associated with visual perception, including the pericalcarine (β = -10.009 cm3, P = 0.013). CONCLUSION Adaptive changes and functional remodeling of brain structures occur in patients with genetically predicted HL. Brain regions functionally associated with auditory perception, visual perception, and memory function are the main brain regions vulnerable in HL.
Collapse
Affiliation(s)
- Xiaoduo Liu
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, 45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Lubo Shi
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, 95 Yong'an Road, Xicheng District, Beijing, 100050, China
| | - Enze Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, National Clinical Research Center for Cardiovascular Diseases, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Shuo Jia
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, National Clinical Research Center for Cardiovascular Diseases, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| |
Collapse
|
152
|
Su S, Wang R, Chen Z, Zhou F. The causal effect of sarcopenia-associated traits on brain cortical structure: A Mendelian randomization study. Arch Gerontol Geriatr 2024; 118:105302. [PMID: 38056106 DOI: 10.1016/j.archger.2023.105302] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/26/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Previous observational studies have reported sarcopenia can affect the structure and function of brain cortical structure. However, the causality inferred from those studies was subjected to residual confounding and reverse causation. Herein, we use a two-sample Mendelian randomization (MR) analysis to illustrate the causal effect of sarcopenia-associated traits on brain cortical structure. METHODS We selected appendicular lean mass (ALM), hand grip strength (left and right) (HGSL and HGSR), and usual walking pace (UWP) to symbolize sarcopenia. The definition of brain cortical structure is human brain cortical surface area (SA) and cortical thickness (TH) globally and in 34 functional regions measured by magnetic resonance imaging. Instrumental variables at the genome-wide significance level were obtained from publicly available datasets, and inverse variance weighted as the primary method was used for MR analysis. RESULT At the global level, we found ALM (β=2604.68, 95 % confidence interval (CI): 1886.17 to 3323.19, P = 1.20 × 10-12) and HGSR (β=4733.05, 95 % CI: 2245.08 to 7221.01, P = 1.93 × 10-4) were associated with increased SA. At the region level, the SA of 25 functional gyrus without global weighted was influenced by ALM. The HGSR significantly increased SA of medial orbitofrontal and precentral gyrus without global weighted and ALM was associated with decrease of TH of lateral occipital gyrus with global weighted. No pleiotropy was detected. CONCLUSION This was the first MR study investigated the causal effect of sarcopenia-associated traits on brain cortical structure. In our study, we revealed genetically predicted sarcopenia-associated traits including ALM and HGSR could affect brain cortical structure.
Collapse
Affiliation(s)
- Shilong Su
- Department of Orthopedics, Peking University Third Hospital, No. 49 North Garden Road, Haidian 100191, Beijing, China; Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian 100191, Beijing, China
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, No. 49 North Garden Road, Haidian 100191, Beijing, China; Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian 100191, Beijing, China
| | - Zhengyang Chen
- Department of Orthopedics, Peking University Third Hospital, No. 49 North Garden Road, Haidian 100191, Beijing, China; Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian 100191, Beijing, China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, No. 49 North Garden Road, Haidian 100191, Beijing, China; Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian 100191, Beijing, China.
| |
Collapse
|
153
|
Goltermann O, Alagöz G, Molz B, Fisher SE. Neuroimaging genomics as a window into the evolution of human sulcal organization. Cereb Cortex 2024; 34:bhae078. [PMID: 38466113 PMCID: PMC10926775 DOI: 10.1093/cercor/bhae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/03/2024] [Accepted: 02/04/2024] [Indexed: 03/12/2024] Open
Abstract
Primate brain evolution has involved prominent expansions of the cerebral cortex, with largest effects observed in the human lineage. Such expansions were accompanied by fine-grained anatomical alterations, including increased cortical folding. However, the molecular bases of evolutionary alterations in human sulcal organization are not yet well understood. Here, we integrated data from recently completed large-scale neuroimaging genetic analyses with annotations of the human genome relevant to various periods and events in our evolutionary history. These analyses identified single-nucleotide polymorphism (SNP) heritability enrichments in fetal brain human-gained enhancer (HGE) elements for a number of sulcal structures, including the central sulcus, which is implicated in human hand dexterity. We zeroed in on a genomic region that harbors DNA variants associated with left central sulcus shape, an HGE element, and genetic loci involved in neurogenesis including ZIC4, to illustrate the value of this approach for probing the complex factors contributing to human sulcal evolution.
Collapse
Affiliation(s)
- Ole Goltermann
- Max Planck School of Cognition, Stephanstrasse 1a, 04103 Leipzig, Germany
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, The Netherlands
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Gökberk Alagöz
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, The Netherlands
| | - Barbara Molz
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, The Netherlands
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition & Behaviour, Radboud University, Thomas van Aquinostraat 4, 6525 GD Nijmegen, The Netherlands
| |
Collapse
|
154
|
Li J, Long Z, Sheng W, Du L, Qiu J, Chen H, Liao W. Transcriptomic Similarity Informs Neuromorphic Deviations in Depression Biotypes. Biol Psychiatry 2024; 95:414-425. [PMID: 37573006 DOI: 10.1016/j.biopsych.2023.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) is complicated by population heterogeneity, motivating the investigation of biotypes through imaging-derived phenotypes. However, neuromorphic heterogeneity in MDD remains unclear, and how the correlated gene expression (CGE) connectome constrains these neuromorphic anomalies in MDD biotypes has not yet been studied. METHODS Here, we related cortical thickness deviations in MDD biotypes to a pattern of CGE connectome. Cortical thickness was estimated from 3-dimensional T1-weighted magnetic resonance images in 2 independent cohorts (discovery cohort: N = 425; replication cohort: N = 217). The transcriptional activity was measured according to Allen Human Brain Atlas. A density peak-based clustering algorithm was used to identify MDD biotypes. RESULTS We found that patients with MDD were clustered into 2 replicated biotypes based on single-patient regional deviations from healthy control participants across 2 datasets. Biotype 1 mainly exhibited cortical thinning across the brain, whereas biotype 2 mainly showed cortical thickening in the brain. Using brainwide gene expression data, we found that deviations of transcriptionally connected neighbors predicted regional deviation for both biotypes. Furthermore, putative CGE-informed epicenters of biotype 1 were concentrated on the cognitive control circuit, whereas biotype 2 epicenters were located in the social perception circuit. The patterns of epicenter likelihood were separately associated with depression- and anxiety-response maps, suggesting that epicenters of MDD biotypes may be associated with clinical efficacies. CONCLUSIONS Our findings linked the CGE connectome and neuromorphic deviations to identify distinct epicenters in MDD biotypes, providing insight into how microscale gene expressions informed MDD biotypes.
Collapse
Affiliation(s)
- Jiao Li
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Zhiliang Long
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Southwest University, Chongqing, P.R. China
| | - Wei Sheng
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Lian Du
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Jiang Qiu
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Southwest University, Chongqing, P.R. China
| | - Huafu Chen
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Wei Liao
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China.
| |
Collapse
|
155
|
Cooper R, Hayes RA, Corcoran M, Sheth KN, Arnold TC, Stein JM, Glahn DC, Jalbrzikowski M. Bridging the gap: improving correspondence between low-field and high-field magnetic resonance images in young people. Front Neurol 2024; 15:1339223. [PMID: 38585353 PMCID: PMC10995930 DOI: 10.3389/fneur.2024.1339223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/19/2024] [Indexed: 04/09/2024] Open
Abstract
Background Portable low-field-strength magnetic resonance imaging (MRI) systems represent a promising alternative to traditional high-field-strength systems with the potential to make MR technology available at scale in low-resource settings. However, lower image quality and resolution may limit the research and clinical potential of these devices. We tested two super-resolution methods to enhance image quality in a low-field MR system and compared their correspondence with images acquired from a high-field system in a sample of young people. Methods T1- and T2-weighted structural MR images were obtained from a low-field (64mT) Hyperfine and high-field (3T) Siemens system in N = 70 individuals (mean age = 20.39 years, range 9-26 years). We tested two super-resolution approaches to improve image correspondence between images acquired at high- and low-field: (1) processing via a convolutional neural network ('SynthSR'), and (2) multi-orientation image averaging. We extracted brain region volumes, cortical thickness, and cortical surface area estimates. We used Pearson correlations to test the correspondence between these measures, and Steiger Z tests to compare the difference in correspondence between standard imaging and super-resolution approaches. Results Single pairs of T1- and T2-weighted images acquired at low field showed high correspondence to high-field-strength images for estimates of total intracranial volume, surface area cortical volume, subcortical volume, and total brain volume (r range = 0.60-0.88). Correspondence was lower for cerebral white matter volume (r = 0.32, p = 0.007, q = 0.009) and non-significant for mean cortical thickness (r = -0.05, p = 0.664, q = 0.664). Processing images with SynthSR yielded significant improvements in correspondence for total brain volume, white matter volume, total surface area, subcortical volume, cortical volume, and total intracranial volume (r range = 0.85-0.97), with the exception of global mean cortical thickness (r = 0.14). An alternative multi-orientation image averaging approach improved correspondence for cerebral white matter and total brain volume. Processing with SynthSR also significantly improved correspondence across widespread regions for estimates of cortical volume, surface area and subcortical volume, as well as within isolated prefrontal and temporal regions for estimates of cortical thickness. Conclusion Applying super-resolution approaches to low-field imaging improves regional brain volume and surface area accuracy in young people. Finer-scale brain measurements, such as cortical thickness, remain challenging with the limited resolution of low-field systems.
Collapse
Affiliation(s)
- Rebecca Cooper
- Department of Psychiatry and Behavioral Sciences, Boston Children’s Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Rebecca A. Hayes
- Department of Psychiatry and Behavioral Sciences, Boston Children’s Hospital, Boston, MA, United States
| | - Mary Corcoran
- Department of Psychiatry and Behavioral Sciences, Boston Children’s Hospital, Boston, MA, United States
| | - Kevin N. Sheth
- Center for Brain and Mind Health, Yale School of Medicine, New Haven, CT, United States
| | - Thomas Campbell Arnold
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| | - Joel M. Stein
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David C. Glahn
- Department of Psychiatry and Behavioral Sciences, Boston Children’s Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, United States
| | - Maria Jalbrzikowski
- Department of Psychiatry and Behavioral Sciences, Boston Children’s Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
156
|
Vosberg DE, Jurisica I, Pausova Z, Paus T. Intrauterine growth and the tangential expansion of the human cerebral cortex in times of food scarcity and abundance. Nat Commun 2024; 15:1205. [PMID: 38350995 PMCID: PMC10864407 DOI: 10.1038/s41467-024-45409-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Tangential growth of the human cerebral cortex is driven by cell proliferation during the first and second trimester of pregnancy. Fetal growth peaks in mid-gestation. Here, we explore how genes associated with fetal growth relate to cortical growth. We find that both maternal and fetal genetic variants associated with higher birthweight predict larger cortical surface area. The relative dominance of the maternal vs. fetal variants in these associations show striking variations across birth years (1943 to 1966). The birth-year patterns vary as a function of the epigenetic status near genes differentially methylated in individuals exposed (or not) to famine during the Dutch Winter of 1944/1945. Thus, it appears that the two sets of molecular processes contribute to early cortical development to a different degree in times of food scarcity or its abundance.
Collapse
Affiliation(s)
- Daniel E Vosberg
- Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
- Department of Neuroscience, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Research Institute of the Hospital for Sick Children, Toronto, ON, Canada
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Departments of Medical Biophysics and Computer Science, and the Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Zdenka Pausova
- Research Institute of the Hospital for Sick Children, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- ECOGENE-21, Chicoutimi, Quebec, Canada
| | - Tomáš Paus
- Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada.
- Department of Neuroscience, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.
- ECOGENE-21, Chicoutimi, Quebec, Canada.
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, ON, Canada.
- Department of Psychiatry, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
157
|
Jia H, Li Z, Guo F, Hua Z, Zhou X, Li X, Li R, Liu Q, Liu Y, Dong H. Cortical structure and the risk of amyotrophic lateral sclerosis: A bidirectional Mendelian randomization study. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110872. [PMID: 37827425 DOI: 10.1016/j.pnpbp.2023.110872] [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: 02/15/2023] [Revised: 09/06/2023] [Accepted: 10/08/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Current observational studies indicate progressive brain atrophy is closely associated with the clinical feature of amyotrophic lateral sclerosis. However, it is unclear whether the changes in cortical structure are the cause or result of ALS. Our study aimed to investigate the causal relationship between cortical structure and ALS risk using a bidirectional two-sample MR study. METHODS We collected publicly available genome-wide association studies' summary statistics for cortical structure from UK Biobank and ENIGMA consortium (n = 33,992) and ALS from the Project MinE (n = 138,086). We used the inverse variance weighted method (IVW) as primary analysis in order to evaluate the causal effects. In addition, the weighted median and MR Egger methods were performed to ensure the robustness and reliability of the IVW results. RESULTS We found the decreased surface of the paracentral lobule and thickness of the frontal pole and middle temporal lobe were suggestively associated with an increased risk of ALS as well as the increased surface of medial orbitofrontal and middle temporal lobe. In another aspect, the causalities between the susceptibility to ALS and the volume of the transverse temporal gyrus and superior temporal gyrus were negative. Besides, the susceptibility to ALS might also contribute to an increased thickness of the postcentral gyrus and superior parietal gyrus. CONCLUSION In this two-sample MR analysis, we observed that multiple cortical brain regions are associated with a higher ALS risk. Further research into the underlying mechanisms is required to back up our findings.
Collapse
Affiliation(s)
- Hongning Jia
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Clinical Neurology, Ministry of Education, Shijiazhuang, Hebei, China; Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China; Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Zhiguang Li
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Department of Neurology, Xingtai Third Hospital, Xingtai, China
| | - Fei Guo
- Department of Basic Medicine, Xingtai Medical College, Xingtai, China
| | - Zixin Hua
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaomeng Zhou
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Clinical Neurology, Ministry of Education, Shijiazhuang, Hebei, China; Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| | - Xin Li
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Clinical Neurology, Ministry of Education, Shijiazhuang, Hebei, China; Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| | - Rui Li
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Clinical Neurology, Ministry of Education, Shijiazhuang, Hebei, China; Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| | - Qi Liu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Clinical Neurology, Ministry of Education, Shijiazhuang, Hebei, China; Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| | - Yaling Liu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Clinical Neurology, Ministry of Education, Shijiazhuang, Hebei, China; Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China.
| | - Hui Dong
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Clinical Neurology, Ministry of Education, Shijiazhuang, Hebei, China; Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China.
| |
Collapse
|
158
|
Busch EL, Rapuano KM, Anderson KM, Rosenberg MD, Watts R, Casey BJ, Haxby JV, Feilong M. Dissociation of Reliability, Heritability, and Predictivity in Coarse- and Fine-Scale Functional Connectomes during Development. J Neurosci 2024; 44:e0735232023. [PMID: 38148152 PMCID: PMC10866091 DOI: 10.1523/jneurosci.0735-23.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 10/09/2023] [Accepted: 11/16/2023] [Indexed: 12/28/2023] Open
Abstract
The functional connectome supports information transmission through the brain at various spatial scales, from exchange between broad cortical regions to finer-scale, vertex-wise connections that underlie specific information processing mechanisms. In adults, while both the coarse- and fine-scale functional connectomes predict cognition, the fine scale can predict up to twice the variance as the coarse-scale functional connectome. Yet, past brain-wide association studies, particularly using large developmental samples, focus on the coarse connectome to understand the neural underpinnings of individual differences in cognition. Using a large cohort of children (age 9-10 years; n = 1,115 individuals; both sexes; 50% female, including 170 monozygotic and 219 dizygotic twin pairs and 337 unrelated individuals), we examine the reliability, heritability, and behavioral relevance of resting-state functional connectivity computed at different spatial scales. We use connectivity hyperalignment to improve access to reliable fine-scale (vertex-wise) connectivity information and compare the fine-scale connectome with the traditional parcel-wise (coarse scale) functional connectomes. Though individual differences in the fine-scale connectome are more reliable than those in the coarse-scale, they are less heritable. Further, the alignment and scale of connectomes influence their ability to predict behavior, whereby some cognitive traits are equally well predicted by both connectome scales, but other, less heritable cognitive traits are better predicted by the fine-scale connectome. Together, our findings suggest there are dissociable individual differences in information processing represented at different scales of the functional connectome which, in turn, have distinct implications for heritability and cognition.
Collapse
Affiliation(s)
- Erica L Busch
- Department of Psychology, Yale University, New Haven, Connecticut, 06510
| | - Kristina M Rapuano
- Department of Psychology, Yale University, New Haven, Connecticut, 06510
| | - Kevin M Anderson
- Department of Psychology, Yale University, New Haven, Connecticut, 06510
| | - Monica D Rosenberg
- Department of Psychology, University of Chicago, Chicago, Illinois, 60637
| | - Richard Watts
- Department of Psychology, Yale University, New Haven, Connecticut, 06510
| | - B J Casey
- Department of Psychology, Yale University, New Haven, Connecticut, 06510
| | - James V Haxby
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire, 03755
| | - Ma Feilong
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire, 03755
| |
Collapse
|
159
|
Ye Z, Zeng Q, Ning L, Huang W, Su Q. Systolic blood pressure is associated with abnormal alterations in brain cortical structure: Evidence from a Mendelian randomization study. Eur J Intern Med 2024; 120:92-98. [PMID: 37852841 DOI: 10.1016/j.ejim.2023.10.018] [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: 08/13/2023] [Revised: 09/09/2023] [Accepted: 10/13/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Hypertension has been recognized as a significant risk factor for cerebrovascular diseases and cognitive decline. However, the specific impact of hypertension, systolic/diastolic blood pressure, pulse pressure (PP) and mean arterial pressure (MAP) on brain cortical structure remains unclear. Mendelian randomization (MR) provides a robust approach to investigate the causal relationship between blood pressure components and brain cortical changes. METHODS In this MR study, data from large-scale genome-wide association studies for blood pressure components and neuroimaging were utilized to conduct our analyses. We leveraged genetic variants associated specifically with hypertension (122,620 cases and 332,683 controls), systolic (469,767 individuals), diastolic (490,469 individuals) blood pressure, PP (810,865 individuals) and MAP (over 1 million individuals) to evaluate their effects on brain cortex surficial area (51,665 individuals) and cortex thickness (51,665 individuals). RESULTS Our findings revealed a significant correlation between systolic blood pressure and abnormal reduction in brain cortex surficial area (β=-1330.69, 95% confident interval [CI]: -2655.35 to -6.02, p = 0.0489); however, no significant relationship was found between systolic blood pressure and brain cortex thickness (β=-0.0078, 95% CI: -0.0178 to 0.0022, p = 0.1287). Additionally, no significant associations were observed between hypertension (β=-200.05, p = 0.6884; β=-0.0051, p = 0.1179, respectively), diastolic blood pressure (β=-460.63, p = 0.5160; β=0.0047, p = 0.2448, respectively), PP (β=1041.84, p = 0.3725; β=-0.0112, p = 0.2212, respectively), MAP (β=-18.84, p = 0.8841; β=0.0002, p = 0.7654, respectively) and both brain cortex surficial area and brain cortex thickness. CONCLUSION Our MR study provides evidence supporting the hypothesis that systolic blood pressure, rather than diastolic blood pressure, PP or MAP, is associated with abnormal changes in brain cortical structure.
Collapse
Affiliation(s)
- Ziliang Ye
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No. 85 Hedi Road, Nanning, Guangxi 530021, China
| | - Qing Zeng
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No. 85 Hedi Road, Nanning, Guangxi 530021, China
| | - Limeng Ning
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No. 85 Hedi Road, Nanning, Guangxi 530021, China
| | - Wanzhong Huang
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No. 85 Hedi Road, Nanning, Guangxi 530021, China
| | - Qiang Su
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No. 85 Hedi Road, Nanning, Guangxi 530021, China.
| |
Collapse
|
160
|
Miller AP, Gizer IR. Neurogenetic and multi-omic sources of overlap among sensation seeking, alcohol consumption, and alcohol use disorder. Addict Biol 2024; 29:e13365. [PMID: 38380706 PMCID: PMC10882188 DOI: 10.1111/adb.13365] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/26/2023] [Accepted: 11/26/2023] [Indexed: 02/22/2024]
Abstract
Sensation seeking is bidirectionally associated with levels of alcohol consumption in both adult and adolescent samples, and shared neurobiological and genetic influences may in part explain these associations. Links between sensation seeking and alcohol use disorder (AUD) may primarily manifest via increased alcohol consumption rather than through direct effects on increasing problems and consequences. Here the overlap among sensation seeking, alcohol consumption, and AUD was examined using multivariate modelling approaches for genome-wide association study (GWAS) summary statistics in conjunction with neurobiologically informed analyses at multiple levels of investigation. Meta-analytic and genomic structural equation modelling (GenomicSEM) approaches were used to conduct GWAS of sensation seeking, alcohol consumption, and AUD. Resulting summary statistics were used in downstream analyses to examine shared brain tissue enrichment of heritability and genome-wide evidence of overlap (e.g., stratified GenomicSEM, RRHO, genetic correlations with neuroimaging phenotypes), and to identify genomic regions likely contributing to observed genetic overlap across traits (e.g., H-MAGMA and LAVA). Across approaches, results supported shared neurogenetic architecture between sensation seeking and alcohol consumption characterised by overlapping enrichment of genes expressed in midbrain and striatal tissues and variants associated with increased cortical surface area. Alcohol consumption and AUD evidenced overlap in relation to variants associated with decreased frontocortical thickness. Finally, genetic mediation models provided evidence of alcohol consumption mediating associations between sensation seeking and AUD. This study extends previous research by examining critical sources of neurogenetic and multi-omic overlap among sensation seeking, alcohol consumption, and AUD which may underlie observed phenotypic associations.
Collapse
Affiliation(s)
- Alex P. Miller
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Ian R. Gizer
- Department of Psychological SciencesUniversity of MissouriColumbiaMissouriUSA
| |
Collapse
|
161
|
Huang W, Wang Y, Huang W. Mangiferin alleviates 6-OHDA-induced Parkinson's disease by inhibiting AKR1C3 to activate Wnt signaling pathway. Neurosci Lett 2024; 821:137608. [PMID: 38142926 DOI: 10.1016/j.neulet.2023.137608] [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: 08/01/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with a lack of effective treatment options. mangiferin, a bioactive compound derived from mango, has been shown to possess strong neuroprotective properties. In this study, we investigated the neuroprotective effects of mangiferin on PD and its underlying mechanisms using both in vitro and in vivo models of 6-OHDA-induced PD. Additionally, we conducted molecular docking experiments to evaluate the interaction between mangiferin and AKR1C3 and β-catenin. Our results demonstrated that treatment with mangiferin significantly attenuated 6-OHDA-induced cell damage in PC12 cells, reducing intracellular oxidative stress, improving mitochondrial membrane potential, and restoring the expression of tyrosine hydroxylase (TH), a characteristic protein of dopaminergic neurons. Furthermore, mangiferin reduced the accumulation of α-synuclein and inhibited the expression of AKR1C3, thereby activating the Wnt/β-catenin signaling pathway. In vivo studies revealed that mangiferin improved motor dysfunction in 6-OHDA-induced PD mice. Molecular docking analysis confirmed the interaction between mangiferin and AKR1C3 and β-catenin. These findings indicate that mangiferin exerts significant neuroprotective effects in 6-OHDA-induced PD by inhibiting AKR1C3 and activating the Wnt/β-catenin signaling pathway. Therefore, mangiferin may emerge as an innovative therapeutic strategy in the comprehensive treatment regimen of PD patients, providing them with better clinical outcomes and quality of life.
Collapse
Affiliation(s)
- Wanran Huang
- Pharmacy Department, The Second Affiliated Hospital of Wenzhou Medical University (The second Affiliated Hospital &Yuying Children's Hospital), Wenzhou, Zhejiang 325024, China
| | - Yanni Wang
- Pharmacy Department, The Third Affiliated Hospital of Wenzhou Medical University, Ruian People' s Hospital, Wenzhou, Zhejiang 325200, China
| | - Wei Huang
- Pharmacy Department, Ruian Hospital of Traditional Chinese Medicine, Wenzhou, Zhejiang 325200, China.
| |
Collapse
|
162
|
Li LJ, Zhong XX, Tan GZ, Song MX, Li P, Liu ZX, Xiong SC, Yang DQ, Liang ZJ. Investigation of causal relationships between cortical structure and osteoporosis using two-sample Mendelian randomization. Cereb Cortex 2024; 34:bhad529. [PMID: 38216542 DOI: 10.1093/cercor/bhad529] [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: 11/12/2023] [Revised: 12/03/2023] [Accepted: 12/16/2023] [Indexed: 01/14/2024] Open
Abstract
The mutual interaction between bone characteristics and brain had been reported previously, yet whether the cortical structure has any relevance to osteoporosis is questionable. Therefore, we applied a two-sample bidirectional Mendelian randomization analysis to investigate this relationship. We utilized the bone mineral density measurements of femoral neck (n = 32,735) and lumbar spine (n = 28,498) and data on osteoporosis (7300 cases and 358,014 controls). The global surficial area and thickness and 34 specific functional regions of 51,665 patients were screened by magnetic resonance imaging. For the primary estimate, we utilized the inverse-variance weighted method. The Mendelian randomization-Egger intercept test, MR-PRESSO, Cochran's Q test, and "leave-one-out" sensitivity analysis were conducted to assess heterogeneity and pleiotropy. We observed suggestive associations between decreased thickness in the precentral region (OR = 0.034, P = 0.003) and increased chance of having osteoporosis. The results also revealed suggestive causality of decreased bone mineral density in femoral neck to declined total cortical surface area (β = 1400.230 mm2, P = 0.003), as well as the vulnerability to osteoporosis and reduced thickness in the Parstriangularis region (β = -0.006 mm, P = 0.002). Our study supports that the brain and skeleton exhibit bidirectional crosstalk, indicating the presence of a mutual brain-bone interaction.
Collapse
Affiliation(s)
- Long-Jun Li
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Xian-Xing Zhong
- Guangdong Research Institute for Orthopedics and Traumatology of Chinese Medicine, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, PR China
| | - Guo-Zhi Tan
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Ming-Xi Song
- Department of Education and Research, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, PR China
| | - Pian Li
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Zhen-Xin Liu
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Si-Cheng Xiong
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Da-Qi Yang
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Zu-Jian Liang
- Department of Preventive Medicine, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, PR China
| |
Collapse
|
163
|
Nie X, Zhang Q, Wang Y, Liu Z, Xie D, Song Q, Yang C, Yu T, Sun Y. Causal effects of osteoporosis on structural changes in specific brain regions: a Mendelian randomization study. Cereb Cortex 2024; 34:bhad528. [PMID: 38216525 DOI: 10.1093/cercor/bhad528] [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: 11/04/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/14/2024] Open
Abstract
Observational studies have reported that osteoporosis is associated with cortical changes in the brain. However, the inherent limitations of observational studies pose challenges in eliminating confounding factors and establishing causal relationships. And previous observational studies have not reported changes in specific brain regions. By employing Mendelian randomization, we have been able to infer a causal relationship between osteoporosis and a reduction in the surficial area (SA) of the brain cortical. This effect is partially mediated by vascular calcification. We found that osteoporosis significantly decreased the SA of global brain cortical (β = -1587.62 mm2, 95%CI: -2645.94 mm2 to -529.32 mm2, P = 0.003) as well as the paracentral gyrus without global weighted (β = - 19.42 mm2, 95%CI: -28.90 mm2 to -9.95 mm2, P = 5.85 × 10-5). Furthermore, we estimated that 42.25% and 47.21% of the aforementioned effects are mediated through vascular calcification, respectively. Osteoporosis leads to a reduction in the SA of the brain cortical, suggesting the presence of the bone-brain axis. Vascular calcification plays a role in mediating this process to a certain extent. These findings establish a theoretical foundation for further investigations into the intricate interplay between bone, blood vessels, and the brain.
Collapse
Affiliation(s)
- Xinlin Nie
- Department of Orthopedic Center, the First Hospital of Jilin University, Changchun 130000, China
| | - Qiong Zhang
- Department of Orthopedic Center, the First Hospital of Jilin University, Changchun 130000, China
| | - Yixuan Wang
- Department of Otolaryngology Head and Neck Surgery, Shaanxi Provincial People's Hospital, Xi'an 710000, China
| | - Zhaoliang Liu
- Department of Orthopedic Center, the First Hospital of Jilin University, Changchun 130000, China
| | - Dongheng Xie
- Department of Orthopedic Center, the First Hospital of Jilin University, Changchun 130000, China
| | - Qingxu Song
- Department of Orthopedic Center, the First Hospital of Jilin University, Changchun 130000, China
| | - Chen Yang
- Department of Orthopedic Center, the First Hospital of Jilin University, Changchun 130000, China
| | - Tiecheng Yu
- Department of Orthopedic Center, the First Hospital of Jilin University, Changchun 130000, China
| | - Yang Sun
- Department of Orthopedic Center, the First Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
164
|
Zhong Y, Li J, Hong Y, Yang S, Pei L, Chen X, Wu H, Wang T. Resting heart rate causally affects the brain cortical structure: Mendelian randomization study. Cereb Cortex 2024; 34:bhad536. [PMID: 38212288 PMCID: PMC10839837 DOI: 10.1093/cercor/bhad536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024] Open
Abstract
Resting heart rate (RHR) has been linked to impaired cortical structure in observational studies. However, the extent to which this association is potentially causal has not been determined. Using genetic data, this study aimed to reveal the causal effect of RHR on brain cortical structure. A Two-Sample Mendelian randomization (MR) analysis was conducted. Sensitivity analyses, weighted median, MR Pleiotropy residual sum and outlier, and MR-Egger regression were conducted to evaluate heterogeneity and pleiotropy. A causal relationship between RHR and cortical structures was identified by MR analysis. On the global scale, elevated RHR was found to decrease global surface area (SA; P < 0.0125). On a regional scale, the elevated RHR significantly decreased the SA of pars triangularis without global weighted (P = 1.58 × 10-4) and the thickness (TH) of the paracentral with global weighted (P = 3.56 × 10-5), whereas it increased the TH of banks of the superior temporal sulcus in the presence of global weighted (P = 1.04 × 10-4). MR study provided evidence that RHR might be causally linked to brain cortical structure, which offers a different way to understand the heart-brain axis theory.
Collapse
Affiliation(s)
- Yinsheng Zhong
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518003, P. R. China
| | - Jun Li
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518003, P. R. China
| | - Yinghui Hong
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518003, P. R. China
| | - Shujun Yang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518003, P. R. China
| | - Liying Pei
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518003, P. R. China
| | - Xuxiang Chen
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518003, P. R. China
| | - Haidong Wu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518003, P. R. China
| | - Tong Wang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518003, P. R. China
| |
Collapse
|
165
|
Liang X, Sun L, Liao X, Lei T, Xia M, Duan D, Zeng Z, Li Q, Xu Z, Men W, Wang Y, Tan S, Gao JH, Qin S, Tao S, Dong Q, Zhao T, He Y. Structural connectome architecture shapes the maturation of cortical morphology from childhood to adolescence. Nat Commun 2024; 15:784. [PMID: 38278807 PMCID: PMC10817914 DOI: 10.1038/s41467-024-44863-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 01/08/2024] [Indexed: 01/28/2024] Open
Abstract
Cortical thinning is an important hallmark of the maturation of brain morphology during childhood and adolescence. However, the connectome-based wiring mechanism that underlies cortical maturation remains unclear. Here, we show cortical thinning patterns primarily located in the lateral frontal and parietal heteromodal nodes during childhood and adolescence, which are structurally constrained by white matter network architecture and are particularly represented using a network-based diffusion model. Furthermore, connectome-based constraints are regionally heterogeneous, with the largest constraints residing in frontoparietal nodes, and are associated with gene expression signatures of microstructural neurodevelopmental events. These results are highly reproducible in another independent dataset. These findings advance our understanding of network-level mechanisms and the associated genetic basis that underlies the maturational process of cortical morphology during childhood and adolescence.
Collapse
Affiliation(s)
- Xinyuan Liang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Lianglong Sun
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Xuhong Liao
- School of Systems Science, Beijing Normal University, Beijing, 100875, China
| | - Tianyuan Lei
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Mingrui Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Dingna Duan
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Zilong Zeng
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Qiongling Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Zhilei Xu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Weiwei Men
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Beijing City Key Laboratory for Medical Physics and Engineering, Institute of Heavy Ion Physics, School of Physics, Peking University, Beijing, 100871, China
| | - Yanpei Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Shuping Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing, 100096, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Beijing City Key Laboratory for Medical Physics and Engineering, Institute of Heavy Ion Physics, School of Physics, Peking University, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Shaozheng Qin
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Sha Tao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Qi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Tengda Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China.
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China.
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China.
| | - Yong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China.
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100875, China.
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| |
Collapse
|
166
|
Li X, Jiang M, Zhao L, Yang K, Lu T, Zhang D, Li J, Wang L. Relationship between autism and brain cortex surface area: genetic correlation and a two-sample Mendelian randomization study. BMC Psychiatry 2024; 24:69. [PMID: 38263034 PMCID: PMC10807092 DOI: 10.1186/s12888-024-05514-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/10/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Alterations in surface area (SA) in specific regions of the cortex have been reported in many individuals with autism spectrum disorder (ASD), however, the genetic background between ASD and SA is still unclear. This study estimated the genetic correlation and causal effect of ASD and cortical SA. METHODS Summarized data of genome-wide association studies (GWAS) were separately downloaded from the Psychiatric Genomics Consortium (18,381 cases of ASD, and 27,969 controls) and the Enhancing Neuroimaging Genetics through Meta-Analysis Consortium (33,992 participants of Europeans). We used Linkage disequilibrium score regression (LDSC) and Heritability Estimation from Summary Statistics (HESS) to calculate the heritability of each trait. As for the genetic correlation between ASD and SA, LDSC was used for global correlation and HESS was used to examine the local genetic covariance further. We used three Mendelian randomization (MR) methods, Inverse-variance weighted, MR-Egger, and weighted median to estimate the causal relationship. RESULTS LDSC observed a nominal significant genetic correlation (rg = 0.1229, P-value = 0.0346) between ASD and SA of the rostral anterior cingulate gyrus whereas analysis through HESS did not reveal any significant loci having genetic covariance. Based on MR results, statistically meaningful estimations were found in the following areas, postcentral cortex (β (SE) = 21.82 (7.84) mm, 95% CI: 6.46 to 37.19 mm, PIVW = 5.38 × 10- 3, PFDR = 3.09 × 10- 2), posterior cingulate gyrus (β (SE) = 6.23 (2.69) mm, 95% CI: 0.96 to 11.49 mm, PIVW = 2.05 × 10- 2, PFDR = 4.26 × 10- 2), supramarginal gyrus (β (SE) = 19.25 (8.43) mm, 95% CI: 29.29 to 35.77 mm, PIVW = 2.24 × 10- 2, PFDR = 4.31 × 10- 2). CONCLUSION Our results provided genetic evidence to support the opinion that individuals with ASD tend to develop differences in cortical SA of special areas. The findings contributed to understanding the genetic relationship between ASD and cortical SA.
Collapse
Grants
- 2019B030335001 Key-Area Research and Development Program of Guangdong Province
- 2019B030335001 Key-Area Research and Development Program of Guangdong Province
- 2019B030335001 Key-Area Research and Development Program of Guangdong Province
- 2019B030335001 Key-Area Research and Development Program of Guangdong Province
- 2019B030335001 Key-Area Research and Development Program of Guangdong Province
- 2019B030335001 Key-Area Research and Development Program of Guangdong Province
- 2019B030335001 Key-Area Research and Development Program of Guangdong Province
- 2019B030335001 Key-Area Research and Development Program of Guangdong Province
- 81971283, 82171537, 82071541, 81671363, and 81730037 National Natural Science Foundation of China
- 81971283, 82171537, 82071541, 81671363, and 81730037 National Natural Science Foundation of China
- 81971283, 82171537, 82071541, 81671363, and 81730037 National Natural Science Foundation of China
- 81971283, 82171537, 82071541, 81671363, and 81730037 National Natural Science Foundation of China
- 81971283, 82171537, 82071541, 81671363, and 81730037 National Natural Science Foundation of China
- 81971283, 82171537, 82071541, 81671363, and 81730037 National Natural Science Foundation of China
- 81971283, 82171537, 82071541, 81671363, and 81730037 National Natural Science Foundation of China
- 81971283, 82171537, 82071541, 81671363, and 81730037 National Natural Science Foundation of China
Collapse
Affiliation(s)
- Xianjing Li
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital, Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Miaomiao Jiang
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital, Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Liyang Zhao
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital, Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Kang Yang
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital, Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Tianlan Lu
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital, Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Dai Zhang
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital, Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- Guangdong Key Laboratory of Mental Health and Cognitive Science, Institute for Brain Research and Rehabilitation (IBRR), South China Normal University, Guangzhou, China
| | - Jun Li
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital, Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China.
| | - Lifang Wang
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital, Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China.
| |
Collapse
|
167
|
Wang M, Wang Z, Yu Y, Zhao D, Shen Z, Wei F. From teeth to brain: dental caries causally affects the cortical thickness of the banks of the superior temporal sulcus. BMC Oral Health 2024; 24:124. [PMID: 38263072 PMCID: PMC10807149 DOI: 10.1186/s12903-024-03899-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/15/2024] [Indexed: 01/25/2024] Open
Abstract
OBJECTIVES Dental caries is one of the most prevalent oral diseases and causes of tooth loss. Cross-sectional studies observed epidemiological associations between dental caries and brain degeneration disorders, while it is unknown whether dental caries causally affect the cerebral structures. This study tested whether genetically proxied DMFS (the sum of Decayed, Missing, and Filled tooth Surfaces) causally impacts the brain cortical structure using Mendelian randomization (MR). METHODS The summary-level GWAS meta-analysis data from the GLIDE consortium were used for DMFS, including 26,792 participants. ENIGMA (Enhancing NeuroImaging Genetics through Meta Analysis) consortium GWAS summary data of 51,665 patients were used for brain structure. This study estimated the causal effects of DMFS on the surface area (SA) and thickness (TH) of the global cortex and functional cortical regions accessed by magnetic resonance imaging (MRI). Inverse-variance weighted (IVW) was used as the primary estimate, the MR pleiotropy residual sum and outlier (MR-PRESSO), the MR-Egger intercept test, and leave-one-out analyses were used to examine the potential horizontal pleiotropy. RESULTS Genetically proxied DMFS decreases the TH of the banks of the superior temporal sulcus (BANSSTS) with or without global weighted (weighted, β = - 0.0277 mm, 95% CI: - 0.0470 mm to - 0.0085 mm, P = 0.0047; unweighted, β = - 0.0311 mm, 95% CI: - 0.0609 mm to - 0.0012 mm, P = 0.0412). The causal associations were robust in various sensitivity analyses. CONCLUSIONS Dental caries causally decrease the cerebral cortical thickness of the BANKSSTS, a cerebral cortical region crucial for language-related functions, and is the most affected brain region in Alzheimer's disease. This investigation provides the first evidence that dental caries causally affects brain structure, proving the existence of teeth-brain axes. This study also suggested that clinicians should highlight the causal effects of dental caries on brain disorders during the diagnosis and treatments, the cortical thickness of BANKSSTS is a promising diagnostic measurement for dental caries-related brain degeneration.
Collapse
Affiliation(s)
- Mengqiao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Ziyao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Yajie Yu
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, 100191, China
| | - Delu Zhao
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Zhiyuan Shen
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| |
Collapse
|
168
|
Sun D, Wang R, Du Q, Zhang Y, Chen H, Shi Z, Wang X, Zhou H. Causal relationship between multiple sclerosis and cortical structure: a Mendelian randomization study. J Transl Med 2024; 22:83. [PMID: 38245759 PMCID: PMC10800041 DOI: 10.1186/s12967-024-04892-7] [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: 06/29/2023] [Accepted: 01/13/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Observational studies have suggested an association between multiple sclerosis (MS) and cortical structure, but the results have been inconsistent. OBJECTIVE We used two-sample Mendelian randomization (MR) to assess the causal relationship between MS and cortical structure. METHODS MS data as the exposure trait, including 14,498 cases and 24,091 controls, were obtained from the International Multiple Sclerosis Genetics Consortium. Genome-wide association study (GWAS) data for cortical surface area (SAw/nw) and thickness (THw/nw) in 51,665 individuals of European ancestry were obtained from the ENIGMA Consortium. The inverse-variance weighted (IVW) method was used as the primary analysis for MR. Sensitivity analyses were conducted to evaluate heterogeneity and pleiotropy. Enrichment analysis was performed on MR analyses filtered by sensitivity analysis. RESULTS After IVW and sensitivity analysis filtering, only six surviving MR results provided suggestive evidence supporting a causal relationship between MS and cortical structure, including lingual SAw (p = .0342, beta (se) = 5.7127 (2.6969)), parahippocampal SAw (p = .0224, beta (se) = 1.5577 (0.6822)), rostral middle frontal SAw (p = .0154, beta (se) = - 9.0301 (3.7281)), cuneus THw (p = .0418, beta (se) = - 0.0020 (0.0010)), lateral orbitofrontal THw (p = .0281, beta (se) = 0.0025 (0.0010)), and lateral orbitofrontal THnw (p = .0417, beta (se) = 0.0029 (0.0014)). Enrichment analysis suggested that leukocyte cell-related pathways, JAK-STAT signaling pathway, NF-kappa B signaling pathway, cytokine-cytokine receptor interaction, and prolactin signaling pathway may be involved in the effect of MS on cortical morphology. CONCLUSION Our results provide evidence supporting a causal relationship between MS and cortical structure. Enrichment analysis suggests that the pathways mediating brain morphology abnormalities in MS patients are mainly related to immune and inflammation-driven pathways.
Collapse
Affiliation(s)
- Dongren Sun
- Department of Neurology, West China Hospital, Sichuan University, Guo Xuexiang #37, Chengdu, 610041, China
| | - Rui Wang
- Department of Neurology, West China Hospital, Sichuan University, Guo Xuexiang #37, Chengdu, 610041, China
| | - Qin Du
- Department of Neurology, West China Hospital, Sichuan University, Guo Xuexiang #37, Chengdu, 610041, China
| | - Ying Zhang
- Department of Neurology, West China Hospital, Sichuan University, Guo Xuexiang #37, Chengdu, 610041, China
| | - Hongxi Chen
- Department of Neurology, West China Hospital, Sichuan University, Guo Xuexiang #37, Chengdu, 610041, China
| | - Ziyan Shi
- Department of Neurology, West China Hospital, Sichuan University, Guo Xuexiang #37, Chengdu, 610041, China
| | - Xiaofei Wang
- Department of Neurology, West China Hospital, Sichuan University, Guo Xuexiang #37, Chengdu, 610041, China.
| | - Hongyu Zhou
- Department of Neurology, West China Hospital, Sichuan University, Guo Xuexiang #37, Chengdu, 610041, China.
| |
Collapse
|
169
|
Cattarinussi G, Pouya P, Grimaldi DA, Dini MZ, Sambataro F, Brambilla P, Delvecchio G. Cortical alterations in relatives of patients with bipolar disorder: A review of magnetic resonance imaging studies. J Affect Disord 2024; 345:234-243. [PMID: 37865341 DOI: 10.1016/j.jad.2023.10.097] [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: 05/29/2023] [Revised: 09/11/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023]
Abstract
INTRODUCTION Bipolar disorder (BD) is a severe mental disorder characterized by high heritability rates. Widespread brain cortical alterations have been reported in BD patients, mostly involving the frontal, temporal and parietal regions. Importantly, also unaffected relatives of BD patients (BD-RELs) present abnormalities in cortical measures, which are not influenced by disease-related factors, such as medication use and illness duration. Here, we collected all available evidence on cortical measures in BD-RELs to further our knowledge on the potential cortical alterations associated with the vulnerability and the resilience to BD. METHODS A search on PubMed, Web of Science and Scopus was performed to identify neuroimaging studies exploring cortical alterations in BD-RELs, including cortical thickness (CT), surface area (SA), gyrification (GI) and cortical complexity. Eleven studies were included. Of these, five assessed CT, five examined CT and SA and one explored CT, SA and GI. RESULTS Overall, a heterogeneous pattern of cortical alterations emerged. The areas more consistently linked with genetic liability for BD were the prefrontal and sensorimotor regions. Mixed evidence was reported in the temporal and cingulate areas. LIMITATIONS The small sample size and the heterogeneity in terms of methodologies and the characteristics of the participants limit the generalizability of our results. CONCLUSIONS Our findings suggest that the genetic liability for BD is related to reduced CT in the prefrontal cortex, which might be a marker of risk for BD, and increased CT within the sensorimotor cortex, which could represent a marker of resilience.
Collapse
Affiliation(s)
- Giulia Cattarinussi
- Department of Neuroscience (DNS), Padua Neuroscience Center, University of Padova, Padua, Italy; Padua Neuroscience Center, University of Padova, Padua, Italy
| | - Parnia Pouya
- Research Center for Evidence-Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Iranian EBM Center: A Joanna Briggs Institute Affiliated Group, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahta Zare Dini
- Research Center for Evidence-Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Iranian EBM Center: A Joanna Briggs Institute Affiliated Group, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fabio Sambataro
- Department of Neuroscience (DNS), Padua Neuroscience Center, University of Padova, Padua, Italy; Padua Neuroscience Center, University of Padova, Padua, Italy
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giuseppe Delvecchio
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| |
Collapse
|
170
|
Oblong LM, Soheili‐Nezhad S, Trevisan N, Shi Y, Beckmann CF, Sprooten E. Principal and independent genomic components of brain structure and function. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12876. [PMID: 38225802 PMCID: PMC10797248 DOI: 10.1111/gbb.12876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 01/17/2024]
Abstract
The highly polygenic and pleiotropic nature of behavioural traits, psychiatric disorders and structural and functional brain phenotypes complicate mechanistic interpretation of related genome-wide association study (GWAS) signals, thereby obscuring underlying causal biological processes. We propose genomic principal and independent component analysis (PCA, ICA) to decompose a large set of univariate GWAS statistics of multimodal brain traits into more interpretable latent genomic components. Here we introduce and evaluate this novel methods various analytic parameters and reproducibility across independent samples. Two UK Biobank GWAS summary statistic releases of 2240 imaging-derived phenotypes (IDPs) were retrieved. Genome-wide beta-values and their corresponding standard-error scaled z-values were decomposed using genomic PCA/ICA. We evaluated variance explained at multiple dimensions up to 200. We tested the inter-sample reproducibility of output of dimensions 5, 10, 25 and 50. Reproducibility statistics of the respective univariate GWAS served as benchmarks. Reproducibility of 10-dimensional PCs and ICs showed the best trade-off between model complexity and robustness and variance explained (PCs: |rz - max| = 0.33, |rraw - max| = 0.30; ICs: |rz - max| = 0.23, |rraw - max| = 0.19). Genomic PC and IC reproducibility improved substantially relative to mean univariate GWAS reproducibility up to dimension 10. Genomic components clustered along neuroimaging modalities. Our results indicate that genomic PCA and ICA decompose genetic effects on IDPs from GWAS statistics with high reproducibility by taking advantage of the inherent pleiotropic patterns. These findings encourage further applications of genomic PCA and ICA as fully data-driven methods to effectively reduce the dimensionality, enhance the signal to noise ratio and improve interpretability of high-dimensional multitrait genome-wide analyses.
Collapse
Affiliation(s)
- Lennart M. Oblong
- Department of Cognitive NeuroscienceDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CentreNijmegenThe Netherlands
| | - Sourena Soheili‐Nezhad
- Department of Cognitive NeuroscienceDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CentreNijmegenThe Netherlands
- Language and Genetics DepartmentMax Planck Institute for PsycholinguisticsNijmegenThe Netherlands
| | - Nicolò Trevisan
- Department of Cognitive NeuroscienceDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CentreNijmegenThe Netherlands
| | - Yingjie Shi
- Department of Cognitive NeuroscienceDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CentreNijmegenThe Netherlands
- Department of Human GeneticsDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CentreNijmegenThe Netherlands
| | - Christian F. Beckmann
- Department of Cognitive NeuroscienceDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CentreNijmegenThe Netherlands
- Centre for Cognitive NeuroimagingDonders Institute for Brain, Cognition and Behaviour, Radboud UniversityNijmegenThe Netherlands
| | - Emma Sprooten
- Department of Cognitive NeuroscienceDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CentreNijmegenThe Netherlands
- Department of Human GeneticsDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CentreNijmegenThe Netherlands
| |
Collapse
|
171
|
Lin Y, Wang S, Zhang L, Yang Q. Elucidating the relationship between breast cancer and brain cortical structure: a Mendelian randomization study. Cereb Cortex 2024; 34:bhad498. [PMID: 38112592 DOI: 10.1093/cercor/bhad498] [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: 11/08/2023] [Revised: 12/02/2023] [Accepted: 12/03/2023] [Indexed: 12/21/2023] Open
Abstract
Cancer-associated cognitive impairment is a significant challenge for individuals who have survived breast cancer, affecting their quality of life. In this study, we conducted an inaugural comprehensive Mendelian randomization analysis discerning the causal relationship between breast cancer, including its two subtypes, and the cerebral cortical structure. Our analysis indicated that estrogen receptor-negative breast cancer significantly decreased surface area (β = -593.01 mm2, 95% CI: -1134.9 to -51.1 mm2, P = 0.032). At the regional level, estrogen receptor-negative breast cancer showed a significant association with surface area and thickness in 17 cortical regions. These regions included the insula, posterior cingulate, superior frontal, precuneus, fusiform, lateral occipital, and rostral middle frontal. Specifically, estrogen receptor-negative breast cancer had a significant impact on decreasing the surface area of the insula without considering global weight (β = -14.09 mm2, 95% CI: -22.91 to -5.27 mm2, P = 0.0017). The results from meta-analysis and LD Score Regression provide support for our findings. This investigation unveils the correlations between breast cancer, its various subcategories, and the cerebral cortical structure. Notably, breast cancer of the estrogen receptor-negative variety may elicit more widespread cerebral atrophy.
Collapse
Affiliation(s)
- Yilong Lin
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361003, China
| | - Songsong Wang
- Department of Urology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Liyi Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361003, China
| | - Qingmo Yang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361003, China
| |
Collapse
|
172
|
Lin YK, Cai XR, Chen JZ, Hong HJ, Tu K, Chen YL, Du Q. Non-alcoholic fatty liver disease causally affects the brain cortical structure: a Mendelian randomization study. Front Neurosci 2024; 17:1305624. [PMID: 38260009 PMCID: PMC10800802 DOI: 10.3389/fnins.2023.1305624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
Background Reduced brain volume, impaired cognition, and possibly a range of psychoneurological disorders have been reported in patients with non-alcoholic fatty liver disease (NAFLD); however, no underlying cause has been specified. Here, Mendelian randomization (MR) was employed to determine the causative NAFLD effects on cortical structure. Methods We used pooled-level data from FinnGen's published genome-wide association study (GWAS) of NAFLD (1908 cases and 340,591 healthy controls), as well as published GWAS with NAFLD activity score (NAS) and fibrosis stage-associated SNPs as genetic tools, in addition to the Enigma Consortium data from 51,665 patients, were used to assess genetic susceptibility in relation to changes with cortical thickness (TH) and surface area (SA). A main estimate was made by means of inverse variance weighted (IVW), while heterogeneity and pleiotropy were detected using MR-Egger, weighted median, and MR Pleiotropy RESidual Sum and Outlier to perform a two-sample MR analysis. Results At the global level, NAFLD reduced SA (beta = -586.72 mm2, se = 217.73, p = 0.007) and several changes in the cortical structure of the cerebral gyrus were found, with no detectable pleiotropy. Conclusion NAFLD causally affects cortical structures, which supports the presence of an intricate liver-brain axis.
Collapse
Affiliation(s)
- Yu-Kai Lin
- Department of Hepatological Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Medical University Cancer Center, Fuzhou, China
| | - Xin-Ran Cai
- Department of Hepatological Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Medical University Cancer Center, Fuzhou, China
| | - Jiang-Zhi Chen
- Department of Hepatological Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Medical University Cancer Center, Fuzhou, China
| | - Hai-Jie Hong
- Department of Hepatological Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Medical University Cancer Center, Fuzhou, China
| | - Kai Tu
- Department of Hepatological Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Medical University Cancer Center, Fuzhou, China
| | - Yan-Ling Chen
- Department of Hepatological Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Medical University Cancer Center, Fuzhou, China
| | - Qiang Du
- Department of Hepatological Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Medical University Cancer Center, Fuzhou, China
| |
Collapse
|
173
|
Faouzi J, Tan M, Casse F, Lesage S, Tesson C, Brice A, Mangone G, Mariani LL, Iwaki H, Colliot O, Pihlstrøm L, Corvol JC. Proxy-analysis of the genetics of cognitive decline in Parkinson's disease through polygenic scores. NPJ Parkinsons Dis 2024; 10:8. [PMID: 38177146 PMCID: PMC10767119 DOI: 10.1038/s41531-023-00619-5] [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: 02/03/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Cognitive decline is common in Parkinson's disease (PD) and its genetic risk factors are not well known to date, besides variants in the GBA and APOE genes. However, variation in complex traits is caused by numerous variants and is usually studied with genome-wide association studies (GWAS), requiring a large sample size, which is difficult to achieve for outcome measures in PD. Taking an alternative approach, we computed 100 polygenic scores (PGS) related to cognitive, dementia, stroke, and brain anatomical phenotypes and investigated their association with cognitive decline in six longitudinal cohorts. The analysis was adjusted for age, sex, genetic ancestry, follow-up duration, GBA and APOE status. Then, we meta-analyzed five of these cohorts, comprising a total of 1702 PD participants with 6156 visits, using the Montreal Cognitive Assessment as a cognitive outcome measure. After correction for multiple comparisons, we found four PGS significantly associated with cognitive decline: intelligence (p = 5.26e-13), cognitive performance (p = 1.46e-12), educational attainment (p = 8.52e-10), and reasoning (p = 3.58e-5). Survival analyses highlighted an offset of several years between the first and last quartiles of PGS, with significant differences for the PGS of cognitive performance (5 years) and educational attainment (7 years). In conclusion, we found four PGS associated with cognitive decline in PD, all associated with general cognitive phenotypes. This study highlights the common genetic factors between cognitive decline in PD and the general population, and the importance of the participant's cognitive reserve for cognitive outcome in PD.
Collapse
Affiliation(s)
- Johann Faouzi
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, F-75013, Paris, France
- Univ Rennes, Ensai, CNRS, CREST-UMR 9194, F-35000, Rennes, France
| | - Manuela Tan
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Fanny Casse
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Suzanne Lesage
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Christelle Tesson
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Alexis Brice
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Département de Génétique, F-75013, Paris, France
| | - Graziella Mangone
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Département de Neurologie, F-75013, Paris, France
- Department of Neurology, Movement Disorder Division, Rush University Medical Center, 1725 W. Harrison Street, Chicago, IL, 60612, USA
| | - Louise-Laure Mariani
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Département de Neurologie, F-75013, Paris, France
| | - Hirotaka Iwaki
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International LLC, Washington, DC, USA
| | - Olivier Colliot
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, F-75013, Paris, France
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Jean-Christophe Corvol
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Département de Neurologie, F-75013, Paris, France.
| |
Collapse
|
174
|
Luppi AI, Girn M, Rosas FE, Timmermann C, Roseman L, Erritzoe D, Nutt DJ, Stamatakis EA, Spreng RN, Xing L, Huttner WB, Carhart-Harris RL. A role for the serotonin 2A receptor in the expansion and functioning of human transmodal cortex. Brain 2024; 147:56-80. [PMID: 37703310 DOI: 10.1093/brain/awad311] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
Integrating independent but converging lines of research on brain function and neurodevelopment across scales, this article proposes that serotonin 2A receptor (5-HT2AR) signalling is an evolutionary and developmental driver and potent modulator of the macroscale functional organization of the human cerebral cortex. A wealth of evidence indicates that the anatomical and functional organization of the cortex follows a unimodal-to-transmodal gradient. Situated at the apex of this processing hierarchy-where it plays a central role in the integrative processes underpinning complex, human-defining cognition-the transmodal cortex has disproportionately expanded across human development and evolution. Notably, the adult human transmodal cortex is especially rich in 5-HT2AR expression and recent evidence suggests that, during early brain development, 5-HT2AR signalling on neural progenitor cells stimulates their proliferation-a critical process for evolutionarily-relevant cortical expansion. Drawing on multimodal neuroimaging and cross-species investigations, we argue that, by contributing to the expansion of the human cortex and being prevalent at the apex of its hierarchy in the adult brain, 5-HT2AR signalling plays a major role in both human cortical expansion and functioning. Owing to its unique excitatory and downstream cellular effects, neuronal 5-HT2AR agonism promotes neuroplasticity, learning and cognitive and psychological flexibility in a context-(hyper)sensitive manner with therapeutic potential. Overall, we delineate a dual role of 5-HT2ARs in enabling both the expansion and modulation of the human transmodal cortex.
Collapse
Affiliation(s)
- Andrea I Luppi
- Department of Clinical Neurosciences and Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
- Leverhulme Centre for the Future of Intelligence, University of Cambridge, Cambridge, CB2 1SB, UK
- The Alan Turing Institute, London, NW1 2DB, UK
| | - Manesh Girn
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
- Psychedelics Division-Neuroscape, Department of Neurology, University of California SanFrancisco, San Francisco, CA 94158, USA
| | - Fernando E Rosas
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
- Data Science Institute, Imperial College London, London, SW7 2AZ, UK
- Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK
| | - Christopher Timmermann
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Leor Roseman
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - David Erritzoe
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - David J Nutt
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Emmanuel A Stamatakis
- Department of Clinical Neurosciences and Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - R Nathan Spreng
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
| | - Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Robin L Carhart-Harris
- Psychedelics Division-Neuroscape, Department of Neurology, University of California SanFrancisco, San Francisco, CA 94158, USA
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
175
|
Abstract
Brain development in humans is achieved through precise spatiotemporal genetic control, the mechanisms of which remain largely elusive. Recently, integration of technological advances in human stem cell-based modelling with genome editing has emerged as a powerful platform to establish causative links between genotypes and phenotypes directly in the human system. Here, we review our current knowledge of complex genetic regulation of each key step of human brain development through the lens of evolutionary specialization and neurodevelopmental disorders and highlight the use of human stem cell-derived 2D cultures and 3D brain organoids to investigate human-enriched features and disease mechanisms. We also discuss opportunities and challenges of integrating new technologies to reveal the genetic architecture of human brain development and disorders.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
176
|
Paus T. Population Neuroscience: Principles and Advances. Curr Top Behav Neurosci 2024; 68:3-34. [PMID: 38589637 DOI: 10.1007/7854_2024_474] [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] [Indexed: 04/10/2024]
Abstract
In population neuroscience, three disciplines come together to advance our knowledge of factors that shape the human brain: neuroscience, genetics, and epidemiology (Paus, Human Brain Mapping 31:891-903, 2010). Here, I will come back to some of the background material reviewed in more detail in our previous book (Paus, Population Neuroscience, 2013), followed by a brief overview of current advances and challenges faced by this integrative approach.
Collapse
Affiliation(s)
- Tomáš Paus
- Department of Psychiatry and Neuroscience, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
177
|
Momoi MY. Overview: Research on the Genetic Architecture of the Developing Cerebral Cortex in Norms and Diseases. Methods Mol Biol 2024; 2794:1-12. [PMID: 38630215 DOI: 10.1007/978-1-0716-3810-1_1] [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] [Indexed: 04/19/2024]
Abstract
The human brain is characterized by high cell numbers, diverse cell types with diverse functions, and intricate connectivity with an exceedingly broad surface of the cortex. Human-specific brain development was accomplished by a long timeline for maturation from the prenatal period to the third decade of life. The long timeline makes complicated architecture and circuits of human cerebral cortex possible, and it makes human brain vulnerable to intrinsic and extrinsic insults resulting in the development of variety of neuropsychiatric disorders. Unraveling the molecular and cellular processes underlying human brain development under the elaborate regulation of gene expression in a spatiotemporally specific manner, especially that of the cortex will provide a biological understanding of human cognition and behavior in health and diseases. Global research consortia and the advancing technologies in brain science including functional genomics equipped with emergent neuroinformatics such as single-cell multiomics, novel human models, and high-volume databases with high-throughput computation facilitate the biological understanding of the development of the human brain cortex. Knowing the process of interplay of the genome and the environment in cortex development will lead us to understand the human-specific cognitive function and its individual diversity. Thus, it is worthwhile to overview the recent progress in neurotechnology to foresee further understanding of the human brain and norms and diseases.
Collapse
Affiliation(s)
- Mariko Y Momoi
- Ryomo Seishi Ryogoen Rehabilitation Hospital for Children with Disabilities, Gunma, Japan
| |
Collapse
|
178
|
Ge YJ, Wu BS, Zhang Y, Chen SD, Zhang YR, Kang JJ, Deng YT, Ou YN, He XY, Zhao YL, Kuo K, Ma Q, Banaschewski T, Barker GJ, Bokde ALW, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Martinot MLP, Artiges E, Nees F, Orfanos DP, Lemaitre H, Paus T, Poustka L, Hohmann S, Millenet S, Fröhner JH, Smolka MN, Vaidya N, Walter H, Whelan R, Feng JF, Tan L, Dong Q, Schumann G, Cheng W, Yu JT. Genetic architectures of cerebral ventricles and their overlap with neuropsychiatric traits. Nat Hum Behav 2024; 8:164-180. [PMID: 37857874 DOI: 10.1038/s41562-023-01722-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 09/12/2023] [Indexed: 10/21/2023]
Abstract
The cerebral ventricles are recognized as windows into brain development and disease, yet their genetic architectures, underlying neural mechanisms and utility in maintaining brain health remain elusive. Here we aggregated genetic and neuroimaging data from 61,974 participants (age range, 9 to 98 years) in five cohorts to elucidate the genetic basis of ventricular morphology and examined their overlap with neuropsychiatric traits. Genome-wide association analysis in a discovery sample of 31,880 individuals identified 62 unique loci and 785 candidate genes associated with ventricular morphology. We replicated over 80% of loci in a well-matched cohort of lateral ventricular volume. Gene set analysis revealed enrichment of ventricular-trait-associated genes in biological processes and disease pathogenesis during both early brain development and degeneration. We explored the age-dependent genetic associations in cohorts of different age groups to investigate the possible roles of ventricular-trait-associated loci in neurodevelopmental and neurodegenerative processes. We describe the genetic overlap between ventricular and neuropsychiatric traits through comprehensive integrative approaches under correlative and causal assumptions. We propose the volume of the inferior lateral ventricles as a heritable endophenotype to predict the risk of Alzheimer's disease, which might be a consequence of prodromal Alzheimer's disease. Our study provides an advance in understanding the genetics of the cerebral ventricles and demonstrates the potential utility of ventricular measurements in tracking brain disorders and maintaining brain health across the lifespan.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Yi 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
| | - 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
| | - 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
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, 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, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xiao-Yu He
- 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
| | - Yong-Li Zhao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Kevin Kuo
- 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
| | - Qing Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gareth J Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, USA
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, UK
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt, Braunschweig and Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U 1299 'Trajectoires développementales & psychiatrie', University Paris-Saclay, CNRS; Ecole Normale Supérieure Paris-Saclay, Centre Borelli, Gif-sur-Yvette, France
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U 1299 'Trajectoires développementales & psychiatrie', University Paris-Saclay, CNRS; Ecole Normale Supérieure Paris-Saclay, Centre Borelli, Gif-sur-Yvette, France
- AP-HP, Sorbonne University, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, Paris, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U 1299 'Trajectoires développementales & psychiatrie', University Paris-Saclay, CNRS; Ecole Normale Supérieure Paris-Saclay, Centre Borelli, Gif-sur-Yvette, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig Holstein, Kiel University, Kiel, Germany
| | | | - Herve Lemaitre
- NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, Bordeaux, France
| | - Tomáš Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
- Departments of Psychiatry and Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, Göttingen, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sabina Millenet
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Juliane H Fröhner
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Stratified Medicine, Department of Psychiatry and Neuroscience, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - 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, Beijing, 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
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 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, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine, Department of Psychiatry and Neuroscience, Charité Universitätsmedizin Berlin, Berlin, Germany
- Centre for Population Neuroscience and Precision Medicine, Institute for Science and Technology of Brain-Inspired Intelligence, 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, Beijing, 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
|
179
|
Chai YH, Han YP, Zhang JY, Zhou JB. Diabetic Retinopathy and Brain Structure, Cognition Function, and Dementia: A Bidirectional Mendelian Randomization Study. J Alzheimers Dis 2024; 97:1211-1221. [PMID: 38217603 DOI: 10.3233/jad-231022] [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] [Indexed: 01/15/2024]
Abstract
BACKGROUND Accumulating evidence has demonstrated that hyperglycemia is a possible risk factor for mild cognitive impairment or Alzheimer's disease. Diabetic retinopathy (DR) has been identified as a risk factor for dementia in patients with diabetes. OBJECTIVE This study aimed to investigate the causal relationships between DR and brain structure, cognitive function, and dementia. METHODS We performed bidirectional two-sample Mendelian randomization for DR, brain structure, cognitive function, and dementia using the inverse-variance weighted method. RESULTS Inverse-variance weighted analysis showed the association of DR with vascular dementia (OR = 1.68, 95% CI: 1.01-2.82), and dementia was significantly associated with the increased risk of non-proliferative DR (NPDR) (OR = 1.76, 95% CI: 1.04-2.98). Furthermore, better cognitive performance was significantly associated with a reduced risk of NPDR (OR = 0.85, 95% CI: 0.74-0.98). No association was observed between DR and brain structure. CONCLUSIONS These findings suggest that the association of DR with vascular dementia. The reciprocal effect of cognitive performance and dementia on NPDR risk highlights the potential benefits of dementia prevention for reducing the burden of DR.
Collapse
Affiliation(s)
- Yin-He Chai
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yi-Peng Han
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jin-Yan Zhang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jian-Bo Zhou
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
180
|
Zhou M, Chen S, Chen Y, Wang C, Chen C. Causal associations between gut microbiota and regional cortical structure: a Mendelian randomization study. Front Neurosci 2023; 17:1296145. [PMID: 38196849 PMCID: PMC10774226 DOI: 10.3389/fnins.2023.1296145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/05/2023] [Indexed: 01/11/2024] Open
Abstract
Introduction Observational studies have reported associations between gut microbiota composition and central nervous system diseases. However, the potential causal relationships and underlying mechanisms remain unclear. Here, we applied Mendelian randomization (MR) to investigate the causal effects of gut microbiota on cortical surface area (SA) and thickness (TH) in the brain. Methods We used genome-wide association study summary statistics of gut microbiota abundance in 18,340 individuals from the MiBioGen Consortium to identify genetic instruments for 196 gut microbial taxa. We then analyzed data from 56,761 individuals from the ENIGMA Consortium to examine associations of genetically predicted gut microbiota with alterations in cortical SA and TH globally and across 34 functional brain regions. Inverse-variance weighted analysis was used as the primary MR method, with MR Egger regression, MR-PRESSO, Cochran's Q test, and leave-one-out analysis to assess heterogeneity and pleiotropy. Results At the functional region level, genetically predicted higher abundance of class Mollicutes was associated with greater SA of the medial orbitofrontal cortex (β = 8.39 mm2, 95% CI: 3.08-13.70 mm2, p = 0.002), as was higher abundance of phylum Tenericutes (β = 8.39 mm2, 95% CI: 3.08-13.70 mm2, p = 0.002). Additionally, higher abundance of phylum Tenericutes was associated with greater SA of the lateral orbitofrontal cortex (β = 10.51 mm2, 95% CI: 3.24-17.79 mm2, p = 0.0046). No evidence of heterogeneity or pleiotropy was detected. Conclusion Specific gut microbiota may causally influence cortical structure in brain regions involved in neuropsychiatric disorders. The findings provide evidence for a gut-brain axis influencing cortical development, particularly in the orbitofrontal cortex during adolescence.
Collapse
Affiliation(s)
- Maochao Zhou
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Song Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yan Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| | | | - Chunmei Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
181
|
Chen G, Li L, Sun T, Jiang C, Xu W, Chen S, Hu C, Yue Y, Wang T, Jiang W, Yuan Y. The Interaction of LAMA2 and Duration of Illness Affects the Thickness of the Right Transverse Temporal Gyrus in Major Depressive Disorder. Neuropsychiatr Dis Treat 2023; 19:2807-2816. [PMID: 38144699 PMCID: PMC10749177 DOI: 10.2147/ndt.s435025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/14/2023] [Indexed: 12/26/2023] Open
Abstract
Background Depression is a heritable brain disorder. Laminin genes were recently identified to affect the brain's overall thickness through neurogenesis, differentiation, and migration in depression. This study aims to explore the effects of the LAMA2's single nucleotide polymorphisms (SNP), a subunit gene of laminin, on the detected brain regions of patients with major depressive disorder (MDD). Methods The study included 89 patients with MDD and 60 healthy controls with T1-weighted structural magnetic resonance imaging and blood samples for genotyping. The interactions between LAMA2 gene SNPs and diagnosis as well as duration of illness (DOI) were explored on brain measures controlled for age, gender, and site. Results The right transverse temporal gyrus and right parahippocampal gyrus showed reduced thickness in MDD. Almost all seven LAMA2 SNPs showed significant interactions with diagnosis on both gyrus (corrected p < 0.05 or trending). In MDD, rs6569604, rs2229848, rs2229849, rs2229850, and rs2784895 interacted with DOI on the right transverse temporal gyrus (corrected p < 0.05), but not the right parahippocampal gyrus. Conclusion The thickness of the right transverse temporal gyrus in patients with MDD may be affected by LAMA2 gene and DOI.
Collapse
Affiliation(s)
- Gang Chen
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Department of Medical Psychology, Huai’an NO 3 People’s Hospital, Huaian, People’s Republic of China
| | - Lei Li
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Department of Sleep Medicine, The Fourth People’s Hospital of Lianyungang, Lianyungang, People’s Republic of China
| | - Taipeng Sun
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Department of Medical Psychology, Huai’an NO 3 People’s Hospital, Huaian, People’s Republic of China
| | - Chenguang Jiang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Wei Xu
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Suzhen Chen
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Changchun Hu
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Yingying Yue
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Tianyu Wang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Wenhao Jiang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
182
|
Dong L, Hou B, Liu C, Mao C, Huang X, Shang L, Chu S, Peng B, Cui L, Feng F, Gao J. Association Between Wnt Target Genes and Cortical Volumes in Alzheimer's Disease. J Mol Neurosci 2023; 73:1010-1016. [PMID: 38135866 PMCID: PMC10754720 DOI: 10.1007/s12031-023-02122-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/16/2023] [Indexed: 12/24/2023]
Abstract
The disproportionate cortical atrophy is an established biomarker for the pathophysiological process of Alzheimer's disease (AD). However, the genetic basis underlying the cortical atrophy remains poorly defined. Herein, we aim to illustrate the effect of the Wnt target genes on the cortical volumes of AD patients. 82 sporadic AD patients were recruited. All the subjects had history survey, blood biochemical examination, cognitive assessment, MRI morphometry and whole exome sequencing. This report focused on 84 common variants (minor allele frequency > 0.01) of 32 Wnt target genes, including the APC, DAAM1, DACT1, DISC1, LATS2, TLR2, WDR61, and the AXIN, DVL, FZD, LRP, TCF/LEF, WNT family genes. The Wnt target genes showed asymmetric effects on the cortical volumes of AD patients. The right temporal/parietal/occipital cortices were more affected than left temporal/parietal/occipital cortices. Nevertheless, the reverse applied to the frontal cortex. The DACT1 affected the cortical thickness most, followed by the TCF3 and APC. The DACT1 rs698025-GG genotype displayed greater right temporal pole and left medial orbito-frontal gyrus than rs698025-GA genotype (2.4 ± 0.4 vs. 2.0 ± 0.6, P = 0.005; 5.2 ± 0.6 vs. 5.0 ± 0.6, P = 0.001). The brain region most influenced by the Wnt target genes was the right calcarine cortex. In conclusion, the common variants of the Wnt target genes exert asymmetric effects on the cortical volumes of AD patients. The Wnt signaling pathway may play a role in the cortical atrophy of AD patients.
Collapse
Affiliation(s)
- Liling Dong
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China
| | - Bo Hou
- Radiology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China
| | - Caiyan Liu
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China
| | - Chenhui Mao
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China
| | - Xinying Huang
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China
| | - Li Shang
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China
| | - Shanshan Chu
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China
| | - Bin Peng
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China
| | - Liying Cui
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China
| | - Feng Feng
- Radiology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China.
| | - Jing Gao
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100005, China.
| |
Collapse
|
183
|
Bhattacharya A, Vo DD, Jops C, Kim M, Wen C, Hervoso JL, Pasaniuc B, Gandal MJ. Isoform-level transcriptome-wide association uncovers genetic risk mechanisms for neuropsychiatric disorders in the human brain. Nat Genet 2023; 55:2117-2128. [PMID: 38036788 PMCID: PMC10703692 DOI: 10.1038/s41588-023-01560-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/05/2023] [Indexed: 12/02/2023]
Abstract
Methods integrating genetics with transcriptomic reference panels prioritize risk genes and mechanisms at only a fraction of trait-associated genetic loci, due in part to an overreliance on total gene expression as a molecular outcome measure. This challenge is particularly relevant for the brain, in which extensive splicing generates multiple distinct transcript-isoforms per gene. Due to complex correlation structures, isoform-level modeling from cis-window variants requires methodological innovation. Here we introduce isoTWAS, a multivariate, stepwise framework integrating genetics, isoform-level expression and phenotypic associations. Compared to gene-level methods, isoTWAS improves both isoform and gene expression prediction, yielding more testable genes, and increased power for discovery of trait associations within genome-wide association study loci across 15 neuropsychiatric traits. We illustrate multiple isoTWAS associations undetectable at the gene-level, prioritizing isoforms of AKT3, CUL3 and HSPD1 in schizophrenia and PCLO with multiple disorders. Results highlight the importance of incorporating isoform-level resolution within integrative approaches to increase discovery of trait associations, especially for brain-relevant traits.
Collapse
Affiliation(s)
- Arjun Bhattacharya
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Institute for Data Science in Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Daniel D Vo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Connor Jops
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Minsoo Kim
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Cindy Wen
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Jonatan L Hervoso
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Michael J Gandal
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
184
|
Wang Q, Xu S, Liu F, Liu Y, Chen K, Huang L, Xu F, Liu Y. Causal relationship between sleep traits and cognitive impairment: A Mendelian randomization study. J Evid Based Med 2023; 16:485-494. [PMID: 38108111 DOI: 10.1111/jebm.12576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/30/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVE Observational studies had demonstrated a link between sleep disturbances and cognitive decline. Here, we aimed to investigate the causal association between genetically predicted sleep traits and cognitive impairment using Mendelian randomization (MR). METHODS Using strict criteria, we selected genetic variants from European ancestry Genome-wide association studies (GWAS) from the Sleep Disorders Knowledge Portal and UK Biobank as instrumental variables for several sleep traits, including insomnia, sleep duration, daytime sleepiness, daytime napping, and chronotype. Summary statistics related to cognitive impairment were derived from five different GWAS, including the Social Science Genetic Association Consortium. The role of self-reported sleep trait phenotypes in the etiology of cognitive impairment was explored using inverse-variance weighted (IVW) tests, MR-Egger tests, and weighted medians, and sensitivity analyses were conducted to ensure robustness. RESULTS In the main IVW analysis, sleep duration (reaction time: β = -0.05, 95% CI -0.07 to -0.04, p = 1.93×10-12 ), daytime sleepiness (average cortical thickness: β = -0.12, 95% CI -0.22 to -0.02, p = 0.023), and daytime napping (fluid intelligence: β = -0.47, 95% CI -0.87 to -0.07, p = 0.021; hippocampal volume in Alzheimer's disease: β = -0.99, 95% CI -1.64 to -0.35, p = 0.002) were significantly negatively correlated with cognitive performance. However, any effects of insomnia and chronotype on cognitive impairment were not determined. CONCLUSIONS Our findings highlighted that focusing on sleep behaviors or distinct sleep patterns-particularly sleep duration, daytime sleepiness, and daytime napping, was a promising approach for preventing cognitive impairment. This study also shed light on risk factors for and potential early markers of cognitive impairment risk factors.
Collapse
Affiliation(s)
- Qing Wang
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Traditional Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Xu
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Traditional Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fenglan Liu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanfei Liu
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Traditional Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Keji Chen
- National Clinical Research Center for Traditional Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 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, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Traditional Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- National Clinical Research Center for Traditional Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
185
|
Ma DR, Li SJ, Shi JJ, Liang YY, Hu ZW, Hao XY, Li MJ, Guo MN, Zuo CY, Yu WK, Mao CY, Tang MB, Zhang C, Xu YM, Wu J, Sun SL, Shi CH. Shared Genetic Architecture between Parkinson's Disease and Brain Structural Phenotypes. Mov Disord 2023; 38:2258-2268. [PMID: 37990409 DOI: 10.1002/mds.29598] [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: 05/10/2023] [Revised: 08/02/2023] [Accepted: 08/21/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Patients with Parkinson's disease (PD) have consistently demonstrated brain structure abnormalities, indicating the presence of shared etiological and pathological processes between PD and brain structures; however, the genetic relationship remains poorly understood. OBJECTIVE The aim of this study was to investigate the extent of shared genetic architecture between PD and brain structural phenotypes (BSPs) and to identify shared genomic loci. METHODS We used the summary statistics from genome-wide association studies to conduct MiXeR and conditional/conjunctional false discovery rate analyses to investigate the shared genetic signatures between PD and BSPs. Subsequent expression quantitative trait loci mapping in the human brain and enrichment analyses were also performed. RESULTS MiXeR analysis identified genetic overlap between PD and various BSPs, including total cortical surface area, average cortical thickness, and specific brain volumetric structures. Further analysis using conditional false discovery rate (FDR) identified 21 novel PD risk loci on associations with BSPs at conditional FDR < 0.01, and the conjunctional FDR analysis demonstrated that PD shared several genomic loci with certain BSPs at conjunctional FDR < 0.05. Among the shared loci, 16 credible mapped genes showed high expression in the brain tissues and were primarily associated with immune function-related biological processes. CONCLUSIONS We confirmed the polygenic overlap with mixed directions of allelic effects between PD and BSPs and identified multiple shared genomic loci and risk genes, which are likely related to immune-related biological processes. These findings provide insight into the complex genetic architecture associated with PD. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Dong-Rui Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuang-Jie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jing-Jing Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yuan-Yuan Liang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zheng-Wei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xiao-Yan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Meng-Jie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Meng-Nan Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Chun-Yan Zuo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Wen-Kai Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Cheng-Yuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Mi-Bo Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Chan Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Jun Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Shi-Lei Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Chang-He Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
186
|
Kerestes R, Laansma MA, Owens-Walton C, Perry A, van Heese EM, Al-Bachari S, Anderson TJ, Assogna F, Aventurato ÍK, van Balkom TD, Berendse HW, van den Berg KR, Mphys RB, Brioschi R, Carr J, Cendes F, Clark LR, Dalrymple-Alford JC, Dirkx MF, Druzgal J, Durrant H, Emsley HC, Garraux G, Haroon HA, Helmich RC, van den Heuvel OA, João RB, Johansson ME, Khachatryan S, Lochner C, McMillan CT, Melzer TR, Mosley P, Newman B, Opriessnig P, Parkes LM, Pellicano C, Piras F, Pitcher TL, Poston KL, Rango M, Roos A, Rummel C, Schmidt R, Schwingenschuh P, Silva LS, Smith V, Squarcina L, Stein DJ, Tavadyan Z, Tsai CC, Vecchio D, Vriend C, Wang JJ, Wiest R, Yasuda CL, Young CB, Jahanshad N, Thompson PM, van der Werf YD, Harding IH, ENIGMA-Parkinson’s Study. Cerebellar Volume and Disease Staging in Parkinson's Disease: An ENIGMA-PD Study. Mov Disord 2023; 38:2269-2281. [PMID: 37964373 PMCID: PMC10754393 DOI: 10.1002/mds.29611] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/14/2023] [Accepted: 09/11/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Increasing evidence points to a pathophysiological role for the cerebellum in Parkinson's disease (PD). However, regional cerebellar changes associated with motor and non-motor functioning remain to be elucidated. OBJECTIVE To quantify cross-sectional regional cerebellar lobule volumes using three dimensional T1-weighted anatomical brain magnetic resonance imaging from the global ENIGMA-PD working group. METHODS Cerebellar parcellation was performed using a deep learning-based approach from 2487 people with PD and 1212 age and sex-matched controls across 22 sites. Linear mixed effects models compared total and regional cerebellar volume in people with PD at each Hoehn and Yahr (HY) disease stage, to an age- and sex- matched control group. Associations with motor symptom severity and Montreal Cognitive Assessment scores were investigated. RESULTS Overall, people with PD had a regionally smaller posterior lobe (dmax = -0.15). HY stage-specific analyses revealed a larger anterior lobule V bilaterally (dmax = 0.28) in people with PD in HY stage 1 compared to controls. In contrast, smaller bilateral lobule VII volume in the posterior lobe was observed in HY stages 3, 4, and 5 (dmax = -0.76), which was incrementally lower with higher disease stage. Within PD, cognitively impaired individuals had lower total cerebellar volume compared to cognitively normal individuals (d = -0.17). CONCLUSIONS We provide evidence of a dissociation between anterior "motor" lobe and posterior "non-motor" lobe cerebellar regions in PD. Whereas less severe stages of the disease are associated with larger motor lobe regions, more severe stages of the disease are marked by smaller non-motor regions. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Rebecca Kerestes
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Max A. Laansma
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Conor Owens-Walton
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Andrew Perry
- Monash Bioinformatics Platform, Monash University, Melbourne, VIC, Australia
| | - Eva M. van Heese
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Sarah Al-Bachari
- Faculty of Health and Medicine, The University of Lancaster, Lancaster, UK
| | - Tim J. Anderson
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Neurology Department, Te Wahtu Ora - Health New Zealand Waitaha Canterbury, Christchurch, New Zew Zealand
| | - Francesca Assogna
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Ítalo K. Aventurato
- Department of Neurology, University of Campinas - UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Tim D. van Balkom
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Amsterdam UMC, Dept. Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Henk W. Berendse
- Amsterdam UMC, Dept. Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Kevin R.E. van den Berg
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
- Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Rebecca Betts Mphys
- School of Physics and Astronomy, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Ricardo Brioschi
- Department of Neurology, University of Campinas - UNICAMP, Campinas, Brazil
| | - Jonathan Carr
- Division of Neurology, Tygerberg Hospital and Stellenbosch University, Cape Town, South Africa
| | - Fernando Cendes
- Department of Neurology, University of Campinas - UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Lyles R. Clark
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John C. Dalrymple-Alford
- New Zealand Brain Research Institute, Christchurch, New Zealand
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - Michiel F. Dirkx
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jason Druzgal
- Department of Radiology and Medical Imaging, University of Virginia, USA
| | - Helena Durrant
- School of Physics and Astronomy, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Hedley C.A. Emsley
- Lancaster Medical School, Lancaster University, Lancaster, UK
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Gaëtan Garraux
- GIGA-CRC in vivo imaging, University of Liège, Belgium
- Department of Neurology, CHU Liège, Liège, Belgium
| | - Hamied A. Haroon
- Division of Psychology, Communication & Human Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Rick C. Helmich
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
- Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Odile A. van den Heuvel
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Amsterdam UMC, Dept. Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Rafael B. João
- Department of Neurology, University of Campinas - UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Martin E. Johansson
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Samson Khachatryan
- Department of Neurology and Neurosurgery, National Institute of Health, Yerevan, Armenia
- Centers for Sleep and Movement Disorders, Somnus Neurology Clinic, Yerevan, Armenia
| | - Christine Lochner
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Cape Town, South Africa
| | - Corey T. McMillan
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Tracy R. Melzer
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - Philip Mosley
- Clinical Brain Networks Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- The Australian eHealth Research Centre, CSIRO Health and Biosecurity, Brisbane, Queensland, Australia
| | - Benjamin Newman
- Department of Radiology and Medical Imaging, University of Virginia, USA
| | - Peter Opriessnig
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Laura M. Parkes
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Clelia Pellicano
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Toni L. Pitcher
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Kathleen L. Poston
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Mario Rango
- Excellence Center for Advanced MR Techniques and Parkinson’s Disease Center, Neurology unit, Fondazione IRCCS Cà Granda Maggiore Policlinico Hospital, University of Milan, Milan, Italy
- Dept of Neurosciences, Neurology Unit, Fondazione Ca’ Granda, IRCCS, Ospedale Policlinico, Univeristy of Milan, Milano, Italy
| | - Annerine Roos
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Christian Rummel
- Support Center for Advanced Neuroimaging, (SCAN) University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Reinhold Schmidt
- Department of Neurology, Medical University of Graz, Graz, Austria
| | | | - Lucas S. Silva
- Department of Neurology, University of Campinas - UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Viktorija Smith
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Letizia Squarcina
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Dan J. Stein
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Zaruhi Tavadyan
- Department of Neurology and Neurosurgery, National Institute of Health, Yerevan, Armenia
- Centers for Sleep and Movement Disorders, Somnus Neurology Clinic, Yerevan, Armenia
| | - Chih-Chien Tsai
- Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Daniela Vecchio
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Chris Vriend
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Dept. Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, the Netherlands
| | - Jiun-Jie Wang
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan City, Taiwan
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung Branch Keelung City, Taiwan
- Healthy Ageing Research Center, ChangGung University, Taiwan
- Department of Chemical Engineering, Ming-Chi University of Technology, New Taipei City, Taiwan
| | - Roland Wiest
- Support Center for Advanced Neuroimaging, (SCAN) University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Clarissa L. Yasuda
- Department of Neurology, University of Campinas - UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Christina B. Young
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Ysbrand D. van der Werf
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Ian H. Harding
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia
| | | |
Collapse
|
187
|
Zhang X, Zhong Y, He K. The causal effects between selenium levels and the brain cortical structure: A two-sample Mendelian randomization study. Brain Behav 2023; 13:e3296. [PMID: 37904336 PMCID: PMC10726828 DOI: 10.1002/brb3.3296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/03/2023] [Accepted: 10/12/2023] [Indexed: 11/01/2023] Open
Abstract
Extensive research has demonstrated the critical role of selenium (Se) and selenoproteins in brain function and cognition. However, the impact of Se on brain cortical structure remains enigmatic. Therefore, this study used Mendelian randomization (MR) analysis to investigate the causal effect between Se levels and brain cortical structure. METHODS This study utilizes 11 genetic variants associated with Se level variations, extracted from a large-scale genome-wide association study (GWAS) encompassed circulating Se levels (n = 5477) and toenail Se levels (n = 4162) in the European population. Outcome data were sourced from the summary statistics of the ENIGMA Consortium, comprising GWAS data from 51,666 individuals. The variables include cortical surface area (SA), thickness (TH) at the global level, and 34 functional cortical regions evaluated by magnetic resonance imaging. The inverse-variance-weighted method was used as the primary estimate. Additionally, sensitivity analyses were conducted to detect potential violations of assumptions underlying MR. RESULTS At the global level, Se levels were not correlated with SA but showed a significant negative correlation with TH (β = -0.00485 mm, SE = 0.00192, p = .0115). Heterogeneity was observed across different brain regions, with positive correlations found between Se levels and the TH of the parahippocampal gyrus, superior frontal gyrus, and frontal pole, whereas negative correlations were found with the TH of the inferior parietal lobe and middle temporal lobe. Regarding SA, Se levels exhibit positive correlations with the pars triangularis, caudal anterior cingulate, inferior parietal lobe, and banks of the superior temporal sulcus. Conversely, negative correlations were observed with the medial orbitofrontal cortex, posterior cingulate gyrus, insula, and the middle, superior, and transverse gyrus of the temporal lobe. No pleiotropy was detected. RESULTS This MR study indicated that Se levels causally influence the brain cortical structure.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Yuqing Zhong
- The First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Kejun He
- Department of NeurosurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
188
|
Seidlitz J, Mallard TT, Vogel JW, Lee YH, Warrier V, Ball G, Hansson O, Hernandez LM, Mandal AS, Wagstyl K, Lombardo MV, Courchesne E, Glessner JT, Satterthwaite TD, Bethlehem RAI, Bernstock JD, Tasaki S, Ng B, Gaiteri C, Smoller JW, Ge T, Gur RE, Gandal MJ, Alexander-Bloch AF. The molecular genetic landscape of human brain size variation. Cell Rep 2023; 42:113439. [PMID: 37963017 PMCID: PMC11694216 DOI: 10.1016/j.celrep.2023.113439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/13/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
Human brain size changes dynamically through early development, peaks in adolescence, and varies up to 2-fold among adults. However, the molecular genetic underpinnings of interindividual variation in brain size remain unknown. Here, we leveraged postmortem brain RNA sequencing and measurements of brain weight (BW) in 2,531 individuals across three independent datasets to identify 928 genome-wide significant associations with BW. Genes associated with higher or lower BW showed distinct neurodevelopmental trajectories and spatial patterns that mapped onto functional and cellular axes of brain organization. Expression of BW genes was predictive of interspecies differences in brain size, and bioinformatic annotation revealed enrichment for neurogenesis and cell-cell communication. Genome-wide, transcriptome-wide, and phenome-wide association analyses linked BW gene sets to neuroimaging measurements of brain size and brain-related clinical traits. Cumulatively, these results represent a major step toward delineating the molecular pathways underlying human brain size variation in health and disease.
Collapse
Affiliation(s)
- Jakob Seidlitz
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Travis T Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA 02142, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02142, USA
| | - Jacob W Vogel
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA; Lifespan Informatics and Neuroimaging Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Younga H Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA 02142, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02142, USA
| | - Varun Warrier
- Department of Psychiatry, University of Cambridge, Cambridge CB2 1TN, UK; Department of Psychology, University of Cambridge, Cambridge CB2 1TN, UK
| | - Gareth Ball
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Melbourne, VIC 3052, Australia
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö P663+Q9, Sweden; Memory Clinic, Skåne University Hospital, Malmö P663+Q9, Sweden
| | - Leanna M Hernandez
- Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA 90024, USA
| | - Ayan S Mandal
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Konrad Wagstyl
- Wellcome Centre for Human Neuroimaging, University College London, London WC1N 3AR, UK
| | - Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Eric Courchesne
- Department of Neuroscience, University of California, San Diego, San Diego, CA 92093, USA; Autism Center of Excellence, University of California, San Diego, San Diego, CA 92093, USA
| | - Joseph T Glessner
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Theodore D Satterthwaite
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA; Lifespan Informatics and Neuroimaging Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | - Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard University, Boston, MA 02115, USA; Department of Neurosurgery, Boston Children's Hospital, Harvard University, Boston, MA 02115, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA 02142, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02142, USA; Center for Precision Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA 02142, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02142, USA; Center for Precision Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Raquel E Gur
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Gandal
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aaron F Alexander-Bloch
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
189
|
Li C, Chen Z, He S, Chen Y, Liu J. Unveiling the influence of daily dietary patterns on brain cortical structure: insights from bidirectional Mendelian randomization. Food Funct 2023; 14:10418-10429. [PMID: 37960880 DOI: 10.1039/d3fo02879h] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Cognitive impairment is a significant concern in aging populations. This study utilized Mendelian randomization analysis to explore the impact of dietary habits and macro-nutrients on cortical structure. A bidirectional Mendelian randomization approach was employed, incorporating large-scale genetic data on dietary habits and brain cortical structure. The results did not reveal significant causal relationships between dietary factors and overall cortical structure and thickness. However, specific dietary factors showed associations with cortical structure in certain regions. For instance, fat intake affected six cortical regions, while milk, protein, fruits, and water were associated with changes in specific regions. Reverse analysis suggested that cortical thickness influenced the consumption of alcohol, carbohydrates, coffee, and fish. These findings contribute to understanding the potential mechanisms underlying the role of dietary factors in cognitive function changes and provide evidence supporting the existence of the gut-brain axis.
Collapse
Affiliation(s)
- Cong Li
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| | - Zhe Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Shaqi He
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| | - Yanjing Chen
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
- Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan Province, 410011, People's Republic of China
| |
Collapse
|
190
|
Michel LC, McCormick EM, Kievit RA. Grey and white matter metrics demonstrate distinct and complementary prediction of differences in cognitive performance in children: Findings from ABCD (N= 11 876). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.529634. [PMID: 36945470 PMCID: PMC10028815 DOI: 10.1101/2023.03.06.529634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Individual differences in cognitive performance in childhood are a key predictor of significant life outcomes such as educational attainment and mental health. Differences in cognitive ability are governed in part by variations in brain structure. However, studies commonly focus on either grey or white matter metrics in humans, leaving open the key question as to whether grey or white matter microstructure play distinct or complementary roles supporting cognitive performance. To compare the role of grey and white matter in supporting cognitive performance, we used regularized structural equation models to predict cognitive performance with grey and white matter measures. Specifically, we compared how grey matter (volume, cortical thickness and surface area) and white matter measures (volume, fractional anisotropy and mean diffusivity) predicted individual differences in cognitive performance. The models were tested in 11,876 children (ABCD Study, 5680 female; 6196 male) at 10 years old. We found that grey and white matter metrics bring partly non-overlapping information to predict cognitive performance. The models with only grey or white matter explained respectively 15.4% and 12.4% of the variance in cognitive performance, while the combined model explained 19.0%. Zooming in we additionally found that different metrics within grey and white matter had different predictive power, and that the tracts/regions that were most predictive of cognitive performance differed across metric. These results show that studies focusing on a single metric in either grey or white matter to study the link between brain structure and cognitive performance are missing a key part of the equation.
Collapse
Affiliation(s)
- Lea C Michel
- Cognitive Neuroscience Department, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Ethan M McCormick
- Cognitive Neuroscience Department, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Methodology and Statistics, Institute of Psychology, Leiden University, Leiden, Netherlands
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, United States
| | - Rogier A Kievit
- Cognitive Neuroscience Department, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
191
|
Su WM, Gu XJ, Dou M, Duan QQ, Jiang Z, Yin KF, Cai WC, Cao B, Wang Y, Chen YP. Systematic druggable genome-wide Mendelian randomisation identifies therapeutic targets for Alzheimer's disease. J Neurol Neurosurg Psychiatry 2023; 94:954-961. [PMID: 37349091 PMCID: PMC10579488 DOI: 10.1136/jnnp-2023-331142] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia. Currently, there are no effective disease-modifying treatments for AD. Mendelian randomisation (MR) has been widely used to repurpose licensed drugs and discover novel therapeutic targets. Thus, we aimed to identify novel therapeutic targets for AD and analyse their pathophysiological mechanisms and potential side effects. METHODS A two-sample MR integrating the identified druggable genes was performed to estimate the causal effects of blood and brain druggable expression quantitative trait loci (eQTLs) on AD. A repeat study was conducted using different blood and brain eQTL data sources to validate the identified genes. Using AD markers with available genome-wide association studies data, we evaluated the causal relationship between established AD markers to explore possible mechanisms. Finally, the potential side effects of the druggable genes for AD treatment were assessed using a phenome-wide MR. RESULTS Overall, 5883 unique druggable genes were aggregated; 33 unique potential druggable genes for AD were identified in at least one dataset (brain or blood), and 5 were validated in a different dataset. Among them, three prior druggable genes (epoxide hydrolase 2 (EPHX2), SERPINB1 and SIGLEC11) reached significant levels in both blood and brain tissues. EPHX2 may mediate the pathogenesis of AD by affecting the entire hippocampal volume. Further phenome-wide MR analysis revealed no potential side effects of treatments targeting EPHX2, SERPINB1 or SIGLEC11. CONCLUSIONS This study provides genetic evidence supporting the potential therapeutic benefits of targeting the three druggable genes for AD treatment, which will be useful for prioritising AD drug development.
Collapse
Affiliation(s)
- Wei-Ming Su
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiao-Jing Gu
- Department of Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Dou
- Chengdu Computer Application Institute, Chinese Academy of Sciences, Chengdu, China
| | - Qing-Qing Duan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zheng Jiang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kang-Fu Yin
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei-Chen Cai
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic medical sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yong-Ping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
192
|
Matsumoto J, Fukunaga M, Miura K, Nemoto K, Okada N, Hashimoto N, Morita K, Koshiyama D, Ohi K, Takahashi T, Koeda M, Yamamori H, Fujimoto M, Yasuda Y, Ito S, Yamazaki R, Hasegawa N, Narita H, Yokoyama S, Mishima R, Miyata J, Kobayashi Y, Sasabayashi D, Harada K, Yamamoto M, Hirano Y, Itahashi T, Nakataki M, Hashimoto RI, Tha KK, Koike S, Matsubara T, Okada G, Yoshimura R, Abe O, van Erp TGM, Turner JA, Jahanshad N, Thompson PM, Onitsuka T, Watanabe Y, Matsuo K, Yamasue H, Okamoto Y, Suzuki M, Ozaki N, Kasai K, Hashimoto R. Cerebral cortical structural alteration patterns across four major psychiatric disorders in 5549 individuals. Mol Psychiatry 2023; 28:4915-4923. [PMID: 37596354 PMCID: PMC10914601 DOI: 10.1038/s41380-023-02224-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 07/30/2023] [Accepted: 08/07/2023] [Indexed: 08/20/2023]
Abstract
According to the operational diagnostic criteria, psychiatric disorders such as schizophrenia (SZ), bipolar disorder (BD), major depressive disorder (MDD), and autism spectrum disorder (ASD) are classified based on symptoms. While its cluster of symptoms defines each of these psychiatric disorders, there is also an overlap in symptoms between the disorders. We hypothesized that there are also similarities and differences in cortical structural neuroimaging features among these psychiatric disorders. T1-weighted magnetic resonance imaging scans were performed for 5,549 subjects recruited from 14 sites. Effect sizes were determined using a linear regression model within each protocol, and these effect sizes were meta-analyzed. The similarity of the differences in cortical thickness and surface area of each disorder group was calculated using cosine similarity, which was calculated from the effect sizes of each cortical regions. The thinnest cortex was found in SZ, followed by BD and MDD. The cosine similarity values between disorders were 0.943 for SZ and BD, 0.959 for SZ and MDD, and 0.943 for BD and MDD, which indicated that a common pattern of cortical thickness alterations was found among SZ, BD, and MDD. Additionally, a generally smaller cortical surface area was found in SZ and MDD than in BD, and the effect was larger in SZ. The cosine similarity values between disorders were 0.945 for SZ and MDD, 0.867 for SZ and ASD, and 0.811 for MDD and ASD, which indicated a common pattern of cortical surface area alterations among SZ, MDD, and ASD. Patterns of alterations in cortical thickness and surface area were revealed in the four major psychiatric disorders. To our knowledge, this is the first report of a cross-disorder analysis conducted on four major psychiatric disorders. Cross-disorder brain imaging research can help to advance our understanding of the pathogenesis of psychiatric disorders and common symptoms.
Collapse
Affiliation(s)
- Junya Matsumoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, 187-8553, Japan
| | - Masaki Fukunaga
- Section of Brain Function Information, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Kenichiro Miura
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, 187-8553, Japan
| | - Kiyotaka Nemoto
- Department of Psychiatry, Institute of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Naohiro Okada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- The International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, 113-0033, Japan
| | - Naoki Hashimoto
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Kentaro Morita
- Department of Rehabilitation, University of Tokyo Hospital, Tokyo, 113-8655, Japan
| | - Daisuke Koshiyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Kazutaka Ohi
- Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
- Department of General Internal Medicine, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Tsutomu Takahashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, 930-0194, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan
| | - Michihiko Koeda
- Department of Neuropsychiatry, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8602, Japan
| | - Hidenaga Yamamori
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, 187-8553, Japan
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
- Japan Community Health Care Organization Osaka Hospital, Osaka, 553-0003, Japan
| | - Michiko Fujimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, 187-8553, Japan
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Yuka Yasuda
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, 187-8553, Japan
- Life Grow Brilliant Mental Clinic, Medical Corporation Foster, Osaka, 530-0013, Japan
| | - Satsuki Ito
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, 187-8553, Japan
- Department of Developmental and Clinical Psychology, The Division of Human Developmental Sciences, Graduate School of Humanity and Sciences, Ochanomizu University, Tokyo, 112-8610, Japan
| | - Ryuichi Yamazaki
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, 187-8553, Japan
- Department of Psychiatry, The Jikei University School of Medicine, Tokyo, 105-8461, Japan
| | - Naomi Hasegawa
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, 187-8553, Japan
| | - Hisashi Narita
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Satoshi Yokoyama
- Department of Psychiatry and Neurosciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Ryo Mishima
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Jun Miyata
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Yuko Kobayashi
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Daiki Sasabayashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, 930-0194, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan
| | - Kenichiro Harada
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Maeri Yamamoto
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yoji Hirano
- Department of Psychiatry, Division of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Institute of Industrial Science, The University of Tokyo, Tokyo, 153-8505, Japan
| | - Takashi Itahashi
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo, 157-8577, Japan
| | - Masahito Nakataki
- Department of Psychiatry, Tokushima University Hospital, Tokushima, 770-8503, Japan
| | - Ryu-Ichiro Hashimoto
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo, 157-8577, Japan
- Department of Language Sciences, Graduate School of Humanities, Tokyo Metropolitan University, Hachioji, 192-0397, Japan
| | - Khin K Tha
- Global Center for Biomedical Science and Engineering, Hokkaido University Faculty of Medicine, Sapporo, 060-8638, Japan
| | - Shinsuke Koike
- The International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, 113-0033, Japan
- University of Tokyo Institute for Diversity & Adaptation of Human Mind (UTIDAHM), Tokyo, 153-8902, Japan
- Center for Evolutionary Cognitive Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, 153-8902, Japan
| | - Toshio Matsubara
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Go Okada
- Department of Psychiatry and Neurosciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Reiji Yoshimura
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Theo G M van Erp
- Clinical Translatational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, 92697, USA
| | - Jessica A Turner
- Department of Psychiatry and Behavioral Health, Wexner Medical Center, the Ohio State University, Columbus, OH, 43210, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90292, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90292, USA
| | - Toshiaki Onitsuka
- National Hospital Organization Sakakibara Hospital, Tsu, 514-1292, Japan
| | - Yoshiyuki Watanabe
- Department of Radiology, Shiga University of Medical Science, Otsu, 520-2192, Japan
| | - Koji Matsuo
- Department of Psychiatry, Faculty of Medicine, Saitama Medical University, Saitama, 350-0495, Japan
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Yasumasa Okamoto
- Department of Psychiatry and Neurosciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Michio Suzuki
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, 930-0194, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan
| | - Norio Ozaki
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- The International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, 113-0033, Japan
- University of Tokyo Institute for Diversity & Adaptation of Human Mind (UTIDAHM), Tokyo, 153-8902, Japan
| | - Ryota Hashimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, 187-8553, Japan.
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan.
| |
Collapse
|
193
|
Mai Z, Mao H. Causal effects of nonalcoholic fatty liver disease on cerebral cortical structure: a Mendelian randomization analysis. Front Endocrinol (Lausanne) 2023; 14:1276576. [PMID: 38027213 PMCID: PMC10646496 DOI: 10.3389/fendo.2023.1276576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Background Previous studies have highlighted changes in the cerebral cortical structure and cognitive function among nonalcoholic fatty liver disease (NAFLD) patients. However, the impact of NAFLD on cerebral cortical structure and specific affected brain regions remains unclear. Therefore, we aimed to explore the potential causal relationship between NAFLD and cerebral cortical structure. Methods We conducted a Mendelian randomization (MR) study using genetic predictors of alanine aminotransferase (ALT), NAFLD, and percent liver fat (PLF) and combined them with genome-wide association study (GWAS) summary statistics from the ENIGMA Consortium. Several methods were used to assess the effect of NAFLD on full cortex and specific brain regions, along with sensitivity analyses. Results At the global level, PLF nominally decreased SA of full cortex; at the functional level, ALT presented a nominal association with reduced SA of parahippocampal gyrus, TH of pars opercularis, TH of pars orbitalis, and TH of pericalcarine cortex. Besides, NAFLD presented a nominal association with reduced SA of parahippocampal gyrus, TH of pars opercularis, TH of pars triangularis and TH of pericalcarine cortex, but increased TH of entorhinal cortex, lateral orbitofrontal cortex and temporal pole. Furthermore, PLF presented a nominal association with reduced SA of parahippocampal gyrus, TH of pars opercularis, TH of cuneus and lingual gyrus, but increased TH of entorhinal cortex. Conclusion NAFLD is suggestively associated with atrophy in specific functional regions of the human brain.
Collapse
Affiliation(s)
- Zhiliang Mai
- Department of Digestive Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Anatomy, Guangdong Medical University, Zhanjiang, China
| | - Hua Mao
- Department of Digestive Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
194
|
Wang Z, Zou J, Zhang L, Ning J, Zhang X, Jiang B, Liang Y, Zhang Y. The impact of early adversity on the cerebral cortex - a Mendelian randomization study. Front Neurosci 2023; 17:1283159. [PMID: 37965215 PMCID: PMC10641447 DOI: 10.3389/fnins.2023.1283159] [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: 08/25/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Background The early adversity is associated with a series of negative outcomes in adulthood, and the impact on the cerebral cortex may be one of the fundamental causes of these adverse consequences in adulthood. In this study, we aim to investigate the causal relationship between early adversity and changes in cerebral cortex structure using Mendelian randomization (MR) analysis. Methods The GWAS summary statistics of 6 early adversity traits were obtained from individuals of European ancestry in the UK Biobank. The GWAS summary statistics of 34 known functional cortical regions were obtained from the ENIGMA Consortium. Causal relationships between the adversity factors and brain cortical structure were assessed using the inverse-variance weighted (IVW), MR-Egger, and weighted median methods, with IVW being the primary evaluation method. Cochran's Q-test, MR-PRESSO, leave-one-out analysis, and funnel plot examination were employed to detect potential heterogeneity and pleiotropy, as well as to identify and exclude outliers. Results At a global level, no causal relationship was found between early adversity and cortical thickness (TH) or surface area (SA) of the brain. However, at the regional level, early adversity was found to potentially influence the TH of the caudal anterior cingulate, superior temporal, entorhinal, paracentral, lateral occipital, banks of the superior temporal sulcus, and supramarginal regions, as well as the SA of the pars triangularis, lateral occipital, parahippocampal, medial orbitofrontal, and isthmus cingulate regions. All findings were nominally significant and passed sensitivity analyses, with no significant heterogeneity or pleiotropy detected. Discussion Our study provides evidence for the association between early adversity and alterations in brain cortical structure, which may serve as a foundation for certain mental disorders. Furthermore, magnetic resonance imaging (MRI) might be considered as a promising tool to aid healthcare professionals in identifying individuals with a history of adverse experiences, allowing for early interventions.
Collapse
Affiliation(s)
- Zhen Wang
- College of Basic Medical Sciences, Dali University, Dali, Yunnan, China
| | - Jing Zou
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Le Zhang
- College of Basic Medical Sciences, Dali University, Dali, Yunnan, China
| | - Jinghua Ning
- College of Basic Medical Sciences, Dali University, Dali, Yunnan, China
| | - Xin Zhang
- College of Basic Medical Sciences, Dali University, Dali, Yunnan, China
| | - Bei Jiang
- Yunnan Key Laboratory of Screening and Research on Anti-pathogenic Plant Resources from West Yunnan (Cultivation), Dali, Yunnan, China
| | - Yi Liang
- Princess Margaret Cancer Centre, University Health Network, TMDT-MaRS Centre, Toronto, ON, Canada
| | - Yuzhe Zhang
- College of Basic Medical Sciences, Dali University, Dali, Yunnan, China
| |
Collapse
|
195
|
Antón-Galindo E, Cabana-Domínguez J, Torrico B, Corominas R, Cormand B, Fernàndez-Castillo N. The pleiotropic contribution of genes in dopaminergic and serotonergic pathways to addiction and related behavioral traits. Front Psychiatry 2023; 14:1293663. [PMID: 37937232 PMCID: PMC10627163 DOI: 10.3389/fpsyt.2023.1293663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/28/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Co-occurrence of substance use disorders (SUD) and other behavioral conditions, such as stress-related, aggressive or risk-taking behaviors, in the same individual has been frequently described. As dopamine (DA) and serotonin (5-HT) have been previously identified as key neurotransmitters for some of these phenotypes, we explored the genetic contribution of these pathways to SUD and these comorbid phenotypes in order to better understand the genetic relationship between them. Methods We tested the association of 275 dopaminergic genes and 176 serotonergic genes with these phenotypes by performing gene-based, gene-set and transcriptome-wide association studies in 11 genome-wide association studies (GWAS) datasets on SUD and related behaviors. Results At the gene-wide level, 68 DA and 27 5-HT genes were found to be associated with at least one GWAS on SUD or related behavior. Among them, six genes had a pleiotropic effect, being associated with at least three phenotypes: ADH1C, ARNTL, CHRNA3, HPRT1, HTR1B and DRD2. Additionally, we found nominal associations between the DA gene sets and SUD, opioid use disorder, antisocial behavior, irritability and neuroticism, and between the 5-HT-core gene set and neuroticism. Predicted gene expression correlates in brain were also found for 19 DA or 5-HT genes. Discussion Our study shows a pleiotropic contribution of dopaminergic and serotonergic genes to addiction and related behaviors such as anxiety, irritability, neuroticism and risk-taking behavior, highlighting a role for DA genes, which could explain, in part, the co-occurrence of these phenotypes.
Collapse
Affiliation(s)
- Ester Antón-Galindo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Judit Cabana-Domínguez
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Bàrbara Torrico
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Roser Corominas
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| |
Collapse
|
196
|
Huang Y, Zhang T, Zhang S, Zhang W, Yang L, Zhu D, Liu T, Jiang X, Han J, Guo L. Genetic Influence on Gyral Peaks. Neuroimage 2023; 280:120344. [PMID: 37619794 DOI: 10.1016/j.neuroimage.2023.120344] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023] Open
Abstract
Genetic mechanisms have been hypothesized to be a major determinant in the formation of cortical folding. Although there is an increasing number of studies examining the heritability of cortical folding, most of them focus on sulcal pits rather than gyral peaks. Gyral peaks, which reflect the highest local foci on gyri and are consistent across individuals, remain unstudied in terms of heritability. To address this knowledge gap, we used high-resolution data from the Human Connectome Project (HCP) to perform classical twin analysis and estimate the heritability of gyral peaks across various brain regions. Our results showed that the heritability of gyral peaks was heterogeneous across different cortical regions, but relatively symmetric between hemispheres. We also found that pits and peaks are different in a variety of anatomic and functional measures. Further, we explored the relationship between the levels of heritability and the formation of cortical folding by utilizing the evolutionary timeline of gyrification. Our findings indicate that the heritability estimates of both gyral peaks and sulcal pits decrease linearly with the evolution timeline of gyrification. This suggests that the cortical folds which formed earlier during gyrification are subject to stronger genetic influences than the later ones. Moreover, the pits and peaks coupled by their time of appearance are also positively correlated in respect of their heritability estimates. These results fill the knowledge gap regarding genetic influences on gyral peaks and significantly advance our understanding of how genetic factors shape the formation of cortical folding. The comparison between peaks and pits suggests that peaks are not a simple morphological mirror of pits but could help complete the understanding of folding patterns.
Collapse
Affiliation(s)
- Ying Huang
- School of Automation, Northwestern Polytechnical University, Xi'an 710129, China; School of Information and Technology, Northwest University, Xi'an 710127, China
| | - Tuo Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an 710129, China.
| | - Songyao Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an 710129, China
| | - Weihan Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an 710129, China
| | - Li Yang
- School of Automation, Northwestern Polytechnical University, Xi'an 710129, China
| | - Dajiang Zhu
- Computer Science & Engineering, University of Texas at Arlington, TX 76010, USA
| | - Tianming Liu
- Cortical Architecture Imaging and Discovery Lab, Department of Computer Science and Bioimaging Research Center, The University of Georgia, Athens, GA 30602, USA
| | - Xi Jiang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610056, China
| | - Junwei Han
- School of Automation, Northwestern Polytechnical University, Xi'an 710129, China
| | - Lei Guo
- School of Automation, Northwestern Polytechnical University, Xi'an 710129, China
| |
Collapse
|
197
|
Chiou KL, Huang X, Bohlen MO, Tremblay S, DeCasien AR, O’Day DR, Spurrell CH, Gogate AA, Zintel TM, Cayo Biobank Research Unit, Andrews MG, Martínez MI, Starita LM, Montague MJ, Platt ML, Shendure J, Snyder-Mackler N. A single-cell multi-omic atlas spanning the adult rhesus macaque brain. SCIENCE ADVANCES 2023; 9:eadh1914. [PMID: 37824616 PMCID: PMC10569716 DOI: 10.1126/sciadv.adh1914] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Cataloging the diverse cellular architecture of the primate brain is crucial for understanding cognition, behavior, and disease in humans. Here, we generated a brain-wide single-cell multimodal molecular atlas of the rhesus macaque brain. Together, we profiled 2.58 M transcriptomes and 1.59 M epigenomes from single nuclei sampled from 30 regions across the adult brain. Cell composition differed extensively across the brain, revealing cellular signatures of region-specific functions. We also identified 1.19 M candidate regulatory elements, many previously unidentified, allowing us to explore the landscape of cis-regulatory grammar and neurological disease risk in a cell type-specific manner. Altogether, this multi-omic atlas provides an open resource for investigating the evolution of the human brain and identifying novel targets for disease interventions.
Collapse
Affiliation(s)
- Kenneth L. Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Xingfan Huang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Martin O. Bohlen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sébastien Tremblay
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex R. DeCasien
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| | - Diana R. O’Day
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Cailyn H. Spurrell
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Aishwarya A. Gogate
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Trisha M. Zintel
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Cayo Biobank Research Unit
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Caribbean Primate Research Center, University of Puerto Rico, San Juan, PR, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Madeline G. Andrews
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Melween I. Martínez
- Caribbean Primate Research Center, University of Puerto Rico, San Juan, PR, USA
| | - Lea M. Starita
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Michael J. Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael L. Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
198
|
Johansen N, Somasundaram S, Travaglini KJ, Yanny AM, Shumyatcher M, Casper T, Cobbs C, Dee N, Ellenbogen R, Ferreira M, Goldy J, Guzman J, Gwinn R, Hirschstein D, Jorstad NL, Keene CD, Ko A, Levi BP, Ojemann JG, Pham T, Shapovalova N, Silbergeld D, Sulc J, Torkelson A, Tung H, Smith K, Lein ES, Bakken TE, Hodge RD, Miller JA. Interindividual variation in human cortical cell type abundance and expression. Science 2023; 382:eadf2359. [PMID: 37824649 DOI: 10.1126/science.adf2359] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 07/30/2023] [Indexed: 10/14/2023]
Abstract
Single-cell transcriptomic studies have identified a conserved set of neocortical cell types from small postmortem cohorts. We extended these efforts by assessing cell type variation across 75 adult individuals undergoing epilepsy and tumor surgeries. Nearly all nuclei map to one of 125 robust cell types identified in the middle temporal gyrus. However, we found interindividual variance in abundances and gene expression signatures, particularly in deep-layer glutamatergic neurons and microglia. A minority of donor variance is explainable by age, sex, ancestry, disease state, and cell state. Genomic variation was associated with expression of 150 to 250 genes for most cell types. This characterization of cellular variation provides a baseline for cell typing in health and disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Tamara Casper
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Charles Cobbs
- Swedish Neuroscience Institute, Seattle,WA 98122, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Richard Ellenbogen
- Department of Neurological Surgery, University of Washington, Seattle, WA 98104, USA
| | - Manuel Ferreira
- Department of Neurological Surgery, University of Washington, Seattle, WA 98104, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Junitta Guzman
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ryder Gwinn
- Swedish Neuroscience Institute, Seattle,WA 98122, USA
| | | | | | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104, USA
| | - Andrew Ko
- Department of Neurological Surgery, University of Washington, Seattle, WA 98104, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jeffrey G Ojemann
- Department of Neurological Surgery, University of Washington, Seattle, WA 98104, USA
| | - Thanh Pham
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Daniel Silbergeld
- Department of Neurological Surgery, University of Washington, Seattle, WA 98104, USA
| | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Amy Torkelson
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kimberly Smith
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | |
Collapse
|
199
|
Sha Z, Warrier V, Bethlehem RA, Schultz LM, Merikangas A, Sun KY, Gur RC, Gur RE, Shinohara RT, Seidlitz J, Almasy L, Andreassen OA, Alexander-Bloch AF. The overlapping genetic architecture of psychiatric disorders and cortical brain structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.05.561040. [PMID: 37873315 PMCID: PMC10592957 DOI: 10.1101/2023.10.05.561040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Both psychiatric vulnerability and cortical structure are shaped by the cumulative effect of common genetic variants across the genome. However, the shared genetic underpinnings between psychiatric disorders and brain structural phenotypes, such as thickness and surface area of the cerebral cortex, remains elusive. In this study, we employed pleiotropy-informed conjunctional false discovery rate analysis to investigate shared loci across genome-wide association scans of regional cortical thickness, surface area, and seven psychiatric disorders in approximately 700,000 individuals of European ancestry. Aggregating regional measures, we identified 50 genetic loci shared between psychiatric disorders and surface area, as well as 26 genetic loci shared with cortical thickness. Risk alleles exhibited bidirectional effects on both cortical thickness and surface area, such that some risk alleles for each disorder increased regional brain size while other risk alleles decreased regional brain size. Due to bidirectional effects, in many cases we observed extensive pleiotropy between an imaging phenotype and a psychiatric disorder even in the absence of a significant genetic correlation between them. The impact of genetic risk for psychiatric disorders on regional brain structure did exhibit a consistent pattern across highly comorbid psychiatric disorders, with 80% of the genetic loci shared across multiple disorders displaying consistent directions of effect. Cortical patterning of genetic overlap revealed a hierarchical genetic architecture, with the association cortex and sensorimotor cortex representing two extremes of shared genetic influence on psychiatric disorders and brain structural variation. Integrating multi-scale functional annotations and transcriptomic profiles, we observed that shared genetic loci were enriched in active genomic regions, converged on neurobiological and metabolic pathways, and showed differential expression in postmortem brain tissue from individuals with psychiatric disorders. Cumulatively, these findings provide a significant advance in our understanding of the overlapping polygenic architecture between psychopathology and cortical brain structure.
Collapse
Affiliation(s)
- Zhiqiang Sha
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Varun Warrier
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | | | - Laura M. Schultz
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Alison Merikangas
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin Y. Sun
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
| | - Russell T. Shinohara
- Penn Statistics in Imaging and Visualization Endeavor (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 423 Guardian Dr, Philadelphia, PA 19104, United States
- Center for Biomedical Image Computing and Analytics (CBICA), Department of Radiology, Perelman School of Medicine, United States
| | - Jakob Seidlitz
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Laura Almasy
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ole A. Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Aaron F. Alexander-Bloch
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| |
Collapse
|
200
|
Zhao S, Chi L, Chen H. CEGA: a method for inferring natural selection by comparative population genomic analysis across species. Genome Biol 2023; 24:219. [PMID: 37789379 PMCID: PMC10548728 DOI: 10.1186/s13059-023-03068-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
We developed maximum likelihood method for detecting positive selection or balancing selection using multilocus or genomic polymorphism and divergence data from two species. The method is especially useful for investigating natural selection in noncoding regions. Simulations demonstrate that the method outperforms existing methods in detecting both positive and balancing selection. We apply the method to population genomic data from human and chimpanzee. The list of genes identified under selection in the noncoding regions is prominently enriched in pathways related to the brain and nervous system. Therefore, our method will serve as a useful tool for comparative population genomic analysis.
Collapse
Affiliation(s)
- Shilei Zhao
- CAS Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
- School of Future Technology, College of Life Sciences and Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lianjiang Chi
- CAS Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
| | - Hua Chen
- CAS Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- China National Center for Bioinformation, Beijing, 100101, China.
- School of Future Technology, College of Life Sciences and Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|