1
|
Qi J, Suo X, Tian C, Xia X, Qin W, Wang P, Tang J, Xu J, Fu J, Liu N, Yu C, Shen H, Dou Y. TESC overexpression mitigates amyloid-β-induced hippocampal atrophy and memory decline. Gene 2025; 933:148939. [PMID: 39278373 DOI: 10.1016/j.gene.2024.148939] [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: 05/16/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND AND OBJECTIVES Genome-wide association studies (GWASs) have identified numerous candidate genes for human brain-imaging phenotypes; however, the biological relevance of many of these genes remains unconfirmed. This study aimed to investigate the causal relationships among tescalcin (TESC) (a GWAS-indicated gene), hippocampal volume, Alzheimer's disease (AD), and the underlying biological mechanisms. METHODS Human transcriptional data were analyzed to confirm relative TESC expression in the hippocampus. In cell experiments, RNA-seq analysis was used to identify the potential biological pathways for TESC overexpression, and immunofluorescence imaging and cell viability assays were used to evaluate the effect of TESC overexpression on neuronal structure and survival. In animal experiments, the effects of TESC overexpression on hippocampal volume and cognitive function in normal mice and amyloid-β (Aβ)-induced AD mice were investigated by 9.4 T magnetic resonance imaging and behavioral tests. Underlying mechanisms were further assessed via western blotting and electrophysiological recordings. RESULTS Human transcriptional data demonstrated that TESC is primarily expressed in the hippocampus and neurons. TESC overexpression enhanced the viability of HT22 cells and reduced Aβ-induced cell death. In mouse models, Tesc-overexpressing mice revealed increased hippocampal volume, likely owing to enhanced cell viability and long-term potentiation (LTP), and reducing apoptotic- and oxidation-induced hippocampal damage. TESC overexpression could significantly mitigate Aβ-induced hippocampal atrophy and memory impairment, potentially by reducing Aβ-induced neuronal apoptosis and LTP weakening. CONCLUSION This study exemplifies the translation of GWAS findings into actionable biological knowledge and suggests that upregulation of TESC may offer a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Jinbo Qi
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Xinjun Suo
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China; School of Medical Technology, Tianjin Medical University, Tianjin 300070, PR China
| | - Chunxiao Tian
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, PR China
| | - Xianyou Xia
- Department of Cell Biology, School of Basic Medicine and Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, PR China
| | - Wen Qin
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Ping Wang
- School of Medical Technology, Tianjin Medical University, Tianjin 300070, PR China
| | - Jie Tang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Jiayuan Xu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Jilian Fu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Nana Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China; School of Medical Technology, Tianjin Medical University, Tianjin 300070, PR China
| | - Hui Shen
- Department of Cell Biology, School of Basic Medicine and Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, PR China.
| | - Yan Dou
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China.
| |
Collapse
|
2
|
Mahmoodi M, Ayatollahi Mehrgardi A, Momen M, Serpell JA, Esmailizadeh A. Deciphering the genetic basis of behavioral traits in dogs: Observed-trait GWAS and latent-trait GWAS analysis reveal key genes and variants. Vet J 2024; 308:106251. [PMID: 39368730 DOI: 10.1016/j.tvjl.2024.106251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Dogs exhibit remarkable phenotypic diversity, particularly in behavioral traits, making them an excellent model for studying the genetic basis of complex behaviors. Behavioral traits such as aggression and fear are highly heritable among different dog breeds, but their genetic basis is largely unknown. We used the genome-wide association study (GWAS) to identify candidate genes associated with nine behavioral traits including; stranger-directed aggression (SDA), owner-directed aggression (ODA), dog-directed aggression (DDA), stranger-directed fear (SDF), nonsocial fear (NF), dog-directed fear (DDF), touch sensitivity (TS), separation-related behavior (SRB) and attachment attention-seeking (AAS). The observed behavioral traits were collected from 38,714 to 40,460 individuals across 108 modern dog breeds. We performed a GWAS based on a latent trait extracted using the confirmatory factor analysis (CFA) method with nine observable behavioral traits and compared the results with those from the GWAS of the observed traits. Using both observed-trait and latent-trait GWAS, we identified 41 significant SNPs that were common between both GWAS methods, of which 26 were pleiotropic, as well as 10 SNPs unique to the latent-trait GWAS, and 5 SNPs unique to the observed-trait GWAS discovered. These SNPs were associated with 21 genes in latent-trait GWAS and 22 genes in the observed-trait GWAS, with 19 genes shared by both. According to previous studies, some of the genes from this study have been reported to be related to behavioral and neurological functions in dogs. In the human population, these identified genes play a role in either the formation of the nervous system or are linked to various mental health conditions. Taken together, our findings suggest that latent-trait GWAS for behavioral traits in dogs identifies significant latent genes that are neurologically prioritized.
Collapse
Affiliation(s)
- Maryam Mahmoodi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran.
| | - Ahmad Ayatollahi Mehrgardi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran.
| | - Mehdi Momen
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - James A Serpell
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ali Esmailizadeh
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
3
|
Ning C, Jin M, Cai Y, Fan L, Hu K, Lu Z, Zhang M, Chen C, Li Y, Hu N, Zhang D, Liu Y, Chen S, Jiang Y, He C, Wang Z, Cao Z, Li H, Li G, Ma Q, Geng H, Tian W, Zhang H, Yang X, Huang C, Wei Y, Li B, Zhu Y, Li X, Miao X, Tian J. Genetic architectures of the human hippocampus and those involved in neuropsychiatric traits. BMC Med 2024; 22:456. [PMID: 39394562 PMCID: PMC11470718 DOI: 10.1186/s12916-024-03682-8] [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: 05/08/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND The hippocampus, with its complex subfields, is linked to numerous neuropsychiatric traits. While most research has focused on its global structure or a few specific subfields, a comprehensive analysis of hippocampal substructures and their genetic correlations across a wide range of neuropsychiatric traits remains underexplored. Given the hippocampus's high heritability, considering hippocampal and subfield volumes (HASV) as endophenotypes for neuropsychiatric conditions is essential. METHODS We analyzed MRI-derived volumetric data of hippocampal and subfield structures from 41,525 UK Biobank participants. Genome-wide association studies (GWAS) on 24 HASV traits were conducted, followed by genetic correlation, overlap, and Mendelian randomization (MR) analyses with 10 common neuropsychiatric traits. Polygenic risk scores (PRS) based on HASV traits were also evaluated for predicting these traits. RESULTS Our analysis identified 352 independent genetic variants surpassing a significance threshold of 2.1 × 10-9 within the 24 HASV traits, located across 93 chromosomal regions. Notably, the regions 12q14.3, 17q21.31, 12q24.22, 6q21, 9q33.1, 6q25.1, and 2q24.2 were found to influence multiple HASVs. Gene set analysis revealed enrichment of neural differentiation and signaling pathways, as well as protein binding and degradation. Of 240 HASV-neuropsychiatric trait pairs, 75 demonstrated significant genetic correlations (P < 0.05/240), revealing 433 pleiotropic loci. Particularly, genes like ACBD4, ARHGAP27, KANSL1, MAPT, ARL17A, and ARL17B were involved in over 50 HASV-neuropsychiatric pairs. Leveraging Mendelian randomization analysis, we further confirmed that atrophy in the left hippocampus, right hippocampus, right hippocampal body, and right CA1-3 region were associated with an increased risk of developing Parkinson's disease (PD). Furthermore, PRS for all four HASVs were significantly linked to a higher risk of Parkinson's disease (PD), with the highest hazard ratio (HR) of 1.30 (95% CI 1.18-1.43, P = 6.15 × 10⁻⁸) for right hippocampal volume. CONCLUSIONS These findings highlight the extensive distribution of pleiotropic genetic determinants between HASVs and neuropsychiatric traits. Moreover, they suggest a significant potential for effectively managing and intervening in these diseases during their early stages.
Collapse
Affiliation(s)
- Caibo Ning
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China
| | - Meng Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yimin Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China
| | - Linyun Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Kexin Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zequn Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Can Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yanmin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Naifan Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Donghui Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yizhuo Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Shuoni Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yuan Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Chunyi He
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zhuo Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zilong Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Hanting Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Gaoyuan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Qianying Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Hui Geng
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Wen Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Heng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Xiaojun Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chaoqun Huang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yongchang Wei
- Department of Gastrointestinal Oncology, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Bin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China
| | - Xiangpan Li
- Department of Radiation Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China.
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China.
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China.
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
4
|
Goel K, Chhetri A, Ludhiadch A, Munshi A. Current Update on Categorization of Migraine Subtypes on the Basis of Genetic Variation: a Systematic Review. Mol Neurobiol 2024; 61:4804-4833. [PMID: 38135854 DOI: 10.1007/s12035-023-03837-3] [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: 09/26/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
Migraine is a complex neurovascular disorder that is characterized by severe behavioral, sensory, visual, and/or auditory symptoms. It has been labeled as one of the ten most disabling medical illnesses in the world by the World Health Organization (Aagaard et al Sci Transl Med 6(237):237ra65, 2014). According to a recent report by the American Migraine Foundation (Shoulson et al Ann Neurol 25(3):252-9, 1989), around 148 million people in the world currently suffer from migraine. On the basis of presence of aura, migraine is classified into two major subtypes: migraine with aura (Aagaard et al Sci Transl Med 6(237):237ra65, 2014) and migraine without aura. (Aagaard K et al Sci Transl Med 6(237):237ra65, 2014) Many complex genetic mechanisms have been proposed in the pathophysiology of migraine but specific pathways associated with the different subtypes of migraine have not yet been explored. Various approaches including candidate gene association studies (CGAS) and genome-wide association studies (Fan et al Headache: J Head Face Pain 54(4):709-715, 2014). have identified the genetic markers associated with migraine and its subtypes. Several single nucleotide polymorphisms (Kaur et al Egyp J Neurol, Psychiatry Neurosurg 55(1):1-7, 2019) within genes involved in ion homeostasis, solute transport, synaptic transmission, cortical excitability, and vascular function have been associated with the disorder. Currently, the diagnosis of migraine is majorly behavioral with no focus on the genetic markers and thereby the therapeutic intervention specific to subtypes. Therefore, there is a need to explore genetic variants significantly associated with MA and MO as susceptibility markers in the diagnosis and targets for therapeutic interventions in the specific subtypes of migraine. Although the proper characterization of pathways based on different subtypes is yet to be studied, this review aims to make a first attempt to compile the information available on various genetic variants and the molecular mechanisms involved with the development of MA and MO. An attempt has also been made to suggest novel candidate genes based on their function to be explored by future research.
Collapse
Affiliation(s)
- Kashish Goel
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Aakash Chhetri
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Abhilash Ludhiadch
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Anjana Munshi
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401.
| |
Collapse
|
5
|
Zuo Q, Wu H, Chen CLP, Lei B, Wang S. Prior-Guided Adversarial Learning With Hypergraph for Predicting Abnormal Connections in Alzheimer's Disease. IEEE TRANSACTIONS ON CYBERNETICS 2024; 54:3652-3665. [PMID: 38236677 DOI: 10.1109/tcyb.2023.3344641] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Alzheimer's disease (AD) is characterized by alterations of the brain's structural and functional connectivity during its progressive degenerative processes. Existing auxiliary diagnostic methods have accomplished the classification task, but few of them can accurately evaluate the changing characteristics of brain connectivity. In this work, a prior-guided adversarial learning with hypergraph (PALH) model is proposed to predict abnormal brain connections using triple-modality medical images. Concretely, a prior distribution from anatomical knowledge is estimated to guide multimodal representation learning using an adversarial strategy. Also, the pairwise collaborative discriminator structure is further utilized to narrow the difference in representation distribution. Moreover, the hypergraph perceptual network is developed to effectively fuse the learned representations while establishing high-order relations within and between multimodal images. Experimental results demonstrate that the proposed model outperforms other related methods in analyzing and predicting AD progression. More importantly, the identified abnormal connections are partly consistent with previous neuroscience discoveries. The proposed model can evaluate the characteristics of abnormal brain connections at different stages of AD, which is helpful for cognitive disease study and early treatment.
Collapse
|
6
|
DeCarli C, Maillard P, Pase MP, Beiser AS, Kojis D, Satizabal CL, Himali JJ, Aparicio HJ, Fletcher E, Seshadri S. Trends in Intracranial and Cerebral Volumes of Framingham Heart Study Participants Born 1930 to 1970. JAMA Neurol 2024; 81:471-480. [PMID: 38526486 PMCID: PMC10964161 DOI: 10.1001/jamaneurol.2024.0469] [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: 11/14/2023] [Accepted: 01/05/2024] [Indexed: 03/26/2024]
Abstract
Importance Human brain development and maintenance is under both genetic and environmental influences that likely affect later-life dementia risk. Objective To examine environmental influences by testing whether time-dependent secular differences occurred in cranial and brain volumes and cortical thickness over birth decades spanning 1930 to 1970. Design, Setting, and Participants This cross-sectional study used data from the community-based Framingham Heart Study cohort for participants born in the decades 1930 to 1970. Participants did not have dementia or history of stroke and had magnetic resonance imaging (MRI) obtained from March 18, 1999, to November 15, 2019. The final analysis dataset was created in October 2023. Exposure Years of birth ranging from 1925 to 1968. Main Measures Cross-sectional analysis of intracranial, cortical gray matter, white matter, and hippocampal volumes as well as cortical surface area and cortical thickness. The secular measure was the decade in which the participant was born. Covariates included age at MRI and sex. Results The main study cohort consisted of 3226 participants with a mean (SD) age of 57.7 (7.8) years at the time of their MRI. A total of 1706 participants were female (53%) and 1520 (47%) were male. The birth decades ranged from the 1930s to 1970s. Significant trends for larger intracranial, hippocampal, and white matter volumes and cortical surface area were associated with progressive birth decades. Comparing the 1930s birth decade to the 1970s accounted for a 6.6% greater volume (1234 mL; 95% CI, 1220-1248, vs 1321 mL; 95% CI, 1301-1341) for ICV, 7.7% greater volume (441.9 mL; 95% CI, 435.2-448.5, vs 476.3 mL; 95% CI, 467.0-485.7) for white matter, 5.7% greater value (6.51 mL; 95% CI, 6.42-6.60, vs 6.89 mL; 95% CI, 6.77-7.02) for hippocampal volume, and a 14.9% greater value (1933 cm2; 95% CI, 1908-1959, vs 2222 cm2; 95% CI, 2186-2259) for cortical surface area. Repeat analysis applied to a subgroup of 1145 individuals of similar age range born in the 1940s (mean [SD] age, 60.0 [2.8] years) and 1950s (mean [SD] age, 59.0 [2.8] years) resulted in similar findings. Conclusion and Relevance In this study, secular trends for larger brain volumes suggested improved brain development among individuals born between 1930 and 1970. Early life environmental influences may explain these results and contribute to the declining dementia incidence previously reported in the Framingham Heart Study cohort.
Collapse
Affiliation(s)
- Charles DeCarli
- Department of Neurology & Imaging of Dementia and Aging Laboratory, University of California Davis, Sacramento, California
| | - Pauline Maillard
- Department of Neurology & Imaging of Dementia and Aging Laboratory, University of California Davis, Sacramento, California
| | - Matthew P. Pase
- Framingham Heart Study, Framingham, Massachusetts
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Alexa S. Beiser
- Framingham Heart Study, Framingham, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
- Department of Neurology, Boston University Chonbanian and Avedisian School of Medicine, Boston, Massachusetts
| | - Daniel Kojis
- Framingham Heart Study, Framingham, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Claudia L. Satizabal
- Framingham Heart Study, Framingham, Massachusetts
- The Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, Texas
| | - Jayandra J. Himali
- Framingham Heart Study, Framingham, Massachusetts
- The Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, Texas
| | - Hugo J. Aparicio
- Framingham Heart Study, Framingham, Massachusetts
- Department of Neurology, Boston University Chonbanian and Avedisian School of Medicine, Boston, Massachusetts
| | - Evan Fletcher
- Department of Neurology & Imaging of Dementia and Aging Laboratory, University of California Davis, Sacramento, California
| | - Sudha Seshadri
- Framingham Heart Study, Framingham, Massachusetts
- Department of Neurology, Boston University Chonbanian and Avedisian School of Medicine, Boston, Massachusetts
- The Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio
| |
Collapse
|
7
|
Garrido-Martín D, Calvo M, Reverter F, Guigó R. A fast non-parametric test of association for multiple traits. Genome Biol 2023; 24:230. [PMID: 37828616 PMCID: PMC10571397 DOI: 10.1186/s13059-023-03076-8] [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: 04/25/2022] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
The increasing availability of multidimensional phenotypic data in large cohorts of genotyped individuals requires efficient methods to identify genetic effects on multiple traits. Permutational multivariate analysis of variance (PERMANOVA) offers a powerful non-parametric approach. However, it relies on permutations to assess significance, which hinders the analysis of large datasets. Here, we derive the limiting null distribution of the PERMANOVA test statistic, providing a framework for the fast computation of asymptotic p values. Our asymptotic test presents controlled type I error and high power, often outperforming parametric approaches. We illustrate its applicability in the context of QTL mapping and GWAS.
Collapse
Affiliation(s)
- Diego Garrido-Martín
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona (UB), Av. Diagonal 643, Barcelona, 08028, Spain.
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona, 08003, Catalonia, Spain.
| | - Miquel Calvo
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona (UB), Av. Diagonal 643, Barcelona, 08028, Spain
| | - Ferran Reverter
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona (UB), Av. Diagonal 643, Barcelona, 08028, Spain
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona, 08003, Catalonia, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| |
Collapse
|
8
|
Rajabli F, Benchek P, Tosto G, Kushch N, Sha J, Bazemore K, Zhu C, Lee WP, Haut J, Hamilton-Nelson KL, Wheeler NR, Zhao Y, Farrell JJ, Grunin MA, Leung YY, Kuksa PP, Li D, Lucio da Fonseca E, Mez JB, Palmer EL, Pillai J, Sherva RM, Song YE, Zhang X, Iqbal T, Pathak O, Valladares O, Kuzma AB, Abner E, Adams PM, Aguirre A, Albert MS, Albin RL, Allen M, Alvarez L, Apostolova LG, Arnold SE, Asthana S, Atwood CS, Ayres G, Baldwin CT, Barber RC, Barnes LL, Barral S, Beach TG, Becker JT, Beecham GW, Beekly D, Benitez BA, Bennett D, Bertelson J, Bird TD, Blacker D, Boeve BF, Bowen JD, Boxer A, Brewer J, Burke JR, Burns JM, Buxbaum JD, Cairns NJ, Cantwell LB, Cao C, Carlson CS, Carlsson CM, Carney RM, Carrasquillo MM, Chasse S, Chesselet MF, Chin NA, Chui HC, Chung J, Craft S, Crane PK, Cribbs DH, Crocco EA, Cruchaga C, Cuccaro ML, Cullum M, Darby E, Davis B, De Jager PL, DeCarli C, DeToledo J, Dick M, Dickson DW, Dombroski BA, Doody RS, Duara R, Ertekin-Taner NI, Evans DA, Faber KM, Fairchild TJ, Fallon KB, Fardo DW, Farlow MR, Fernandez-Hernandez V, Ferris S, Foroud TM, Frosch MP, et alRajabli F, Benchek P, Tosto G, Kushch N, Sha J, Bazemore K, Zhu C, Lee WP, Haut J, Hamilton-Nelson KL, Wheeler NR, Zhao Y, Farrell JJ, Grunin MA, Leung YY, Kuksa PP, Li D, Lucio da Fonseca E, Mez JB, Palmer EL, Pillai J, Sherva RM, Song YE, Zhang X, Iqbal T, Pathak O, Valladares O, Kuzma AB, Abner E, Adams PM, Aguirre A, Albert MS, Albin RL, Allen M, Alvarez L, Apostolova LG, Arnold SE, Asthana S, Atwood CS, Ayres G, Baldwin CT, Barber RC, Barnes LL, Barral S, Beach TG, Becker JT, Beecham GW, Beekly D, Benitez BA, Bennett D, Bertelson J, Bird TD, Blacker D, Boeve BF, Bowen JD, Boxer A, Brewer J, Burke JR, Burns JM, Buxbaum JD, Cairns NJ, Cantwell LB, Cao C, Carlson CS, Carlsson CM, Carney RM, Carrasquillo MM, Chasse S, Chesselet MF, Chin NA, Chui HC, Chung J, Craft S, Crane PK, Cribbs DH, Crocco EA, Cruchaga C, Cuccaro ML, Cullum M, Darby E, Davis B, De Jager PL, DeCarli C, DeToledo J, Dick M, Dickson DW, Dombroski BA, Doody RS, Duara R, Ertekin-Taner NI, Evans DA, Faber KM, Fairchild TJ, Fallon KB, Fardo DW, Farlow MR, Fernandez-Hernandez V, Ferris S, Foroud TM, Frosch MP, Fulton-Howard B, Galasko DR, Gamboa A, Gearing M, Geschwind DH, Ghetti B, Gilbert JR, Goate AM, Grabowski TJ, Graff-Radford NR, Green RC, Growdon JH, Hakonarson H, Hall J, Hamilton RL, Harari O, Hardy J, Harrell LE, Head E, Henderson VW, Hernandez M, Hohman T, Honig LS, Huebinger RM, Huentelman MJ, Hulette CM, Hyman BT, Hynan LS, Ibanez L, Jarvik GP, Jayadev S, Jin LW, Johnson K, Johnson L, Kamboh MI, Karydas AM, Katz MJ, Kauwe JS, Kaye JA, Keene CD, Khaleeq A, Kim R, Knebl J, Kowall NW, Kramer JH, Kukull WA, LaFerla FM, Lah JJ, Larson EB, Lerner A, Leverenz JB, Levey AI, Lieberman AP, Lipton RB, Logue M, Lopez OL, Lunetta KL, Lyketsos CG, Mains D, Margaret FE, Marson DC, Martin ERR, Martiniuk F, Mash DC, Masliah E, Massman P, Masurkar A, McCormick WC, McCurry SM, McDavid AN, McDonough S, McKee AC, Mesulam M, Miller BL, Miller CA, Miller JW, Montine TJ, Monuki ES, Morris JC, Mukherjee S, Myers AJ, Nguyen T, O'Bryant S, Olichney JM, Ory M, Palmer R, Parisi JE, Paulson HL, Pavlik V, Paydarfar D, Perez V, Peskind E, Petersen RC, Pierce A, Polk M, Poon WW, Potter H, Qu L, Quiceno M, Quinn JF, Raj A, Raskind M, Reiman EM, Reisberg B, Reisch JS, Ringman JM, Roberson ED, Rodriguear M, Rogaeva E, Rosen HJ, Rosenberg RN, Royall DR, Sager MA, Sano M, Saykin AJ, Schneider JA, Schneider LS, Seeley WW, Slifer SH, Small S, Smith AG, Smith JP, Sonnen JA, Spina S, St George-Hyslop P, Stern RA, Stevens AB, Strittmatter SM, Sultzer D, Swerdlow RH, Tanzi RE, Tilson JL, Trojanowski JQ, Troncoso JC, Tsuang DW, Van Deerlin VM, van Eldik LJ, Vance JM, Vardarajan BN, Vassar R, Vinters HV, Vonsattel JP, Weintraub S, Welsh-Bohmer KA, Whitehead PL, Wijsman EM, Wilhelmsen KC, Williams B, Williamson J, Wilms H, Wingo TS, Wisniewski T, Woltjer RL, Woon M, Wright CB, Wu CK, Younkin SG, Yu CE, Yu L, Zhu X, Kunkle BW, Bush WS, Wang LS, Farrer LA, Haines JL, Mayeux R, Pericak-Vance MA, Schellenberg GD, Jun GR, Reitz C, Naj AC. Multi-ancestry genome-wide meta-analysis of 56,241 individuals identifies LRRC4C, LHX5-AS1 and nominates ancestry-specific loci PTPRK , GRB14 , and KIAA0825 as novel risk loci for Alzheimer's disease: the Alzheimer's Disease Genetics Consortium. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.06.23292311. [PMID: 37461624 PMCID: PMC10350126 DOI: 10.1101/2023.07.06.23292311] [Show More Authors] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Limited ancestral diversity has impaired our ability to detect risk variants more prevalent in non-European ancestry groups in genome-wide association studies (GWAS). We constructed and analyzed a multi-ancestry GWAS dataset in the Alzheimer's Disease (AD) Genetics Consortium (ADGC) to test for novel shared and ancestry-specific AD susceptibility loci and evaluate underlying genetic architecture in 37,382 non-Hispanic White (NHW), 6,728 African American, 8,899 Hispanic (HIS), and 3,232 East Asian individuals, performing within-ancestry fixed-effects meta-analysis followed by a cross-ancestry random-effects meta-analysis. We identified 13 loci with cross-ancestry associations including known loci at/near CR1 , BIN1 , TREM2 , CD2AP , PTK2B , CLU , SHARPIN , MS4A6A , PICALM , ABCA7 , APOE and two novel loci not previously reported at 11p12 ( LRRC4C ) and 12q24.13 ( LHX5-AS1 ). Reflecting the power of diverse ancestry in GWAS, we observed the SHARPIN locus using 7.1% the sample size of the original discovering single-ancestry GWAS (n=788,989). We additionally identified three GWS ancestry-specific loci at/near ( PTPRK ( P =2.4×10 -8 ) and GRB14 ( P =1.7×10 -8 ) in HIS), and KIAA0825 ( P =2.9×10 -8 in NHW). Pathway analysis implicated multiple amyloid regulation pathways (strongest with P adjusted =1.6×10 -4 ) and the classical complement pathway ( P adjusted =1.3×10 -3 ). Genes at/near our novel loci have known roles in neuronal development ( LRRC4C, LHX5-AS1 , and PTPRK ) and insulin receptor activity regulation ( GRB14 ). These findings provide compelling support for using traditionally-underrepresented populations for gene discovery, even with smaller sample sizes.
Collapse
|
9
|
Liu N, Zhang L, Tian T, Cheng J, Zhang B, Qiu S, Geng Z, Cui G, Zhang Q, Liao W, Yu Y, Zhang H, Gao B, Xu X, Han T, Yao Z, Qin W, Liu F, Liang M, Xu Q, Fu J, Xu J, Zhu W, Zhang P, Li W, Shi D, Wang C, Lui S, Yan Z, Chen F, Li J, Zhang J, Wang D, Shen W, Miao Y, Xian J, Gao JH, Zhang X, Li MJ, Xu K, Zuo XN, Wang M, Ye Z, Yu C. Cross-ancestry genome-wide association meta-analyses of hippocampal and subfield volumes. Nat Genet 2023:10.1038/s41588-023-01425-8. [PMID: 37337106 DOI: 10.1038/s41588-023-01425-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/11/2023] [Indexed: 06/21/2023]
Abstract
The hippocampus is critical for memory and cognition and neuropsychiatric disorders, and its subfields differ in architecture and function. Genome-wide association studies on hippocampal and subfield volumes are mainly conducted in European populations; however, other ancestral populations are under-represented. Here we conduct cross-ancestry genome-wide association meta-analyses in 65,791 individuals for hippocampal volume and 38,977 for subfield volumes, including 7,009 individuals of East Asian ancestry. We identify 339 variant-trait associations at P < 1.13 × 10-9 for 44 hippocampal traits, including 23 new associations. Common genetic variants have similar effects on hippocampal traits across ancestries, although ancestry-specific associations exist. Cross-ancestry analysis improves the fine-mapping precision and the prediction performance of polygenic scores in under-represented populations. These genetic variants are enriched for Wnt signaling and neuron differentiation and affect cognition, emotion and neuropsychiatric disorders. These findings may provide insight into the genetic architectures of hippocampal and subfield volumes.
Collapse
Affiliation(s)
- Nana Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Longjiang Zhang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tian Tian
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bing Zhang
- Department of Radiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shijun Qiu
- Department of Medical Imaging, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Zuojun Geng
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guangbin Cui
- Functional and Molecular Imaging Key Lab of Shaanxi Province & Department of Radiology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Quan Zhang
- Department of Radiology, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- Molecular Imaging Research Center of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Zhang
- Department of Radiology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Bo Gao
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Radiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wen Qin
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Liang
- School of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, China
| | - Qiang Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Jilian Fu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiayuan Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wei Li
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Dapeng Shi
- Department of Radiology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China
| | - Caihong Wang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Su Lui
- Department of Radiology, Huaxi MR Research Center (HMRRC), Functional and Molecular lmaging key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| | - Zhihan Yan
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feng Chen
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Jiance Li
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Zhang
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wen Shen
- Department of Radiology, Tianjin First Center Hospital, Tianjin, China
| | - Yanwei Miao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xiaochu Zhang
- Division of Life Science and Medicine, University of Science & Technology of China, Hefei, China
| | - Mulin Jun Li
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Kai Xu
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xi-Nian Zuo
- Developmental Population Neuroscience Research Center at IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China.
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
10
|
Watts R, Rader L, Grant J, Filippi CG. Genetic and Environmental Contributions to Subcortical Gray Matter Microstructure and Volume in the Developing Brain. Behav Genet 2023; 53:208-218. [PMID: 37129746 PMCID: PMC10154259 DOI: 10.1007/s10519-023-10142-1] [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: 09/29/2022] [Accepted: 04/12/2023] [Indexed: 05/03/2023]
Abstract
Using baseline (ages 9-10) and two-year follow-up (ages 11-12) data from monozygotic and dizygotic twins enrolled in the longitudinal Adolescent Brain Cognitive DevelopmentSM Study, we investigated the genetic and environmental contributions to microstructure and volume of nine subcortical gray matter regions. Microstructure was assessed using diffusion MRI data analyzed using restriction spectrum imaging (RSI) and diffusion tensor imaging (DTI) models. The highest heritability estimates (estimate [95% confidence interval]) for microstructure were found using the RSI model in the pallidum (baseline: 0.859 [0.818, 0.889], follow-up: 0.835 [0.787, 0.871]), putamen (baseline: 0.859 [0.819, 0.889], follow-up: 0.874 [0.838, 0.902]), and thalamus (baseline: 0.855 [0.814, 0.887], follow-up: 0.819 [0.769, 0.857]). For volumes the corresponding regions were the caudate (baseline: 0.831 [0.688, 0.992], follow-up: 0.848 [0.701, 1.011]) and putamen (baseline: 0.906 [0.875, 0.914], follow-up: 0.906 [0.885, 0.923]). The subcortical regions displayed high genetic stability (rA = 0.743-1.000) across time and exhibited unique environmental correlations (rE = 0.194-0.610). Individual differences in both gray matter microstructure and volumes can be largely explained by additive genetic effects in this sample.
Collapse
Affiliation(s)
- Richard Watts
- Department of Psychology, Yale University, 2 Hillhouse Avenue, New Haven, CT, 06520, USA.
| | - Lydia Rader
- Institute for Behavioral Genetics, Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Justin Grant
- Department of Radiology, Tufts University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
11
|
Tissink E, Werme J, de Lange SC, Savage JE, Wei Y, de Leeuw CA, Nagel M, Posthuma D, van den Heuvel MP. The Genetic Architectures of Functional and Structural Connectivity Properties within Cerebral Resting-State Networks. eNeuro 2023; 10:ENEURO.0242-22.2023. [PMID: 36882310 PMCID: PMC10089056 DOI: 10.1523/eneuro.0242-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/12/2022] [Accepted: 01/08/2023] [Indexed: 03/09/2023] Open
Abstract
Functional connectivity within resting-state networks (RSN-FC) is vital for cognitive functioning. RSN-FC is heritable and partially translates to the anatomic architecture of white matter, but the genetic component of structural connections of RSNs (RSN-SC) and their potential genetic overlap with RSN-FC remain unknown. Here, we perform genome-wide association studies (N discovery = 24,336; N replication = 3412) and annotation on RSN-SC and RSN-FC. We identify genes for visual network-SC that are involved in axon guidance and synaptic functioning. Genetic variation in RSN-FC impacts biological processes relevant to brain disorders that previously were only phenotypically associated with RSN-FC alterations. Correlations of the genetic components of RSNs are mostly observed within the functional domain, whereas less overlap is observed within the structural domain and between the functional and structural domains. This study advances the understanding of the complex functional organization of the brain and its structural underpinnings from a genetics viewpoint.
Collapse
Affiliation(s)
- Elleke Tissink
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands
| | - Josefin Werme
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands
| | - Siemon C de Lange
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, Amsterdam 1105 BA, The Netherlands
| | - Jeanne E Savage
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands
| | - Yongbin Wei
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing 100876, China
| | - Christiaan A de Leeuw
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands
| | - Mats Nagel
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands
- Department of Clinical Genetics, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam University Medical Centre, Amsterdam 1081 HZ, The Netherlands
| | - Martijn P van den Heuvel
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands
- Department of Clinical Genetics, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam University Medical Centre, Amsterdam 1081 HZ, The Netherlands
| |
Collapse
|
12
|
DeCarli C, Pase M, Beiser A, Kojis D, Satizabal C, Himali J, Aparicio H, Flether E, Maillard P, Seshadri S. Secular Trends in Head Size and Cerebral Volumes In the Framingham Heart Study for Birth Years 1902-1985. RESEARCH SQUARE 2023:rs.3.rs-2524684. [PMID: 36778357 PMCID: PMC9915799 DOI: 10.21203/rs.3.rs-2524684/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background Recent data suggest that dementia incidence is declining. We investigated whether similar secular trends consisting of increasing size of brain structures and improving memory performance could be simultaneously occurring as a possible explanation. Method The Framingham Heart Study is a 3 generation, longitudinal study that includes cognitive assessment and medical surveillance. This study cohort consisted of 4,506 unique, non-demented, stroke free, individuals with brain MRI, cognitive assessment, and demographic information spanning dates of birth from 1902 to 1985. Outcomes consisted of height, MRI, and memory measures. Covariates included age at MRI, sex, decade of birth, and all interactions. Models with neuropsychological outcomes also included educational achievement as a covariate. Results Height and intracranial (TCV), hippocampus and cortical gray matter volumes were significantly larger, and memory performance significantly better, with advancing decades of birth after adjusting for age, sex, and interactions. Sensitivity analysis using progressively restricted age-ranges to reduce the association between age and decade of birth, confirmed the findings. Mediation analysis showed that hippocampal volume mediated approximately 5-7% of the effect of decade of birth on logical memory performance. Discussion These findings indicate improvement in brain health and memory performance with advancing decades of birth. Although brain structures are under substantial genetic influence, we conclude that improved early life environmental influences over ensuing decades likely explain these results. We hypothesize that these secular improvements are consistent with declining dementia incidence in this cohort potentially through a mechanism of increased brain reserve.
Collapse
Affiliation(s)
| | | | - Alexa Beiser
- Department of Biostatistics, Boston University School of Public Health
| | | | - Claudia Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases and Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX
| | - Jayandra Himali
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases and Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | | | | | | | | |
Collapse
|
13
|
Bernstein HG, Keilhoff G, Dobrowolny H, Steiner J. The many facets of CD26/dipeptidyl peptidase 4 and its inhibitors in disorders of the CNS - a critical overview. Rev Neurosci 2023; 34:1-24. [PMID: 35771831 DOI: 10.1515/revneuro-2022-0026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/10/2022] [Indexed: 01/11/2023]
Abstract
Dipeptidyl peptidase 4 is a serine protease that cleaves X-proline or X-alanine in the penultimate position. Natural substrates of the enzyme are glucagon-like peptide-1, glucagon inhibiting peptide, glucagon, neuropeptide Y, secretin, substance P, pituitary adenylate cyclase-activating polypeptide, endorphins, endomorphins, brain natriuretic peptide, beta-melanocyte stimulating hormone and amyloid peptides as well as some cytokines and chemokines. The enzyme is involved in the maintenance of blood glucose homeostasis and regulation of the immune system. It is expressed in many organs including the brain. DPP4 activity may be effectively depressed by DPP4 inhibitors. Apart from enzyme activity, DPP4 acts as a cell surface (co)receptor, associates with adeosine deaminase, interacts with extracellular matrix, and controls cell migration and differentiation. This review aims at revealing the impact of DPP4 and DPP4 inhibitors for several brain diseases (virus infections affecting the brain, tumours of the CNS, neurological and psychiatric disorders). Special emphasis is given to a possible involvement of DPP4 expressed in the brain.While prominent contributions of extracerebral DPP4 are evident for a majority of diseases discussed herein; a possible role of "brain" DPP4 is restricted to brain cancers and Alzheimer disease. For a number of diseases (Covid-19 infection, type 2 diabetes, Alzheimer disease, vascular dementia, Parkinson disease, Huntington disease, multiple sclerosis, stroke, and epilepsy), use of DPP4 inhibitors has been shown to have a disease-mitigating effect. However, these beneficial effects should mostly be attributed to the depression of "peripheral" DPP4, since currently used DPP4 inhibitors are not able to pass through the intact blood-brain barrier.
Collapse
Affiliation(s)
- Hans-Gert Bernstein
- Department of Psychiatry and Psychotherapy, Otto v. Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto v. Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry and Psychotherapy, Otto v. Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Johann Steiner
- Department of Psychiatry and Psychotherapy, Otto v. Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| |
Collapse
|
14
|
Jia Y, Cheng S, Liu L, Cheng B, Liang C, Ye J, Chu X, Yao Y, Wen Y, Kafle OP, Zhang F. Evaluating the Genetic Effects of Gut Microbiota on the Development of Neuroticism and General Happiness: A Polygenic Score Analysis and Interaction Study Using UK Biobank Data. Genes (Basel) 2023; 14:156. [PMID: 36672898 PMCID: PMC9858947 DOI: 10.3390/genes14010156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Limited efforts have been invested in exploring the interaction effects between genetic factors and gut microbiota on neuroticism and general happiness. The polygenic risk scores (PRS) of gut microbiota were calculated from individual-level genotype data of the UK Biobank cohort. Linear regression models were then used to assess the associations between individual PRS of gut microbiota and mental traits and interaction analysis was performed by PLINK2.0. KOBAS-i was used to conduct gene ontology (GO) enrichment analysis of the identified genes. We observed suggestive significant associations between neuroticism and PRS for the genus Bifidobacterium (rank-normal transformation, RNT) (beta = -1.10, P = 4.16 × 10-3) and the genus Desulfovibrio (RNT) (beta = 0.54, P = 7.46 × 10-3). PRS for the genus Bifidobacterium (hurdle binary, HB) (beta = 1.99, P = 5.24 × 10-3) and the genus Clostridium (RNT) (beta = 1.26, P = 9.27 × 10-3) were found to be suggestive positively associated with general happiness. Interaction analysis identified several significant genes that interacted with gut microbiota, such as RORA (rs575949009, beta = -45.00, P = 1.82 × 10-9) for neuroticism and ASTN2 (rs36005728, beta = 19.15, P = 3.37 × 10-8) for general happiness. Our study results support the genetic effects of gut microbiota on the development of neuroticism and general happiness.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
15
|
Küçükali F, Neumann A, Van Dongen J, De Pooter T, Joris G, De Rijk P, Ohlei O, Dobricic V, Bos I, Vos SJB, Engelborghs S, De Roeck E, Vandenberghe R, Gabel S, Meersmans K, Tsolaki M, Verhey F, Martinez‐Lage P, Tainta M, Frisoni G, Blin O, Richardson JC, Bordet R, Scheltens P, Popp J, Peyratout G, Johannsen P, Frölich L, Freund‐Levi Y, Streffer J, Lovestone S, Legido‐Quigley C, Kate MT, Barkhof F, Zetterberg H, Bertram L, Strazisar M, Visser PJ, Van Broeckhoven C, Sleegers K, Alzheimer's Disease Neuroimaging Initiative (ADNI), the EMIF‐AD Study Group. Whole‐exome rare‐variant analysis of Alzheimer's disease and related biomarker traits. Alzheimers Dement 2022. [DOI: 10.1002/alz.12842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 09/16/2022] [Accepted: 09/28/2022] [Indexed: 12/08/2022]
|
16
|
Ciobanu LG, Stankov L, Schubert KO, Amare AT, Jawahar MC, Lawrence-Wood E, Mills NT, Knight M, Clark SR, Aidman E. General intelligence and executive functioning are overlapping but separable at genetic and molecular pathway levels: An analytical review of existing GWAS findings. PLoS One 2022; 17:e0272368. [PMID: 36251633 PMCID: PMC9576059 DOI: 10.1371/journal.pone.0272368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/18/2022] [Indexed: 11/05/2022] Open
Abstract
Understanding the genomic architecture and molecular mechanisms of cognitive functioning in healthy individuals is critical for developing tailored interventions to enhance cognitive functioning, as well as for identifying targets for treating impaired cognition. There has been substantial progress in uncovering the genetic composition of the general cognitive ability (g). However, there is an ongoing debate whether executive functioning (EF)–another key predictor of cognitive health and performance, is separable from general g. To provide an analytical review on existing findings on genetic influences on the relationship between g and EF, we re-analysed a subset of genome-wide association studies (GWAS) from the GWAS catalogue that used measures of g and EF as outcomes in non-clinical populations. We identified two sets of single nucleotide polymorphisms (SNPs) associated with g (1,372 SNPs across 12 studies), and EF (300 SNPs across 5 studies) at p<5x10-6. A comparative analysis of GWAS-identified g and EF SNPs in high linkage disequilibrium (LD), followed by pathway enrichment analyses suggest that g and EF are overlapping but separable at genetic variant and molecular pathway levels, however more evidence is required to characterize the genetic overlap/distinction between the two constructs. While not without limitations, these findings may have implications for navigating further research towards translatable genetic findings for cognitive remediation, enhancement, and augmentation.
Collapse
Affiliation(s)
- Liliana G. Ciobanu
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
- * E-mail:
| | - Lazar Stankov
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - K. Oliver Schubert
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
- Northern Adelaide Mental Health Services, Adelaide, SA, Australia
| | - Azmeraw T. Amare
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
- National Health and Medical Research Council (NHMRC) Centre of Research Excellence in Frailty and Healthy Ageing, University of Adelaide, Adelaide, Australia
| | | | | | - Natalie T. Mills
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Matthew Knight
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
- Weapons and Combat Systems Division, Defence Science & Technology Group, Edinburgh, SA, Australia
| | - Scott R. Clark
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Eugene Aidman
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Land Division, Defence Science & Technology Group, Edinburgh, SA, Australia
| |
Collapse
|
17
|
Gonzales MM, Wiedner C, Wang C, Liu Q, Bis JC, Li Z, Himali JJ, Ghosh S, Thomas EA, Parent DM, Kautz TF, Pase MP, Aparicio HJ, Djoussé L, Mukamal KJ, Psaty BM, Longstreth WT, Mosley TH, Gudnason V, Mbangdadji D, Lopez OL, Yaffe K, Sidney S, Bryan RN, Nasrallah IM, DeCarli CS, Beiser AS, Launer LJ, Fornage M, Tracy RP, Seshadri S, Satizabal CL. A population-based meta-analysis of circulating GFAP for cognition and dementia risk. Ann Clin Transl Neurol 2022; 9:1574-1585. [PMID: 36056631 PMCID: PMC9539381 DOI: 10.1002/acn3.51652] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Expression of glial fibrillary acidic protein (GFAP), a marker of reactive astrocytosis, colocalizes with neuropathology in the brain. Blood levels of GFAP have been associated with cognitive decline and dementia status. However, further examinations at a population-based level are necessary to broaden generalizability to community settings. METHODS Circulating GFAP levels were assayed using a Simoa HD-1 analyzer in 4338 adults without prevalent dementia from four longitudinal community-based cohort studies. The associations between GFAP levels with general cognition, total brain volume, and hippocampal volume were evaluated with separate linear regression models in each cohort with adjustment for age, sex, education, race, diabetes, systolic blood pressure, antihypertensive medication, body mass index, apolipoprotein E ε4 status, site, and time between GFAP blood draw and the outcome. Associations with incident all-cause and Alzheimer's disease dementia were evaluated with adjusted Cox proportional hazard models. Meta-analysis was performed on the estimates derived from each cohort using random-effects models. RESULTS Meta-analyses indicated that higher circulating GFAP associated with lower general cognition (ß = -0.09, [95% confidence interval [CI]: -0.15 to -0.03], p = 0.005), but not with total brain or hippocampal volume (p > 0.05). However, each standard deviation unit increase in log-transformed GFAP levels was significantly associated with a 2.5-fold higher risk of incident all-cause dementia (Hazard Ratio [HR]: 2.47 (95% CI: 1.52-4.01)) and Alzheimer's disease dementia (HR: 2.54 [95% CI: 1.42-4.53]) over up to 15-years of follow-up. INTERPRETATION Results support the potential role of circulating GFAP levels for aiding dementia risk prediction and improving clinical trial stratification in community settings.
Collapse
Affiliation(s)
- Mitzi M. Gonzales
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Department of NeurologyUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Crystal Wiedner
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Chen‐Pin Wang
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Department of Population Health SciencesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- South Texas Veterans Health Care System, Geriatric ResearchEducation & Clinical CenterSan AntonioTexasUSA
| | - Qianqian Liu
- Department of Population Health SciencesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Joshua C. Bis
- Cardiovascular Health Research UnitUniversity of WashingtonSeattleWashingtonUSA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, Intramural Research ProgramNational Institute on AgingBethesdaMarylandUSA
| | - Jayandra J. Himali
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Department of Population Health SciencesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- The Framingham Heart StudyFraminghamMassachusettsUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of MedicineBostonMassachusettsUSA
| | - Saptaparni Ghosh
- The Framingham Heart StudyFraminghamMassachusettsUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| | - Emy A. Thomas
- Brown Foundation of Molecular Medicine, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Danielle M. Parent
- Department of Pathology and Laboratory Medicine, and Biochemistry, Larner College of MedicineUniversity of VermontBurlingtonVermontUSA
| | - Tiffany F. Kautz
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Matthew P. Pase
- The Framingham Heart StudyFraminghamMassachusettsUSA
- School of Psychological Sciences, Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
- Harvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Hugo J. Aparicio
- The Framingham Heart StudyFraminghamMassachusettsUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| | - Luc Djoussé
- Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Boston Veterans Affairs Healthcare SystemBostonMassachusettsUSA
| | - Kenneth J. Mukamal
- Department of MedicineBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Bruce M. Psaty
- Cardiovascular Health Research UnitUniversity of WashingtonSeattleWashingtonUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
- Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
- Department of Health Systems and Population HealthUniversity of WashingtonSeattleWashingtonUSA
| | - William T. Longstreth
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
| | - Thomas H. Mosley
- The MIND CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Vilmundur Gudnason
- Icelandic Heart Association Research InstituteKópavogurIceland
- Department of CardiologyUniversity of IcelandReykjavikIceland
| | - Djass Mbangdadji
- Laboratory of Epidemiology and Population Sciences, Intramural Research ProgramNational Institute on AgingBethesdaMarylandUSA
| | - Oscar L. Lopez
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Kristine Yaffe
- Department of PsychiatryUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCaliforniaUSA
- San Francisco VA Medical CenterSan FranciscoCaliforniaUSA
| | - Stephen Sidney
- Kaiser Permanente Medical Center ProgramOaklandCaliforniaUSA
| | - R. Nick Bryan
- Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ilya M. Nasrallah
- Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Alexa S. Beiser
- The Framingham Heart StudyFraminghamMassachusettsUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of MedicineBostonMassachusettsUSA
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research ProgramNational Institute on AgingBethesdaMarylandUSA
| | - Myriam Fornage
- Brown Foundation of Molecular Medicine, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, and Biochemistry, Larner College of MedicineUniversity of VermontBurlingtonVermontUSA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Department of NeurologyUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- The Framingham Heart StudyFraminghamMassachusettsUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Department of Population Health SciencesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- The Framingham Heart StudyFraminghamMassachusettsUSA
| |
Collapse
|
18
|
Mai H, Bao J, Thompson PM, Kim D, Shen L. Identifying genes associated with brain volumetric differences through tissue specific transcriptomic inference from GWAS summary data. BMC Bioinformatics 2022; 23:398. [PMID: 36171548 PMCID: PMC9520794 DOI: 10.1186/s12859-022-04947-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Brain volume has been widely studied in the neuroimaging field, since it is an important and heritable trait associated with brain development, aging and various neurological and psychiatric disorders. Genome-wide association studies (GWAS) have successfully identified numerous associations between genetic variants such as single nucleotide polymorphisms and complex traits like brain volume. However, it is unclear how these genetic variations influence regional gene expression levels, which may subsequently lead to phenotypic changes. S-PrediXcan is a tissue-specific transcriptomic data analysis method that can be applied to bridge this gap. In this work, we perform an S-PrediXcan analysis on GWAS summary data from two large imaging genetics initiatives, the UK Biobank and Enhancing Neuroimaging Genetics through Meta Analysis, to identify tissue-specific transcriptomic effects on two closely related brain volume measures: total brain volume (TBV) and intracranial volume (ICV). RESULTS As a result of the analysis, we identified 10 genes that are highly associated with both TBV and ICV. Nine out of 10 genes were found to be associated with TBV in another study using a different gene-based association analysis. Moreover, most of our discovered genes were also found to be correlated with multiple cognitive and behavioral traits. Further analyses revealed the protein-protein interactions, associated molecular pathways and biological functions that offer insight into how these genes function and interact with others. CONCLUSIONS These results confirm that S-PrediXcan can identify genes with tissue-specific transcriptomic effects on complex traits. The analysis also suggested novel genes whose expression levels are related to brain volumetric traits. This provides important insights into the genetic mechanisms of the human brain.
Collapse
Affiliation(s)
- Hung Mai
- Perelman School of Medicine, University of Pennsylvania, B306 Richards Building, 3700 Hamilton Walk, Philadelphia, PA, USA
- School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Perelman School of Medicine, University of Pennsylvania, B306 Richards Building, 3700 Hamilton Walk, Philadelphia, PA, USA
- School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Institute for Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dokyoon Kim
- Perelman School of Medicine, University of Pennsylvania, B306 Richards Building, 3700 Hamilton Walk, Philadelphia, PA, USA
| | - Li Shen
- Perelman School of Medicine, University of Pennsylvania, B306 Richards Building, 3700 Hamilton Walk, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Liu M, Yu C, Zhang Z, Song M, Sun X, Piálek J, Jacob J, Lu J, Cong L, Zhang H, Wang Y, Li G, Feng Z, Du Z, Wang M, Wan X, Wang D, Wang YL, Li H, Wang Z, Zhang B, Zhang Z. Whole-genome sequencing reveals the genetic mechanisms of domestication in classical inbred mice. Genome Biol 2022; 23:203. [PMID: 36163035 PMCID: PMC9511766 DOI: 10.1186/s13059-022-02772-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background The laboratory mouse was domesticated from the wild house mouse. Understanding the genetics underlying domestication in laboratory mice, especially in the widely used classical inbred mice, is vital for studies using mouse models. However, the genetic mechanism of laboratory mouse domestication remains unknown due to lack of adequate genomic sequences of wild mice. Results We analyze the genetic relationships by whole-genome resequencing of 36 wild mice and 36 inbred strains. All classical inbred mice cluster together distinctly from wild and wild-derived inbred mice. Using nucleotide diversity analysis, Fst, and XP-CLR, we identify 339 positively selected genes that are closely associated with nervous system function. Approximately one third of these positively selected genes are highly expressed in brain tissues, and genetic mouse models of 125 genes in the positively selected genes exhibit abnormal behavioral or nervous system phenotypes. These positively selected genes show a higher ratio of differential expression between wild and classical inbred mice compared with all genes, especially in the hippocampus and frontal lobe. Using a mutant mouse model, we find that the SNP rs27900929 (T>C) in gene Astn2 significantly reduces the tameness of mice and modifies the ratio of the two Astn2 (a/b) isoforms. Conclusion Our study indicates that classical inbred mice experienced high selection pressure during domestication under laboratory conditions. The analysis shows the positively selected genes are closely associated with behavior and the nervous system in mice. Tameness may be related to the Astn2 mutation and regulated by the ratio of the two Astn2 (a/b) isoforms. Supplementary Information The online version contains supplementary material available at 10.1186/s13059-022-02772-1.
Collapse
Affiliation(s)
- Ming Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,International Society of Zoological Sciences, Beijing, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Caixia Yu
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.,National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Zhichao Zhang
- Novogene Bioinformatics Institute, Beijing, China.,Glbizzia Biosciences, Beijing, China
| | - Mingjing Song
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiuping Sun
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing, China
| | - Jaroslav Piálek
- House Mouse Group, Research Facility Studenec, Institute of Vertebrate Biology of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jens Jacob
- Julius Kühn-Institute, Federal Research Centre for Cultivated Plants, Institute for Plant Protection in Horticulture and Forests / Institute for Epidemiology and Pathogen Diagnostics, Münster, Germany
| | - Jiqi Lu
- School of Life Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Cong
- Institute of Plant Protection, Heilongjiang Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Hongmao Zhang
- School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Yong Wang
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Guoliang Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyong Feng
- Plant Protection Research Institute Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Zhenglin Du
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.,National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Meng Wang
- Novogene Bioinformatics Institute, Beijing, China
| | - Xinru Wan
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Dawei Wang
- Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hongjun Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306, USA
| | - Bing Zhang
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China. .,International Society of Zoological Sciences, Beijing, China. .,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
20
|
Janahi M, Aksman L, Schott JM, Mokrab Y, Altmann A. Nomograms of human hippocampal volume shifted by polygenic scores. eLife 2022; 11:e78232. [PMID: 35938915 PMCID: PMC9391046 DOI: 10.7554/elife.78232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022] Open
Abstract
Nomograms are important clinical tools applied widely in both developing and aging populations. They are generally constructed as normative models identifying cases as outliers to a distribution of healthy controls. Currently used normative models do not account for genetic heterogeneity. Hippocampal volume (HV) is a key endophenotype for many brain disorders. Here, we examine the impact of genetic adjustment on HV nomograms and the translational ability to detect dementia patients. Using imaging data from 35,686 healthy subjects aged 44-82 from the UK Biobank (UKB), we built HV nomograms using Gaussian process regression (GPR), which - compared to a previous method - extended the application age by 20 years, including dementia critical age ranges. Using HV polygenic scores (HV-PGS), we built genetically adjusted nomograms from participants stratified into the top and bottom 30% of HV-PGS. This shifted the nomograms in the expected directions by ~100 mm3 (2.3% of the average HV), which equates to 3 years of normal aging for a person aged ~65. Clinical impact of genetically adjusted nomograms was investigated by comparing 818 subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database diagnosed as either cognitively normal (CN), having mild cognitive impairment (MCI) or Alzheimer's disease (AD) patients. While no significant change in the survival analysis was found for MCI-to-AD conversion, an average of 68% relative decrease was found in intra-diagnostic-group variance, highlighting the importance of genetic adjustment in untangling phenotypic heterogeneity.
Collapse
Affiliation(s)
- Mohammed Janahi
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College LondonLondonUnited Kingdom
- Medical and Population Genomics Lab, Human Genetics Department, Research Branch, Sidra MedicineDohaQatar
| | - Leon Aksman
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Jonathan M Schott
- Dementia Research Centre (DRC), Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Younes Mokrab
- Medical and Population Genomics Lab, Human Genetics Department, Research Branch, Sidra MedicineDohaQatar
- Department of Genetic Medicine, Weill Cornell Medicine-QatarDohaQatar
| | - Andre Altmann
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College LondonLondonUnited Kingdom
| |
Collapse
|
21
|
Yang XZ, Wan MY, Zhang DD, Dai Y, Pan ZA, Zhai FF, Han F, Liu JY, Zhou LX, Ni J, Yao M, Jin ZY, Cui LY, Zhang SY, Zhu YC. Investigating the Genetic Characteristics of Hippocampal Volume and Plasma β-Amyloid in a Chinese Community-Dwelling Population. Neurology 2022; 99:e234-e244. [PMID: 35623891 DOI: 10.1212/wnl.0000000000200554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/02/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The genetic characteristics and correlations of hippocampal volume (HV) and plasma β-amyloid (Aβ), probable endophenotypes for dementia, remain to be explored in a Chinese community cohort. Using whole-exome sequencing (WES) and single nucleotide polymorphism (SNP) array genotyping, we sought to identify rare and common variants and genes influencing these 2 endophenotypes and calculate their heritability and genetic correlation. METHODS Association analyses with both WES and SNP array genotyping data were performed for HV and plasma Aβ with mixed-effect linear regression model adjusted for sex, age, and total intracranial volume or APOE ε4 while considering familial relatedness. We also performed gene-level analysis for common and gene burden analysis for rare variants. Heritability and genetic correlation were examined further. RESULTS A total of 1,261 participants from a Chinese community cohort were included and we identified 1 gene, PTPRT, for HV, with the top significant SNPs by whole genome-wide association study (GWAS). rs6030076 (p = 5.48 × 10-8, β = -0.092, SE 0.017) from WES and rs6030088 (p = 8.24 × 10-9, β = -105.22, SE 18.09) from SNP array data were both located in this gene. Gene burden analysis based on rare mutations detected 6 genes to be significantly associated with Aβ. The SNP-based heritability was 0.43 ± 0.13 for HV and 0.2-0.3 for plasma Aβ. The SNP-based genetic correlation between HV and plasma Aβ was negative. DISCUSSION In this study, we identified several SNPs and 1 gene, PTPRT, which were not reported in previous GWAS, associated with HV. The heritability and the genetic correlation gave an overview of HV and plasma Aβ. Our findings provide insights into the mechanisms behind the individual variances in these endophenotypes.
Collapse
Affiliation(s)
- Xin-Zhuang Yang
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng-Yao Wan
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ding-Ding Zhang
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Dai
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zi-Ang Pan
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei-Fei Zhai
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Han
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Yi Liu
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Xin Zhou
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Ni
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming Yao
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng-Yu Jin
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Ying Cui
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Yang Zhang
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Cheng Zhu
- From the Department of Neurology (X.-Z.Y., M.-Y.W., D.-D.Z., Y.D., Z.-A.P., F.-F.Z., F.H., J.-Y.L., L.-X.Z., J.N., M.Y., L.-Y.C., Y.-C.Z.), Medical Research Center (X.-Z.Y., D.-D.Z.), and Departments of Radiology (Z.-Y.J.) and Cardiology (S.-Y.Z.), State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
22
|
Genetic Specificity of Hippocampal Subfield Volumes, Relative to Hippocampal Formation, Identified in 2148 Young Adult Twins and Siblings. Twin Res Hum Genet 2022; 25:129-139. [PMID: 35791873 DOI: 10.1017/thg.2022.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The hippocampus is a complex brain structure with key roles in cognitive and emotional processing and with subregion abnormalities associated with a range of disorders and psychopathologies. Here we combine data from two large independent young adult twin/sibling cohorts to obtain the most accurate estimates to date of genetic covariation between hippocampal subfield volumes and the hippocampus as a single volume. The combined sample included 2148 individuals, comprising 1073 individuals from 627 families (mean age = 22.3 years) from the Queensland Twin IMaging (QTIM) Study, and 1075 individuals from 454 families (mean age = 28.8 years) from the Human Connectome Project (HCP). Hippocampal subfields were segmented using FreeSurfer version 6.0 (CA4 and dentate gyrus were phenotypically and genetically indistinguishable and were summed to a single volume). Multivariate twin modeling was conducted in OpenMx to decompose variance into genetic and environmental sources. Bivariate analyses of hippocampal formation and each subfield volume showed that 10%-72% of subfield genetic variance was independent of the hippocampal formation, with greatest specificity found for the smaller volumes; for example, CA2/3 with 42% of genetic variance being independent of the hippocampus; fissure (63%); fimbria (72%); hippocampus-amygdala transition area (41%); parasubiculum (62%). In terms of genetic influence, whole hippocampal volume is a good proxy for the largest hippocampal subfields, but a poor substitute for the smaller subfields. Additive genetic sources accounted for 49%-77% of total variance for each of the subfields in the combined sample multivariate analysis. In addition, the multivariate analyses were sufficiently powered to identify common environmental influences (replicated in QTIM and HCP for the molecular layer and CA4/dentate gyrus, and accounting for 7%-16% of total variance for 8 of 10 subfields in the combined sample). This provides the clearest indication yet from a twin study that factors such as home environment may influence hippocampal volumes (albeit, with caveats).
Collapse
|
23
|
Le Grand Q, Satizabal CL, Sargurupremraj M, Mishra A, Soumaré A, Laurent A, Crivello F, Tsuchida A, Shin J, Macalli M, Singh B, Beiser AS, DeCarli C, Fletcher E, Paus T, Lathrop M, Adams HHH, Bis JC, Seshadri S, Tzourio C, Mazoyer B, Debette S. Genomic Studies Across the Lifespan Point to Early Mechanisms Determining Subcortical Volumes. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:616-628. [PMID: 34700051 PMCID: PMC9395126 DOI: 10.1016/j.bpsc.2021.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/28/2021] [Accepted: 10/14/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Subcortical brain structures play a key role in pathological processes of age-related neurodegenerative disorders. Mounting evidence also suggests that early-life factors may have an impact on the development of common late-life neurological diseases, including genetic factors that can influence both brain maturation and neurodegeneration. METHODS Using large population-based brain imaging datasets across the lifespan (N ≤ 40,628), we aimed to 1) estimate the heritability of subcortical volumes in young (18-35 years), middle (35-65 years), and older (65+ years) age, and their genetic correlation across age groups; 2) identify whether genetic loci associated with subcortical volumes in older persons also show associations in early adulthood, and explore underlying genes using transcriptome-wide association studies; and 3) explore their association with neurological phenotypes. RESULTS Heritability of subcortical volumes consistently decreased with increasing age. Genetic risk scores for smaller caudate nucleus, putamen, and hippocampus volume in older adults were associated with smaller volumes in young adults. Individually, 10 loci associated with subcortical volumes in older adults also showed associations in young adults. Within these loci, transcriptome-wide association studies showed that expression of several genes in brain tissues (especially MYLK2 and TUFM) was associated with subcortical volumes in both age groups. One risk variant for smaller caudate nucleus volume (TUFM locus) was associated with lower cognitive performance. Genetically predicted Alzheimer's disease was associated with smaller subcortical volumes in middle and older age. CONCLUSIONS Our findings provide novel insights into the genetic determinants of subcortical volumes across the lifespan. More studies are needed to decipher the underlying biology and clinical impact.
Collapse
Affiliation(s)
- Quentin Le Grand
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, Texas; Department of Population Health Sciences, UT Health San Antonio, San Antonio, Texas; Framingham Heart Study, Framingham, Massachusetts; Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Muralidharan Sargurupremraj
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, Texas
| | - Aniket Mishra
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France
| | - Aicha Soumaré
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France
| | - Alexandre Laurent
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France
| | - Fabrice Crivello
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France
| | - Ami Tsuchida
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France
| | - Jean Shin
- Department of Physiology, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Department of Nutritional Sciences, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Mélissa Macalli
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France
| | - Baljeet Singh
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California Davis, Davis, California
| | - Alexa S Beiser
- Framingham Heart Study, Framingham, Massachusetts; Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Charles DeCarli
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California Davis, Davis, California
| | - Evan Fletcher
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California Davis, Davis, California
| | - Tomas Paus
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Psychology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, Centre Hospitalier Universitaire Sainte-Justine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Mark Lathrop
- McGill Genome Center, McGill University, Montreal, Quebec, Canada
| | - Hieab H H Adams
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, Texas; Department of Population Health Sciences, UT Health San Antonio, San Antonio, Texas; Framingham Heart Study, Framingham, Massachusetts; Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Christophe Tzourio
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France; Bordeaux University Hospital, Department of Medical Informatics, Bordeaux, France
| | - Bernard Mazoyer
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; Bordeaux University Hospital, Department of Neuroradiology, Bordeaux, France
| | - Stéphanie Debette
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France; Bordeaux University Hospital, Department of Neurology, Institute of Neurodegenerative Diseases, Bordeaux, France.
| |
Collapse
|
24
|
OUP accepted manuscript. Brain 2022; 145:3214-3224. [DOI: 10.1093/brain/awac105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/11/2022] [Accepted: 03/04/2022] [Indexed: 11/15/2022] Open
|
25
|
Turner JA, Calhoun VD, Thompson PM, Jahanshad N, Ching CRK, Thomopoulos SI, Verner E, Strauss GP, Ahmed AO, Turner MD, Basodi S, Ford JM, Mathalon DH, Preda A, Belger A, Mueller BA, Lim KO, van Erp TGM. ENIGMA + COINSTAC: Improving Findability, Accessibility, Interoperability, and Re-usability. Neuroinformatics 2022; 20:261-275. [PMID: 34846691 PMCID: PMC9149142 DOI: 10.1007/s12021-021-09559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2021] [Indexed: 01/07/2023]
Abstract
The FAIR principles, as applied to clinical and neuroimaging data, reflect the goal of making research products Findable, Accessible, Interoperable, and Reusable. The use of the Collaborative Informatics and Neuroimaging Suite Toolkit for Anonymized Computation (COINSTAC) platform in the Enhancing Neuroimaging Genetics through Meta-Analysis (ENIGMA) consortium combines the technological approach of decentralized analyses with the sociological approach of sharing data. In addition, ENIGMA + COINSTAC provides a platform to facilitate the use of machine-actionable data objects. We first present how ENIGMA and COINSTAC support the FAIR principles, and then showcase their integration with a decentralized meta-analysis of sex differences in negative symptom severity in schizophrenia, and finally present ongoing activities and plans to advance FAIR principles in ENIGMA + COINSTAC. ENIGMA and COINSTAC currently represent efforts toward improved Access, Interoperability, and Reusability. We highlight additional improvements needed in these areas, as well as future connections to other resources for expanded Findability.
Collapse
Affiliation(s)
- Jessica A Turner
- Psychology Department, Georgia State University, Atlanta, GA, USA.
| | - Vince D Calhoun
- Psychology Department, Georgia State University, Atlanta, GA, USA
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, 30303, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Christopher R K Ching
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Eric Verner
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, 30303, USA
| | - Gregory P Strauss
- Departments of Psychology and Neuroscience, University of Georgia, Athens, GA, USA
| | - Anthony O Ahmed
- Weill Cornell Medicine, Department of Psychiatry, White Plains, NY, 10605, USA
| | - Matthew D Turner
- Psychology Department, Georgia State University, Atlanta, GA, USA
| | - Sunitha Basodi
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, 30303, USA
| | - Judith M Ford
- Veterans Affairs San Francisco Healthcare System, San Francisco, CA, 94121, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, 94121, USA
| | - Daniel H Mathalon
- Veterans Affairs San Francisco Healthcare System, San Francisco, CA, 94121, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, 94121, USA
| | - Adrian Preda
- Department of Psychiatry and Human Behavior, University of California Irvine, University of California Irvine Medical Center, 101 The City Drive S, Orange, CA, 92868, USA
| | - Aysenil Belger
- Department of Psychiatry and Frank Porter Graham Child Development Institute, University of North Carolina at Chapel Hill, 105 Smith Level Road, Chapel Hill, NC, 27599-8180, USA
| | - Bryon A Mueller
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, 55414, USA
| | - Kelvin O Lim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, 55414, USA
| | - Theo G M van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, 5251 California Ave, Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, 309 Qureshey Research Lab, Irvine, CA, 92697, USA
| |
Collapse
|
26
|
Wasmann JWA, Lanting CP, Huinck WJ, Mylanus EAM, van der Laak JWM, Govaerts PJ, Swanepoel DW, Moore DR, Barbour DL. Computational Audiology: New Approaches to Advance Hearing Health Care in the Digital Age. Ear Hear 2021; 42:1499-1507. [PMID: 33675587 PMCID: PMC8417156 DOI: 10.1097/aud.0000000000001041] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The global digital transformation enables computational audiology for advanced clinical applications that can reduce the global burden of hearing loss. In this article, we describe emerging hearing-related artificial intelligence applications and argue for their potential to improve access, precision, and efficiency of hearing health care services. Also, we raise awareness of risks that must be addressed to enable a safe digital transformation in audiology. We envision a future where computational audiology is implemented via interoperable systems using shared data and where health care providers adopt expanded roles within a network of distributed expertise. This effort should take place in a health care system where privacy, responsibility of each stakeholder, and patients' safety and autonomy are all guarded by design.
Collapse
Affiliation(s)
- Jan-Willem A Wasmann
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center Nijmegen, the Netherlands
| | - Cris P Lanting
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center Nijmegen, the Netherlands
| | - Wendy J Huinck
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center Nijmegen, the Netherlands
| | - Emmanuel A M Mylanus
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center Nijmegen, the Netherlands
| | - Jeroen W M van der Laak
- Department of Pathology, Radboud University Medical Center Nijmegen, the Netherlands
- Center for Medical Image Science and Visualization, Linköping University, Sweden
| | | | - De Wet Swanepoel
- Department of Speech-Language Pathology and Audiology, University of Pretoria, South Africa
| | - David R Moore
- Communication Sciences Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Otolaryngology, University of Cincinnati, Cincinnati, Ohio, USA
- Manchester Centre for Audiology and Deafness, University of Manchester, Manchester, United Kingdom
| | - Dennis L Barbour
- Department of Biomedical Engineering. Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
27
|
Abstract
Sporadic late-onset Alzheimer's disease (SLOAD) and familial early-onset Alzheimer's disease (FEOAD) associated with dominant mutations in APP, PSEN1 and PSEN2, are thought to represent a spectrum of the same disorder based on near identical behavioral and histopathological features. Hence, FEOAD transgenic mouse models have been used in past decades as a surrogate to study SLOAD pathogenic mechanisms and as the gold standard to validate drugs used in clinical trials. Unfortunately, such research has yielded little output in terms of therapeutics targeting the disease's development and progression. In this short review, we interrogate the widely accepted view of one, dimorphic disease through the prism of the Bmi1+/- mouse model and the distinct chromatin signatures observed between SLOAD and FEOAD brains.
Collapse
Affiliation(s)
| | - Ryan Hogan
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Anthony Flamier
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Gilbert Bernier
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont; Department of Neurosciences, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
28
|
Fears SC, Service SK, Kremeyer B, Araya C, Araya X, Bejarano J, Ramirez M, Castrillón G, Gomez-Franco J, Lopez MC, Montoya G, Montoya P, Aldana I, Teshiba TM, Al-Sharif NB, Jalbrzikowski M, Tishler TA, Escobar J, Ruiz-Linares A, Lopez-Jaramillo C, Macaya G, Molina J, Reus VI, Cantor RM, Sabatti C, Freimer NB, Bearden CE. Genome-wide mapping of brain phenotypes in extended pedigrees with strong genetic loading for bipolar disorder. Mol Psychiatry 2021; 26:5229-5238. [PMID: 32606377 DOI: 10.1038/s41380-020-0805-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 02/08/2023]
Abstract
Bipolar disorder is a highly heritable illness, associated with alterations of brain structure. As such, identification of genes influencing inter-individual differences in brain morphology may help elucidate the underlying pathophysiology of bipolar disorder (BP). To identify quantitative trait loci (QTL) that contribute to phenotypic variance of brain structure, structural neuroimages were acquired from family members (n = 527) of extended pedigrees heavily loaded for bipolar disorder ascertained from genetically isolated populations in Latin America. Genome-wide linkage and association analysis were conducted on the subset of heritable brain traits that showed significant evidence of association with bipolar disorder (n = 24) to map QTL influencing regional measures of brain volume and cortical thickness. Two chromosomal regions showed significant evidence of linkage; a QTL on chromosome 1p influencing corpus callosum volume and a region on chromosome 7p linked to cortical volume. Association analysis within the two QTLs identified three SNPs correlated with the brain measures.
Collapse
Affiliation(s)
- Scott C Fears
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA.
- Section of Mental Health, Greater Los Angeles Veterans Administration, Los Angeles, CA, USA.
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA.
| | - Susan K Service
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - Barbara Kremeyer
- Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
| | - Carmen Araya
- Cell and Molecular Biology Research Center, Universidad de Costa Rica, San Pedro de Montes de Oca, Costa Rica
| | - Xinia Araya
- Cell and Molecular Biology Research Center, Universidad de Costa Rica, San Pedro de Montes de Oca, Costa Rica
| | - Julio Bejarano
- Cell and Molecular Biology Research Center, Universidad de Costa Rica, San Pedro de Montes de Oca, Costa Rica
| | - Margarita Ramirez
- Cell and Molecular Biology Research Center, Universidad de Costa Rica, San Pedro de Montes de Oca, Costa Rica
| | | | | | - Maria C Lopez
- Grupo de Investigación en Psiquiatría (Research Group in Psychiatry (GIPSI)), Departamento de Psiquiatría, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Gabriel Montoya
- Grupo de Investigación en Psiquiatría (Research Group in Psychiatry (GIPSI)), Departamento de Psiquiatría, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Patricia Montoya
- Grupo de Investigación en Psiquiatría (Research Group in Psychiatry (GIPSI)), Departamento de Psiquiatría, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Ileana Aldana
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
| | - Terri M Teshiba
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
| | - Noor B Al-Sharif
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
| | - Maria Jalbrzikowski
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
| | - Todd A Tishler
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
| | - Javier Escobar
- Department of Psychiatry and Family Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Andrés Ruiz-Linares
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, China
- UMR 7268 ADES, CNRS, Aix-Marseille Université, EFS, Faculté de médecine Timone, Marseille, 13005, France
- Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, London, WC1E 6BT, UK
| | - Carlos Lopez-Jaramillo
- Grupo de Investigación en Psiquiatría (Research Group in Psychiatry (GIPSI)), Departamento de Psiquiatría, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Gabriel Macaya
- Cell and Molecular Biology Research Center, Universidad de Costa Rica, San Pedro de Montes de Oca, Costa Rica
| | - Julio Molina
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
- BioCiencias Lab, Guatemala, Guatemala
| | - Victor I Reus
- Department of Psychiatry, University of California, San Francisco, CA, USA
| | - Rita M Cantor
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Chiara Sabatti
- Department of Health Research and Policy, Stanford University, Stanford, CA, USA
| | - Nelson B Freimer
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - Carrie E Bearden
- Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| |
Collapse
|
29
|
Affiliation(s)
- Francisco Xavier Castellanos
- Department of Child and Adolescent Psychiatry, NYU Grossman School of Medicine, and Nathan Kline Institute for Psychiatric Research, New York
| |
Collapse
|
30
|
Takagi Y, Okada N, Ando S, Yahata N, Morita K, Koshiyama D, Kawakami S, Sawada K, Koike S, Endo K, Yamasaki S, Nishida A, Kasai K, Tanaka SC. Intergenerational transmission of the patterns of functional and structural brain networks. iScience 2021; 24:102708. [PMID: 34258550 PMCID: PMC8253972 DOI: 10.1016/j.isci.2021.102708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/04/2021] [Accepted: 06/08/2021] [Indexed: 01/22/2023] Open
Abstract
There is clear evidence of intergenerational transmission of life values, cognitive traits, psychiatric disorders, and even aspects of daily decision making. To investigate biological substrates of this phenomenon, the brain has received increasing attention as a measurable biomarker and potential target for intervention. However, no previous study has quantitatively and comprehensively investigated the effects of intergenerational transmission on functional and structural brain networks. Here, by employing an unusually large cohort dataset (N = 84 parent-child dyads; 45 sons, 39 daughters, 81 mothers, and 3 fathers), we show that patterns of functional and structural brain networks are preserved over a generation. We also demonstrate that several demographic factors and behavioral/physiological phenotypes have a relationship with brain similarity. Collectively, our results provide a comprehensive picture of neurobiological substrates of intergenerational transmission and demonstrate the usability of our dataset for investigating the neurobiological substrates of intergenerational transmission.
Collapse
Affiliation(s)
- Yu Takagi
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- ATR Brain Information Communication Research Laboratory Group, Kyoto, Japan
| | - Naohiro Okada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
| | - Shuntaro Ando
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Research Center for Social Science & Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Noriaki Yahata
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kentaro Morita
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Rehabilitation, The University of Tokyo Hospital, Tokyo, Japan
| | - Daisuke Koshiyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shintaro Kawakami
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kingo Sawada
- Office for Mental Health Support, Mental Health Unit, Division for Practice Research, Center for Research on Counseling and Support Services, The University of Tokyo, Tokyo, Japan
| | - Shinsuke Koike
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
- University of Tokyo Institute for Diversity and Adaptation of Human Mind (UTIDAHM), The University of Tokyo, Tokyo, Japan
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, Japan
- University of Tokyo Center for Integrative Science of Human Behavior (CiSHuB), Tokyo, Japan
| | - Kaori Endo
- Research Center for Social Science & Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Syudo Yamasaki
- Research Center for Social Science & Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Atsushi Nishida
- Research Center for Social Science & Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
- University of Tokyo Institute for Diversity and Adaptation of Human Mind (UTIDAHM), The University of Tokyo, Tokyo, Japan
- University of Tokyo Center for Integrative Science of Human Behavior (CiSHuB), Tokyo, Japan
| | - Saori C Tanaka
- ATR Brain Information Communication Research Laboratory Group, Kyoto, Japan
| |
Collapse
|
31
|
Zhao B, Shan Y, Yang Y, Yu Z, Li T, Wang X, Luo T, Zhu Z, Sullivan P, Zhao H, Li Y, Zhu H. Transcriptome-wide association analysis of brain structures yields insights into pleiotropy with complex neuropsychiatric traits. Nat Commun 2021; 12:2878. [PMID: 34001886 PMCID: PMC8128893 DOI: 10.1038/s41467-021-23130-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/16/2021] [Indexed: 02/03/2023] Open
Abstract
Structural variations of the human brain are heritable and highly polygenic traits, with hundreds of associated genes identified in recent genome-wide association studies (GWAS). Transcriptome-wide association studies (TWAS) can both prioritize these GWAS findings and also identify additional gene-trait associations. Here we perform cross-tissue TWAS analysis of 211 structural neuroimaging and discover 278 associated genes exceeding Bonferroni significance threshold of 1.04 × 10-8. The TWAS-significant genes for brain structures have been linked to a wide range of complex traits in different domains. Through TWAS gene-based polygenic risk scores (PRS) prediction, we find that TWAS PRS gains substantial power in association analysis compared to conventional variant-based GWAS PRS, and up to 6.97% of phenotypic variance (p-value = 7.56 × 10-31) can be explained in independent testing data sets. In conclusion, our study illustrates that TWAS can be a powerful supplement to traditional GWAS in imaging genetics studies for gene discovery-validation, genetic co-architecture analysis, and polygenic risk prediction.
Collapse
Affiliation(s)
- Bingxin Zhao
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yue Shan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yue Yang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhaolong Yu
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tianyou Luo
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ziliang Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Patrick Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hongyu Zhao
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
32
|
Olsen A, Babikian T, Bigler ED, Caeyenberghs K, Conde V, Dams-O'Connor K, Dobryakova E, Genova H, Grafman J, Håberg AK, Heggland I, Hellstrøm T, Hodges CB, Irimia A, Jha RM, Johnson PK, Koliatsos VE, Levin H, Li LM, Lindsey HM, Livny A, Løvstad M, Medaglia J, Menon DK, Mondello S, Monti MM, Newcombe VFJ, Petroni A, Ponsford J, Sharp D, Spitz G, Westlye LT, Thompson PM, Dennis EL, Tate DF, Wilde EA, Hillary FG. Toward a global and reproducible science for brain imaging in neurotrauma: the ENIGMA adult moderate/severe traumatic brain injury working group. Brain Imaging Behav 2021; 15:526-554. [PMID: 32797398 PMCID: PMC8032647 DOI: 10.1007/s11682-020-00313-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The global burden of mortality and morbidity caused by traumatic brain injury (TBI) is significant, and the heterogeneity of TBI patients and the relatively small sample sizes of most current neuroimaging studies is a major challenge for scientific advances and clinical translation. The ENIGMA (Enhancing NeuroImaging Genetics through Meta-Analysis) Adult moderate/severe TBI (AMS-TBI) working group aims to be a driving force for new discoveries in AMS-TBI by providing researchers world-wide with an effective framework and platform for large-scale cross-border collaboration and data sharing. Based on the principles of transparency, rigor, reproducibility and collaboration, we will facilitate the development and dissemination of multiscale and big data analysis pipelines for harmonized analyses in AMS-TBI using structural and functional neuroimaging in combination with non-imaging biomarkers, genetics, as well as clinical and behavioral measures. Ultimately, we will offer investigators an unprecedented opportunity to test important hypotheses about recovery and morbidity in AMS-TBI by taking advantage of our robust methods for large-scale neuroimaging data analysis. In this consensus statement we outline the working group's short-term, intermediate, and long-term goals.
Collapse
Affiliation(s)
- Alexander Olsen
- Department of Psychology, Norwegian University of Science and Technology, 7491, Trondheim, Norway.
- Department of Physical Medicine and Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - Talin Babikian
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA
- UCLA Steve Tisch BrainSPORT Program, Los Angeles, CA, USA
| | - Erin D Bigler
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Psychology and Neuroscience Center, Brigham Young University, Provo, UT, USA
| | - Karen Caeyenberghs
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Burwood, Australia
| | - Virginia Conde
- Department of Psychology, Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Kristen Dams-O'Connor
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ekaterina Dobryakova
- Center for Traumatic Brain Injury, Kessler Foundation, East Hanover, NJ, USA
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Helen Genova
- Center for Traumatic Brain Injury, Kessler Foundation, East Hanover, NJ, USA
| | - Jordan Grafman
- Cognitive Neuroscience Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Department of Physical Medicine & Rehabilitation, Neurology, Department of Psychiatry & Department of Psychology, Cognitive Neurology and Alzheimer's, Center, Feinberg School of Medicine, Weinberg, Chicago, IL, USA
| | - Asta K Håberg
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hopsital, Trondheim University Hospital, Trondheim, Norway
| | - Ingrid Heggland
- Section for Collections and Digital Services, NTNU University Library, Norwegian University of Science and Technology, Trondheim, Norway
| | - Torgeir Hellstrøm
- Department of Physical Medicine and Rehabilitation, Oslo University Hospital, Oslo, Norway
| | - Cooper B Hodges
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Psychology, Brigham Young University, Provo, UT, USA
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Andrei Irimia
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Ruchira M Jha
- Departments of Critical Care Medicine, Neurology, Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, Pittsburgh, PA, USA
- Clinical and Translational Science Institute, Pittsburgh, PA, USA
| | - Paula K Johnson
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT, USA
- Neuroscience Center, Brigham Young University, Provo, UT, USA
| | - Vassilis E Koliatsos
- Departments of Pathology(Neuropathology), Neurology, and Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Neuropsychiatry Program, Sheppard and Enoch Pratt Hospital, Baltimore, MD, USA
| | - Harvey Levin
- H. Ben Taub Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Lucia M Li
- C3NL, Imperial College London, London, UK
- UK DRI Centre for Health Care and Technology, Imperial College London, London, UK
| | - Hannah M Lindsey
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Psychology, Brigham Young University, Provo, UT, USA
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Abigail Livny
- Department of Diagnostic Imaging, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
- Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Marianne Løvstad
- Sunnaas Rehabilitation Hospital, Nesodden, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - John Medaglia
- Department of Psychology, Drexel University, Philadelphia, PA, USA
- Department of Neurology, Drexel University, Philadelphia, PA, USA
| | - David K Menon
- Division of Anaesthesia, University of Cambridge, Cambridge, UK
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Martin M Monti
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurosurgery, Brain Injury Research Center (BIRC), UCLA, Los Angeles, CA, USA
| | | | - Agustin Petroni
- Department of Psychology, Norwegian University of Science and Technology, 7491, Trondheim, Norway
- Department of Computer Science, Faculty of Exact & Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
- National Scientific & Technical Research Council, Institute of Research in Computer Science, Buenos Aires, Argentina
| | - Jennie Ponsford
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Australia
- Monash Epworth Rehabilitation Research Centre, Epworth Healthcare, Melbourne, Australia
| | - David Sharp
- Department of Brain Sciences, Imperial College London, London, UK
- Care Research & Technology Centre, UK Dementia Research Institute, London, UK
| | - Gershon Spitz
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Australia
| | - Lars T Westlye
- Department of Psychology, University of Oslo, Oslo, Norway
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of USC, Marina del Rey, CA, USA
- Departments of Neurology, Pediatrics, Psychiatry, Radiology, Engineering, and Ophthalmology, USC, Los Angeles, CA, USA
| | - Emily L Dennis
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT, USA
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of USC, Marina del Rey, CA, USA
| | - David F Tate
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT, USA
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Elisabeth A Wilde
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT, USA
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
- H. Ben Taub Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Frank G Hillary
- Department of Neurology, Hershey Medical Center, State College, PA, USA.
| |
Collapse
|
33
|
Farias FHG, Benitez BA, Cruchaga C. Quantitative endophenotypes as an alternative approach to understanding genetic risk in neurodegenerative diseases. Neurobiol Dis 2021; 151:105247. [PMID: 33429041 DOI: 10.1016/j.nbd.2020.105247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 01/02/2023] Open
Abstract
Endophenotypes, as measurable intermediate features of human diseases, reflect underlying molecular mechanisms. The use of quantitative endophenotypes in genetic studies has improved our understanding of pathophysiological changes associated with diseases. The main advantage of the quantitative endophenotypes approach to study human diseases over a classic case-control study design is the inferred biological context that can enable the development of effective disease-modifying treatments. Here, we summarize recent progress on biomarkers for neurodegenerative diseases, including cerebrospinal fluid and blood-based, neuroimaging, neuropathological, and clinical studies. This review focuses on how endophenotypic studies have successfully linked genetic modifiers to disease risk, disease onset, or progression rate and provided biological context to genes identified in genome-wide association studies. Finally, we review critical methodological considerations for implementing this approach and future directions.
Collapse
Affiliation(s)
- Fabiana H G Farias
- Department of Psychiatry, Washington University, St. Louis, MO 63110, United States of America; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, United States of America
| | - Bruno A Benitez
- Department of Psychiatry, Washington University, St. Louis, MO 63110, United States of America; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, United States of America
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, MO 63110, United States of America; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, United States of America; Hope Center for Neurologic Diseases, Washington University, St. Louis, MO 63110, United States of America; The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, 63110, United States of America; Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, United States of America.
| |
Collapse
|
34
|
Medland SE, Grasby KL, Jahanshad N, Painter JN, Colodro-Conde L, Bralten J, Hibar DP, Lind PA, Pizzagalli F, Thomopoulos SI, Stein JL, Franke B, Martin NG, Thompson PM. Ten years of enhancing neuro-imaging genetics through meta-analysis: An overview from the ENIGMA Genetics Working Group. Hum Brain Mapp 2020; 43:292-299. [PMID: 33300665 PMCID: PMC8675405 DOI: 10.1002/hbm.25311] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/22/2022] Open
Abstract
Here we review the motivation for creating the enhancing neuroimaging genetics through meta-analysis (ENIGMA) Consortium and the genetic analyses undertaken by the consortium so far. We discuss the methodological challenges, findings, and future directions of the genetics working group. A major goal of the working group is tackling the reproducibility crisis affecting "candidate gene" and genome-wide association analyses in neuroimaging. To address this, we developed harmonized analytic methods, and support their use in coordinated analyses across sites worldwide, which also makes it possible to understand heterogeneity in results across sites. These efforts have resulted in the identification of hundreds of common genomic loci robustly associated with brain structure. We have found both pleiotropic and specific genetic effects associated with brain structures, as well as genetic correlations with psychiatric and neurological diseases.
Collapse
Affiliation(s)
- Sarah E Medland
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,School of Psychology, University of Queensland, Brisbane, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Katrina L Grasby
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, USA
| | - Jodie N Painter
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Lucía Colodro-Conde
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,School of Psychology, University of Queensland, Brisbane, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.,Faculty of Psychology, University of Murcia, Murcia, Spain
| | - Janita Bralten
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Derrek P Hibar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, USA.,Personalized Healthcare, Genentech, Inc., South San Francisco, California, USA
| | - Penelope A Lind
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Fabrizio Pizzagalli
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, USA
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, USA
| | - Jason L Stein
- Department of Genetics & UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Barbara Franke
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, USA
| | | |
Collapse
|
35
|
Knutson KA, Deng Y, Pan W. Implicating causal brain imaging endophenotypes in Alzheimer's disease using multivariable IWAS and GWAS summary data. Neuroimage 2020; 223:117347. [PMID: 32898681 PMCID: PMC7778364 DOI: 10.1016/j.neuroimage.2020.117347] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
Recent evidence suggests the existence of many undiscovered heritable brain phenotypes involved in Alzheimer's Disease (AD) pathogenesis. This finding necessitates methods for the discovery of causal brain changes in AD that integrate Magnetic Resonance Imaging measures and genotypic data. However, existing approaches for causal inference in this setting, such as the univariate Imaging Wide Association Study (UV-IWAS), suffer from inconsistent effect estimation and inflated Type I errors in the presence of genetic pleiotropy, the phenomenon in which a variant affects multiple causal intermediate risk phenotypes. In this study, we implement a multivariate extension to the IWAS model, namely MV-IWAS, to consistently estimate and test for the causal effects of multiple brain imaging endophenotypes from the Alzheimer's Disease Neuroimaging Initiative (ADNI) in the presence of pleiotropic and possibly correlated SNPs. We further extend MV-IWAS to incorporate variant-specific direct effects on AD, analogous to the existing Egger regression Mendelian Randomization approach, which allows for testing of remaining pleiotropy after adjusting for multiple intermediate pathways. We propose a convenient approach for implementing MV-IWAS that solely relies on publicly available GWAS summary data and a reference panel. Through simulations with either individual-level or summary data, we demonstrate the well controlled Type I errors and superior power of MV-IWAS over UV-IWAS in the presence of pleiotropic SNPs. We apply the summary statistic based tests to 1578 heritable imaging derived phenotypes (IDPs) from the UK Biobank. MV-IWAS detected numerous IDPs as possible false positives by UV-IWAS while uncovering many additional causal neuroimaging phenotypes in AD which are strongly supported by the existing literature.
Collapse
Affiliation(s)
- Katherine A Knutson
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota United States
| | - Yangqing Deng
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota United States
| | - Wei Pan
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota United States.
| |
Collapse
|
36
|
Dunn AR, Hadad N, Neuner SM, Zhang JG, Philip VM, Dumitrescu L, Hohman TJ, Herskowitz JH, O’Connell KMS, Kaczorowski CC. Identifying Mechanisms of Normal Cognitive Aging Using a Novel Mouse Genetic Reference Panel. Front Cell Dev Biol 2020; 8:562662. [PMID: 33042997 PMCID: PMC7517308 DOI: 10.3389/fcell.2020.562662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
Developing strategies to maintain cognitive health is critical to quality of life during aging. The basis of healthy cognitive aging is poorly understood; thus, it is difficult to predict who will have normal cognition later in life. Individuals may have higher baseline functioning (cognitive reserve) and others may maintain or even improve with age (cognitive resilience). Understanding the mechanisms underlying cognitive reserve and resilience may hold the key to new therapeutic strategies for maintaining cognitive health. However, reserve and resilience have been inconsistently defined in human studies. Additionally, our understanding of the molecular and cellular bases of these phenomena is poor, compounded by a lack of longitudinal molecular and cognitive data that fully capture the dynamic trajectories of cognitive aging. Here, we used a genetically diverse mouse population (B6-BXDs) to characterize individual differences in cognitive abilities in adulthood and investigate evidence of cognitive reserve and/or resilience in middle-aged mice. We tested cognitive function at two ages (6 months and 14 months) using y-maze and contextual fear conditioning. We observed heritable variation in performance on these traits (h 2 RIx̄ = 0.51-0.74), suggesting moderate to strong genetic control depending on the cognitive domain. Due to the polygenetic nature of cognitive function, we did not find QTLs significantly associated with y-maze, contextual fear acquisition (CFA) or memory, or decline in cognitive function at the genome-wide level. To more precisely interrogate the molecular regulation of variation in these traits, we employed RNA-seq and identified gene networks related to transcription/translation, cellular metabolism, and neuronal function that were associated with working memory, contextual fear memory, and cognitive decline. Using this method, we nominate the Trio gene as a modulator of working memory ability. Finally, we propose a conceptual framework for identifying strains exhibiting cognitive reserve and/or resilience to assess whether these traits can be observed in middle-aged B6-BXDs. Though we found that earlier cognitive reserve evident early in life protects against cognitive impairment later in life, cognitive performance and age-related decline fell along a continuum, with no clear genotypes emerging as exemplars of exceptional reserve or resilience - leading to recommendations for future use of aging mouse populations to understand the nature of cognitive reserve and resilience.
Collapse
Affiliation(s)
- Amy R. Dunn
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Niran Hadad
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Sarah M. Neuner
- The Jackson Laboratory, Bar Harbor, ME, United States
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ji-Gang Zhang
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer’s Center and Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Center and Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeremy H. Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics and Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | | | | |
Collapse
|
37
|
Jian X, Sofer T, Tarraf W, Bressler J, Faul JD, Zhao W, Ratliff SM, Lamar M, Launer LJ, Laurie CC, Schneiderman N, Weir DR, Wright CB, Yaffe K, Zeng D, DeCarli C, Mosley TH, Smith JA, González HM, Fornage M. Genome-wide association study of cognitive function in diverse Hispanics/Latinos: results from the Hispanic Community Health Study/Study of Latinos. Transl Psychiatry 2020; 10:245. [PMID: 32699239 PMCID: PMC7376098 DOI: 10.1038/s41398-020-00930-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/19/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022] Open
Abstract
Cognitive function such as reasoning, attention, memory, and language is strongly correlated with brain aging. Compared to non-Hispanic whites, Hispanics/Latinos have a higher risk of cognitive impairment and dementia. The genetic determinants of cognitive function have not been widely explored in this diverse and admixed population. We conducted a genome-wide association analysis of cognitive function in up to 7600 middle aged and older Hispanics/Latinos (mean = 55 years) from the Hispanic Community Health Study / Study of Latinos (HCHS/SOL). Four cognitive measures were examined: the Brief Spanish English Verbal Learning Test (B-SEVLT), the Word Fluency Test (WFT), the Digit Symbol Substitution Test (DSST), the Six-Item Screener (SIS). Four novel loci were identified: one for B-SEVLT at 4p14, two for WFT at 3p14.1 and 6p21.32, and one for DSST at 10p13. These loci implicate genes highly expressed in brain and previously connected to neurological diseases (UBE2K, FRMD4B, the HLA gene complex). By applying tissue-specific gene expression prediction models to our genotype data, additional genes highly expressed in brain showed suggestive associations with cognitive measures possibly indicating novel biological mechanisms, including IFT122 in the hippocampus for SIS, SNX31 in the basal ganglia for B-SEVLT, RPS6KB2 in the frontal cortex for WFT, and CSPG5 in the hypothalamus for DSST. These findings provide new information about the genetic determinants of cognitive function in this unique population. In addition, we derived a measure of general cognitive function based on these cognitive tests and generated genome-wide association summary results, providing a resource to the research community for comparison, replication, and meta-analysis in future genetic studies in Hispanics/Latinos.
Collapse
Affiliation(s)
- Xueqiu Jian
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Tamar Sofer
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wassim Tarraf
- Institute of Gerontology and Department of Health Care Sciences, Wayne State University, Detroit, MI, USA
| | - Jan Bressler
- Department of Epidemiology, Human Genetics and Environmental Sciences and Human Genetics Center, The University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhao
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Scott M Ratliff
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Melissa Lamar
- Department of Behavioral Sciences, Rush Medical College, Chicago, IL, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Bethesda, MD, USA
| | - Cathy C Laurie
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Neil Schneiderman
- Department of Psychology, University of Miami, Coral Gables, FL, USA
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Clinton B Wright
- Division of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Kristine Yaffe
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Donglin Zeng
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Charles DeCarli
- Department of Neurology, School of Medicine and Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California, Davis, Sacramento, CA, USA
| | - Thomas H Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center and Department of Medicine, The University of Mississippi Medical Center, Jackson, MS, USA
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Hector M González
- Department of Neurosciences and Shiley-Marcos Alzheimer's Disease Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Department of Epidemiology, Human Genetics and Environmental Sciences and Human Genetics Center, The University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA.
| |
Collapse
|
38
|
Long L, Galovic M, Chen Y, Postma T, Vos SB, Xiao F, Wu W, Song Y, Huang S, Koepp M, Xiao B. Shared hippocampal abnormalities in sporadic temporal lobe epilepsy patients and their siblings. Epilepsia 2020; 61:735-746. [PMID: 32196657 DOI: 10.1111/epi.16477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To examine the shared familial contribution to hippocampal and extrahippocampal morphological abnormalities in patients with sporadic temporal lobe epilepsy (TLE) and their unaffected siblings. METHODS We collected clinical, electrophysiological, and T1-weighted magnetic resonance imaging (MRI) data of 18 sporadic patients with TLE without lesions other than hippocampal sclerosis (12 right, 6 left), their 18 unaffected full siblings, and 18 matched healthy volunteers. We compared between-group differences in cortical thickness and volumes of five subcortical areas (hippocampus, amygdala, thalamus, putamen, and pallidum). We determined the subregional extent of hippocampal abnormalities using surface shape analysis. All our imaging results were corrected for multiple comparisons using random field theory. RESULTS We detected smaller hippocampal volumes in patients (right TLE: median right hippocampus 1.92 mL, interquartile range [IQR] 1.39-2.62, P < .001; left TLE: left hippocampus 2.05 mL, IQR 1.99-2.33, P = .01) and their unaffected siblings (right hippocampus 2.65 mL, IQR 2.32-2.80, P < .001; left hippocampus 2.39 mL, IQR 2.18-2.53, P < .001) compared to healthy controls (right hippocampus 2.94 mL, IQR 2.77-3.24; left hippocampus 2.71 mL, IQR 2.37-2.89). Surface shape analysis showed that patients with TLE had bilateral subregional atrophy in both hippocampi (right > left). Similar but less-pronounced subregional atrophy was detected in the right hippocampus of unaffected siblings. Patients with TLE had reduced cortical thickness in bilateral premotor/prefrontal cortices and the right precentral gyrus. Siblings did not show abnormalities in cortical or subcortical areas other than the hippocampus. SIGNIFICANCE Our results demonstrate a shared vulnerability of the hippocampus in both patients with TLE and their unaffected siblings, pointing to a contribution of familial factors to hippocampal atrophy. This neuroimaging trait could represent an endophenotype of TLE, which might precede the onset of epilepsy in some individuals.
Collapse
Affiliation(s)
- Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Marian Galovic
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK.,MRI Unit, Chalfont Centre for Epilepsy, Chalfont St Peter, UK.,Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Yayu Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Tjardo Postma
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Sjoerd B Vos
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK.,MRI Unit, Chalfont Centre for Epilepsy, Chalfont St Peter, UK.,Centre for Medical Image Computing, University College London, London, UK
| | - Fenglai Xiao
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK.,MRI Unit, Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Wenyue Wu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanmin Song
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, China
| | - Sha Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Matthias Koepp
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK.,MRI Unit, Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Campos AI, García-Marín LM, Byrne EM, Martin NG, Cuéllar-Partida G, Rentería ME. Insights into the aetiology of snoring from observational and genetic investigations in the UK Biobank. Nat Commun 2020; 11:817. [PMID: 32060260 PMCID: PMC7021827 DOI: 10.1038/s41467-020-14625-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/22/2020] [Indexed: 12/15/2022] Open
Abstract
Although snoring is common in the general population, its aetiology has been largely understudied. Here we report a genetic study on snoring (n ~ 408,000; snorers ~ 152,000) using data from the UK Biobank. We identify 42 genome-wide significant loci, with an SNP-based heritability estimate of ~10% on the liability scale. Genetic correlations with body mass index, alcohol intake, smoking, schizophrenia, anorexia nervosa and neuroticism are observed. Gene-based associations identify 173 genes, including DLEU7, MSRB3 and POC5, highlighting genes expressed in the brain, cerebellum, lungs, blood and oesophagus. We use polygenic scores (PGS) to predict recent snoring and probable obstructive sleep apnoea (OSA) in an independent Australian sample (n ~ 8000). Mendelian randomization analyses suggest a potential causal relationship between high BMI and snoring. Altogether, our results uncover insights into the aetiology of snoring as a complex sleep-related trait and its role in health and disease beyond it being a cardinal symptom of OSA. Snoring is common in the population and tends to be more prevalent in older and/or male individuals. Here, the authors perform GWAS for habitual snoring, identify 41 genomic loci and explore potential causal relationships with anthropometric and cardiometabolic disease traits.
Collapse
Affiliation(s)
- Adrián I Campos
- Genetic Epidemiology Lab, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Luis M García-Marín
- Genetic Epidemiology Lab, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Zapopan, Jalisco, México
| | - Enda M Byrne
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Nicholas G Martin
- Genetic Epidemiology Lab, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Gabriel Cuéllar-Partida
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia. .,University of Queensland Diamantina Institute, Brisbane, QLD, Australia.
| | - Miguel E Rentería
- Genetic Epidemiology Lab, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia. .,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
40
|
Adhikari BM, Jahanshad N, Shukla D, Turner J, Grotegerd D, Dannlowski U, Kugel H, Engelen J, Dietsche B, Krug A, Kircher T, Fieremans E, Veraart J, Novikov DS, Boedhoe PSW, van der Werf YD, van den Heuvel OA, Ipser J, Uhlmann A, Stein DJ, Dickie E, Voineskos AN, Malhotra AK, Pizzagalli F, Calhoun VD, Waller L, Veer IM, Walter H, Buchanan RW, Glahn DC, Hong LE, Thompson PM, Kochunov P. A resting state fMRI analysis pipeline for pooling inference across diverse cohorts: an ENIGMA rs-fMRI protocol. Brain Imaging Behav 2020; 13:1453-1467. [PMID: 30191514 DOI: 10.1007/s11682-018-9941-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Large-scale consortium efforts such as Enhancing NeuroImaging Genetics through Meta-Analysis (ENIGMA) and other collaborative efforts show that combining statistical data from multiple independent studies can boost statistical power and achieve more accurate estimates of effect sizes, contributing to more reliable and reproducible research. A meta- analysis would pool effects from studies conducted in a similar manner, yet to date, no such harmonized protocol exists for resting state fMRI (rsfMRI) data. Here, we propose an initial pipeline for multi-site rsfMRI analysis to allow research groups around the world to analyze scans in a harmonized way, and to perform coordinated statistical tests. The challenge lies in the fact that resting state fMRI measurements collected by researchers over the last decade vary widely, with variable quality and differing spatial or temporal signal-to-noise ratio (tSNR). An effective harmonization must provide optimal measures for all quality data. Here we used rsfMRI data from twenty-two independent studies with approximately fifty corresponding T1-weighted and rsfMRI datasets each, to (A) review and aggregate the state of existing rsfMRI data, (B) demonstrate utility of principal component analysis (PCA)-based denoising and (C) develop a deformable ENIGMA EPI template based on the representative anatomy that incorporates spatial distortion patterns from various protocols and populations.
Collapse
Affiliation(s)
- Bhim M Adhikari
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Neda Jahanshad
- Imaging Genetics Center, Keck School of Medicine of USC, Marina del Rey, Los Angeles, CA, USA
| | - Dinesh Shukla
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jessica Turner
- Department of Psychology, Georgia State University, Atlanta, GA, USA
| | | | - Udo Dannlowski
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Harald Kugel
- Department of Clinical Radiology, University of Münster, Münster, Germany
| | - Jennifer Engelen
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Bruno Dietsche
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Els Fieremans
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Jelle Veraart
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Dmitry S Novikov
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Premika S W Boedhoe
- Department of Psychiatry, Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, Netherlands
| | - Ysbrand D van der Werf
- Department of Psychiatry, Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, Netherlands
| | - Odile A van den Heuvel
- Department of Psychiatry, Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, Netherlands
| | - Jonathan Ipser
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Anne Uhlmann
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Dan J Stein
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Erin Dickie
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Aristotle N Voineskos
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Anil K Malhotra
- Department of Psychiatry, The Zucker Hillside Hospital, Glen Oaks, New York, NY, USA
| | - Fabrizio Pizzagalli
- Imaging Genetics Center, Keck School of Medicine of USC, Marina del Rey, Los Angeles, CA, USA
| | - Vince D Calhoun
- The Mind Research Network & The University of New Mexico, Albuquerque, NM, USA
| | - Lea Waller
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Campus Matte, Berlin, Germany
| | - Ilja M Veer
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Campus Matte, Berlin, Germany
| | - Hernik Walter
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Campus Matte, Berlin, Germany
| | - Robert W Buchanan
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David C Glahn
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, USA
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Paul M Thompson
- Imaging Genetics Center, Keck School of Medicine of USC, Marina del Rey, Los Angeles, CA, USA
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
van der Meer D, Rokicki J, Kaufmann T, Córdova-Palomera A, Moberget T, Alnæs D, Bettella F, Frei O, Doan NT, Sønderby IE, Smeland OB, Agartz I, Bertolino A, Bralten J, Brandt CL, Buitelaar JK, Djurovic S, van Donkelaar M, Dørum ES, Espeseth T, Faraone SV, Fernández G, Fisher SE, Franke B, Haatveit B, Hartman CA, Hoekstra PJ, Håberg AK, Jönsson EG, Kolskår KK, Le Hellard S, Lund MJ, Lundervold AJ, Lundervold A, Melle I, Monereo Sánchez J, Norbom LC, Nordvik JE, Nyberg L, Oosterlaan J, Papalino M, Papassotiropoulos A, Pergola G, de Quervain DJF, Richard G, Sanders AM, Selvaggi P, Shumskaya E, Steen VM, Tønnesen S, Ulrichsen KM, Zwiers MP, Andreassen OA, Westlye LT, for the Alzheimer’s Disease Neuroimaging Initiative, for the Pediatric Imaging, Neurocognition and Genetics Study. Brain scans from 21,297 individuals reveal the genetic architecture of hippocampal subfield volumes. Mol Psychiatry 2020; 25:3053-3065. [PMID: 30279459 PMCID: PMC6445783 DOI: 10.1038/s41380-018-0262-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/09/2018] [Accepted: 09/06/2018] [Indexed: 11/09/2022]
Abstract
The hippocampus is a heterogeneous structure, comprising histologically distinguishable subfields. These subfields are differentially involved in memory consolidation, spatial navigation and pattern separation, complex functions often impaired in individuals with brain disorders characterized by reduced hippocampal volume, including Alzheimer's disease (AD) and schizophrenia. Given the structural and functional heterogeneity of the hippocampal formation, we sought to characterize the subfields' genetic architecture. T1-weighted brain scans (n = 21,297, 16 cohorts) were processed with the hippocampal subfields algorithm in FreeSurfer v6.0. We ran a genome-wide association analysis on each subfield, co-varying for whole hippocampal volume. We further calculated the single-nucleotide polymorphism (SNP)-based heritability of 12 subfields, as well as their genetic correlation with each other, with other structural brain features and with AD and schizophrenia. All outcome measures were corrected for age, sex and intracranial volume. We found 15 unique genome-wide significant loci across six subfields, of which eight had not been previously linked to the hippocampus. Top SNPs were mapped to genes associated with neuronal differentiation, locomotor behaviour, schizophrenia and AD. The volumes of all the subfields were estimated to be heritable (h2 from 0.14 to 0.27, all p < 1 × 10-16) and clustered together based on their genetic correlations compared with other structural brain features. There was also evidence of genetic overlap of subicular subfield volumes with schizophrenia. We conclude that hippocampal subfields have partly distinct genetic determinants associated with specific biological processes and traits. Taking into account this specificity may increase our understanding of hippocampal neurobiology and associated pathologies.
Collapse
Affiliation(s)
- Dennis van der Meer
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Jaroslav Rokicki
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Aldo Córdova-Palomera
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.168010.e0000000419368956Department of Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, USA
| | - Torgeir Moberget
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dag Alnæs
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Francesco Bettella
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Oleksandr Frei
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nhat Trung Doan
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ida E. Sønderby
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav B. Smeland
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid Agartz
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alessandro Bertolino
- grid.7644.10000 0001 0120 3326Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy ,Azienda Ospedaliero-Universitaria Consorziale Policlinico, Bari, Italy
| | - Janita Bralten
- grid.10417.330000 0004 0444 9382Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands ,grid.5590.90000000122931605Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Christine L. Brandt
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jan K. Buitelaar
- grid.5590.90000000122931605Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Srdjan Djurovic
- grid.55325.340000 0004 0389 8485Department of Medical Genetics, Oslo University Hospital, Oslo, Norway ,grid.7914.b0000 0004 1936 7443NORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marjolein van Donkelaar
- grid.10417.330000 0004 0444 9382Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands ,grid.5590.90000000122931605Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Erlend S. Dørum
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway ,grid.416731.60000 0004 0612 1014Sunnaas Rehabilitation Hospital HT, Nesodden, Norway
| | - Thomas Espeseth
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway
| | - Stephen V. Faraone
- grid.411023.50000 0000 9159 4457Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY USA
| | - Guillén Fernández
- grid.5590.90000000122931605Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Simon E. Fisher
- grid.5590.90000000122931605Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands ,grid.419550.c0000 0004 0501 3839Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, Netherlands
| | - Barbara Franke
- grid.10417.330000 0004 0444 9382Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands ,grid.5590.90000000122931605Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Beathe Haatveit
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway
| | - Catharina A. Hartman
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, Interdisciplinary Center Psychopathology and Emotion Regulation, Groningen, The Netherlands
| | - Pieter J. Hoekstra
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, Department of Child and Adolescent Psychiatry, Groningen, Netherlands
| | - Asta K. Håberg
- grid.5947.f0000 0001 1516 2393Department of Neuromedicine and Movement Science, NTNU – Norwegian University of Science and Technology, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim, Norway
| | - Erik G. Jönsson
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.4714.60000 0004 1937 0626Centre for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Knut K. Kolskår
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway ,grid.416731.60000 0004 0612 1014Sunnaas Rehabilitation Hospital HT, Nesodden, Norway
| | - Stephanie Le Hellard
- grid.7914.b0000 0004 1936 7443NORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway ,grid.412008.f0000 0000 9753 1393Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Martina J. Lund
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Astri J. Lundervold
- grid.7914.b0000 0004 1936 7443Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Arvid Lundervold
- grid.7914.b0000 0004 1936 7443Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ingrid Melle
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jennifer Monereo Sánchez
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Linn C. Norbom
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway
| | - Jan E. Nordvik
- grid.416731.60000 0004 0612 1014Sunnaas Rehabilitation Hospital HT, Nesodden, Norway
| | - Lars Nyberg
- grid.12650.300000 0001 1034 3451Departments of Radiation Sciences and Integrative Medical Biology, Umeå Center for Functional Brain Imaging (UFB), Umeå University, Umeå, Sweden
| | - Jaap Oosterlaan
- Amsterdam UMC, University of Amsterdam & Vrije Universiteit Amsterdam, Emma Neuroscience Group at Emma Children’s Hospital, department of Pediatrics, Amsterdam Reproduction & Development, Amsterdam, The Netherlands
| | - Marco Papalino
- grid.7644.10000 0001 0120 3326Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Andreas Papassotiropoulos
- grid.6612.30000 0004 1937 0642Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland ,grid.6612.30000 0004 1937 0642Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland ,grid.6612.30000 0004 1937 0642Life Sciences Training Facility, Department Biozentrum, University of Basel, Basel, Switzerland
| | - Giulio Pergola
- grid.7644.10000 0001 0120 3326Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Dominique J. F. de Quervain
- grid.6612.30000 0004 1937 0642Division of Cognitive Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
| | - Geneviève Richard
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway ,grid.416731.60000 0004 0612 1014Sunnaas Rehabilitation Hospital HT, Nesodden, Norway
| | - Anne-Marthe Sanders
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway ,grid.416731.60000 0004 0612 1014Sunnaas Rehabilitation Hospital HT, Nesodden, Norway
| | - Pierluigi Selvaggi
- grid.7644.10000 0001 0120 3326Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy ,grid.13097.3c0000 0001 2322 6764Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Elena Shumskaya
- grid.10417.330000 0004 0444 9382Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands ,grid.5590.90000000122931605Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Vidar M. Steen
- grid.7914.b0000 0004 1936 7443NORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway ,grid.412008.f0000 0000 9753 1393Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Siren Tønnesen
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kristine M. Ulrichsen
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway ,grid.416731.60000 0004 0612 1014Sunnaas Rehabilitation Hospital HT, Nesodden, Norway
| | - Marcel P. Zwiers
- grid.5590.90000000122931605Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Ole A. Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars T. Westlye
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Psychology, University of Oslo, Oslo, Norway
| | | | | |
Collapse
|
42
|
Abstract
Radiogenomics, defined as the integrated analysis of radiologic imaging and genetic data, is a well-established tool shown to augment neuroimaging in the clinical diagnosis, prognostication, and scientific study of late-onset Alzheimer disease (LOAD). Early work using candidate single nucleotide polymorphisms (SNPs) identified genetic variation in APOE, BIN1, CLU, and CR1 as key modifiers of brain structure and function using magnetic resonance imaging (MRI). More recently, polygenic risk scores used in conjunction with MRI and positron emission tomography have shown great promise as a risk-stratification tool for clinical trials and care-management decisions. In addition, recent work using multimodal MRI and positron emission tomography as proxies of LOAD progression has identified novel risk variants that are enhancing our understanding of LOAD pathophysiology and progression. Herein, we highlight key studies and trends in the radiogenomics of LOAD over the past two decades and their implications for clinical practice and scientific research.
Collapse
|
43
|
Reiterer M, Schmidt-Kastner R, Milton SL. Methionine sulfoxide reductase (Msr) dysfunction in human brain disease. Free Radic Res 2019; 53:1144-1154. [PMID: 31775527 DOI: 10.1080/10715762.2019.1662899] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Extensive research has shown that oxidative stress is strongly associated with aging, senescence and several diseases, including neurodegenerative and psychiatric disorders. Oxidative stress is caused by the overproduction of reactive oxygen species (ROS) that can be counteracted by both enzymatic and nonenzymatic antioxidants. One of these antioxidant mechanisms is the widely studied methionine sulfoxide reductase system (Msr). Methionine is one of the most easily oxidized amino acids and Msr can reverse this oxidation and restore protein function, with MsrA and MsrB reducing different stereoisomers. This article focuses on experimental and genetic research performed on Msr and its link to brain diseases. Studies on several model systems as well as genome-wide association studies are compiled to highlight the role of MSRA in schizophrenia, Alzheimer's disease, and Parkinson's disease. Genetic variation of MSRA may also contribute to the risk of psychosis, personality traits, and metabolic factors.
Collapse
Affiliation(s)
- Melissa Reiterer
- Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, USA
| | | | - Sarah L Milton
- Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
44
|
Zhao B, Luo T, Li T, Li Y, Zhang J, Shan Y, Wang X, Yang L, Zhou F, Zhu Z, Zhu H. Genome-wide association analysis of 19,629 individuals identifies variants influencing regional brain volumes and refines their genetic co-architecture with cognitive and mental health traits. Nat Genet 2019; 51:1637-1644. [PMID: 31676860 PMCID: PMC6858580 DOI: 10.1038/s41588-019-0516-6] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 09/23/2019] [Indexed: 12/19/2022]
Abstract
Volumetric variations of the human brain are heritable and are associated with many brain-related complex traits. Here we performed genome-wide association studies (GWAS) of 101 brain volumetric phenotypes using the UK Biobank sample including 19,629 participants. GWAS identified 365 independent genetic variants exceeding a significance threshold of 4.9 × 10-10, adjusted for testing multiple phenotypes. A gene-based association study found 157 associated genes (124 new), and functional gene mapping analysis linked 146 additional genes. Many of the discovered genetic variants and genes have previously been implicated in cognitive and mental health traits. Through genome-wide polygenic-risk-score prediction, more than 6% of the phenotypic variance (P = 3.13 × 10-24) in four other independent studies could be explained by the UK Biobank GWAS results. In conclusion, our study identifies many new genetic associations at the variant, locus and gene levels and advances our understanding of the pleiotropy and genetic co-architecture between brain volumes and other traits.
Collapse
Affiliation(s)
- Bingxin Zhao
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tianyou Luo
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jingwen Zhang
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Yue Shan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Liuqing Yang
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Fan Zhou
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ziliang Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
45
|
Ho G, Takamatsu Y, Waragai M, Wada R, Sugama S, Takenouchi T, Fujita M, Ali A, Hsieh MHI, Hashimoto M. Current and future clinical utilities of Parkinson's disease and dementia biomarkers: can they help us conquer the disease? Expert Rev Neurother 2019; 19:1149-1161. [PMID: 31359797 DOI: 10.1080/14737175.2019.1649141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: Biomarkers for Parkinson's disease and Alzheimer's disease are essential, not only for disease detection, but also provide insight into potential disease relationships leading to better detection and therapy. As metabolic disease is known to increase neurodegeneration risk, such mechanisms may reveal such novel targets for PD and AD. Moreover, metabolic disease, including insulin resistance, offer novel biomarker and therapeutic targets for neurodegeneration, including glucagon-like-peptide-1, dipeptidyl peptidase-4 and adiponectin. Areas covered: The authors reviewed PubMed-listed research articles, including ours, on a number of putative PD, AD and neurodegenerative disease targets of interest, focusing on the relevance of metabolic syndrome and insulin resistance mechanisms, especially type II diabetes, to PD and AD. We highlighted various issues surrounding the current state of knowledge and propose avenues for future development. Expert opinion: Biomarkers for PD and AD are indispensable for disease diagnosis, prognostication and tracking disease severity, especially for clinical therapy trials. Although no validated PD biomarkers exist, their potential utility has generated tremendous interest. Combining insulin-resistance biomarkers with other core biomarkers or using them to predict non-motor symptoms of PD may be clinically useful. Collectively, although still unclear, potential biomarkers and therapies can aid in shedding new light on novel aspects of both PD and AD.
Collapse
Affiliation(s)
- Gilbert Ho
- PCND Neuroscience Research Institute , Poway , CA , USA
| | | | - Masaaki Waragai
- Tokyo Metropolitan Institute of Medical Science , Tokyo , Japan
| | - Ryoko Wada
- Tokyo Metropolitan Institute of Medical Science , Tokyo , Japan
| | - Shuei Sugama
- Department of Physiology, Nippon Medical School , Tokyo , Japan
| | - Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization , Tsukuba , Japan
| | - Masayo Fujita
- Tokyo Metropolitan Institute of Medical Science , Tokyo , Japan
| | - Alysha Ali
- PCND Neuroscience Research Institute , Poway , CA , USA
| | | | | |
Collapse
|
46
|
Le BD, Stein JL. Mapping causal pathways from genetics to neuropsychiatric disorders using genome-wide imaging genetics: Current status and future directions. Psychiatry Clin Neurosci 2019; 73:357-369. [PMID: 30864184 PMCID: PMC6625892 DOI: 10.1111/pcn.12839] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/17/2022]
Abstract
Imaging genetics aims to identify genetic variants associated with the structure and function of the human brain. Recently, collaborative consortia have been successful in this goal, identifying and replicating common genetic variants influencing gross human brain structure as measured through magnetic resonance imaging. In this review, we contextualize imaging genetic associations as one important link in understanding the causal chain from genetic variant to increased risk for neuropsychiatric disorders. We provide examples in other fields of how identifying genetic variant associations to disease and multiple phenotypes along the causal chain has revealed a mechanistic understanding of disease risk, with implications for how imaging genetics can be similarly applied. We discuss current findings in the imaging genetics research domain, including that common genetic variants can have a slightly larger effect on brain structure than on risk for disorders like schizophrenia, indicating a somewhat simpler genetic architecture. Also, gross brain structure measurements share a genetic basis with some, but not all, neuropsychiatric disorders, invalidating the previously held belief that they are broad endophenotypes, yet pinpointing brain regions likely involved in the pathology of specific disorders. Finally, we suggest that in order to build a more detailed mechanistic understanding of the effects of genetic variants on the brain, future directions in imaging genetics research will require observations of cellular and synaptic structure in specific brain regions beyond the resolution of magnetic resonance imaging. We expect that integrating genetic associations at biological levels from synapse to sulcus will reveal specific causal pathways impacting risk for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Brandon D. Le
- Department of Genetics & UNC Neuroscience Center, University of North Carolina at Chapel Hill, USA
| | - Jason L. Stein
- Department of Genetics & UNC Neuroscience Center, University of North Carolina at Chapel Hill, USA
| |
Collapse
|
47
|
van Erp TGM, Walton E, Hibar DP, Schmaal L, Jiang W, Glahn DC, Pearlson GD, Yao N, Fukunaga M, Hashimoto R, Okada N, Yamamori H, Clark VP, Mueller BA, de Zwarte SMC, Ophoff RA, van Haren NEM, Andreassen OA, Gurholt TP, Gruber O, Kraemer B, Richter A, Calhoun VD, Crespo-Facorro B, Roiz-Santiañez R, Tordesillas-Gutiérrez D, Loughland C, Catts S, Fullerton JM, Green MJ, Henskens F, Jablensky A, Mowry BJ, Pantelis C, Quidé Y, Schall U, Scott RJ, Cairns MJ, Seal M, Tooney PA, Rasser PE, Cooper G, Weickert CS, Weickert TW, Hong E, Kochunov P, Gur RE, Gur RC, Ford JM, Macciardi F, Mathalon DH, Potkin SG, Preda A, Fan F, Ehrlich S, King MD, De Haan L, Veltman DJ, Assogna F, Banaj N, de Rossi P, Iorio M, Piras F, Spalletta G, Pomarol-Clotet E, Kelly S, Ciufolini S, Radua J, Murray R, Marques TR, Simmons A, Borgwardt S, Schönborn-Harrisberger F, Riecher-Rössler A, Smieskova R, Alpert KI, Bertolino A, Bonvino A, Di Giorgio A, Neilson E, Mayer AR, Yun JY, Cannon DM, Lebedeva I, Tomyshev AS, Akhadov T, Kaleda V, Fatouros-Bergman H, Flyckt L, Karolinska Schizophrenia Project (KaSP), Rosa PGP, Serpa MH, Zanetti MV, Hoschl C, Skoch A, Spaniel F, Tomecek D, McIntosh AM, Whalley HC, Knöchel C, et alvan Erp TGM, Walton E, Hibar DP, Schmaal L, Jiang W, Glahn DC, Pearlson GD, Yao N, Fukunaga M, Hashimoto R, Okada N, Yamamori H, Clark VP, Mueller BA, de Zwarte SMC, Ophoff RA, van Haren NEM, Andreassen OA, Gurholt TP, Gruber O, Kraemer B, Richter A, Calhoun VD, Crespo-Facorro B, Roiz-Santiañez R, Tordesillas-Gutiérrez D, Loughland C, Catts S, Fullerton JM, Green MJ, Henskens F, Jablensky A, Mowry BJ, Pantelis C, Quidé Y, Schall U, Scott RJ, Cairns MJ, Seal M, Tooney PA, Rasser PE, Cooper G, Weickert CS, Weickert TW, Hong E, Kochunov P, Gur RE, Gur RC, Ford JM, Macciardi F, Mathalon DH, Potkin SG, Preda A, Fan F, Ehrlich S, King MD, De Haan L, Veltman DJ, Assogna F, Banaj N, de Rossi P, Iorio M, Piras F, Spalletta G, Pomarol-Clotet E, Kelly S, Ciufolini S, Radua J, Murray R, Marques TR, Simmons A, Borgwardt S, Schönborn-Harrisberger F, Riecher-Rössler A, Smieskova R, Alpert KI, Bertolino A, Bonvino A, Di Giorgio A, Neilson E, Mayer AR, Yun JY, Cannon DM, Lebedeva I, Tomyshev AS, Akhadov T, Kaleda V, Fatouros-Bergman H, Flyckt L, Karolinska Schizophrenia Project (KaSP), Rosa PGP, Serpa MH, Zanetti MV, Hoschl C, Skoch A, Spaniel F, Tomecek D, McIntosh AM, Whalley HC, Knöchel C, Oertel-Knöchel V, Howells FM, Stein DJ, Temmingh HS, Uhlmann A, Lopez-Jaramillo C, Dima D, Faskowitz JI, Gutman BA, Jahanshad N, Thompson PM, Turner JA. Reply to: New Meta- and Mega-analyses of Magnetic Resonance Imaging Findings in Schizophrenia: Do They Really Increase Our Knowledge About the Nature of the Disease Process? Biol Psychiatry 2019; 85:e35-e39. [PMID: 30470561 PMCID: PMC7041557 DOI: 10.1016/j.biopsych.2018.10.003] [Show More Authors] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/05/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Theo GM van Erp
- Department of Psychiatry and Human Behavior, University of
California, Irvine, Irvine, CA, USA,Corresponding Author: Theo G.M. van Erp, Clinical
Translational Neuroscience Laboratory, Department of Psychiatry and Human
Behavior, School of Medicine, University of California Irvine, 5251 California
Avenue, Suite 240, Irvine, CA 92617, voice: (949) 824-3331,
| | - Esther Walton
- Medical Research Council Integrative Epidemiology Unit and
Bristol Medical School, Population Health Sciences, University of Bristol, United
Kingdom
| | - Derrek P Hibar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging
& Informatics Institute, Keck School of Medicine of the University of Southern
California, Marina del Rey, CA, USA,Janssen Research & Development, San Diego, CA,
USA
| | - Lianne Schmaal
- Orygen, The National Centre of Excellence in Youth Mental
Health, Melbourne, VIC, Australia,Centre for Youth Mental Health, The University of
Melbourne, Melbourne, VIC, Australia,Department of Psychiatry and Amsterdam Neuroscience, VU
University Medical Center, Amsterdam, The Netherlands
| | - Wenhao Jiang
- Department of Psychology, Georgia State University,
Atlanta, GA, USA
| | - David C Glahn
- Department of Psychiatry, Yale University, New Haven, CT,
USA,Olin Neuropsychiatric Research Center, Institute of
Living, Hartford Hospital, Hartford, CT, USA
| | - Godfrey D Pearlson
- Department of Psychiatry, Yale University, New Haven, CT,
USA,Olin Neuropsychiatric Research Center, Institute of
Living, Hartford Hospital, Hartford, CT, USA
| | - Nailin Yao
- Department of Psychiatry, Yale University, New Haven, CT,
USA,Olin Neuropsychiatric Research Center, Institute of
Living, Hartford Hospital, Hartford, CT, USA
| | - Masaki Fukunaga
- Division of Cerebral Integration, National Institute for
Physiological Sciences, Okazaki, Aichi, Japan
| | - Ryota Hashimoto
- Molecular Research Center for Children's Mental
Development, United Graduate School of Child Development, Osaka University, Suita,
Osaka, Japan,Department of Psychiatry, Osaka University Graduate
School of Medicine, Suita, Osaka, Japan
| | - Naohiro Okada
- Department of Neuropsychiatry, Graduate school of
Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hidenaga Yamamori
- Department of Psychiatry, Osaka University Graduate
School of Medicine, Suita, Osaka, Japan
| | - Vincent P Clark
- University of New Mexico, Albuquerque, NM, USA,Mind Research Network, Albuquerque, NM, USA
| | - Bryon A Mueller
- Department of Psychiatry, University of Minnesota,
Minneapolis, MN, USA
| | - Sonja MC de Zwarte
- Department of Psychiatry and Brain Center Rudolf Magnus,
University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel A Ophoff
- Department of Psychiatry and Brain Center Rudolf Magnus,
University Medical Center Utrecht, Utrecht, The Netherlands,University of California Los Angeles Center for
Neurobehavioral Genetics, Los Angeles, CA, USA
| | - Neeltje EM van Haren
- Department of Psychiatry and Brain Center Rudolf Magnus,
University Medical Center Utrecht, Utrecht, The Netherlands,Department of child and adolescent
psychiatry/psychology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT),
K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine,
University of Oslo, Oslo, Norway,Norwegian Centre for Mental Disorders Research (NORMENT),
K.G. Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction,
Oslo University Hospital, Oslo, Norway
| | - Tiril P Gurholt
- Norwegian Centre for Mental Disorders Research (NORMENT),
K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine,
University of Oslo, Oslo, Norway,Department of Psychiatric Research, Diakonhjemmet
Hospital, Oslo, Norway
| | - Oliver Gruber
- Section for Experimental Psychopathology and
Neuroimaging, Department of General Psychiatry, Heidelberg University Hospital,
Heidelberg, Germany,Center for Translational Research in Systems Neuroscience
and Psychiatry, Department of Psychiatry, Georg August University, Göttingen,
Germany
| | - Bernd Kraemer
- Section for Experimental Psychopathology and
Neuroimaging, Department of General Psychiatry, Heidelberg University Hospital,
Heidelberg, Germany,Center for Translational Research in Systems Neuroscience
and Psychiatry, Department of Psychiatry, Georg August University, Göttingen,
Germany
| | - Anja Richter
- Section for Experimental Psychopathology and
Neuroimaging, Department of General Psychiatry, Heidelberg University Hospital,
Heidelberg, Germany,Center for Translational Research in Systems Neuroscience
and Psychiatry, Department of Psychiatry, Georg August University, Göttingen,
Germany
| | - Vince D Calhoun
- University of New Mexico, Albuquerque, NM, USA,Mind Research Network, Albuquerque, NM, USA
| | - Benedicto Crespo-Facorro
- Department of Psychiatry, University Hospital
Marqués de Valdecilla, School of Medicine, University of Cantabria-Valdecilla
Biomedical Research Institute, Marqués de Valdecilla Research Institute
(IDIVAL), Santander, Spain,Centro Investigación Biomédica en Red de
Salud Mental (CIBERSAM), Santander, Spain
| | - Roberto Roiz-Santiañez
- Department of Psychiatry, University Hospital
Marqués de Valdecilla, School of Medicine, University of Cantabria-Valdecilla
Biomedical Research Institute, Marqués de Valdecilla Research Institute
(IDIVAL), Santander, Spain,Centro Investigación Biomédica en Red de
Salud Mental (CIBERSAM), Santander, Spain
| | - Diana Tordesillas-Gutiérrez
- Department of Psychiatry, University Hospital
Marqués de Valdecilla, School of Medicine, University of Cantabria-Valdecilla
Biomedical Research Institute, Marqués de Valdecilla Research Institute
(IDIVAL), Santander, Spain,Centro Investigación Biomédica en Red de
Salud Mental (CIBERSAM), Santander, Spain,Neuroimaging Unit.Technological Facilities, Valdecilla
Biomedical Research Institute IDIVAL, Santander, Cantabria, Spain Dresden, Dresden,
Germany
| | - Carmel Loughland
- Hunter Medical Research Institute, Newcastle, NSW,
Australia,Priority Research Centre for Brain & Mental Health,
The University of Newcastle, Newcastle, NSW, Australia,Hunter New England Local Health District, Newcastle,
NSW, Australia
| | | | - Janice M Fullerton
- Neuroscience Research Australia, Sydney, NSW,
Australia,School of Medical Sciences, University of New South
Wales, Sydney, NSW, Australia
| | - Melissa J Green
- School of Psychiatry, University of New South Wales,
Sydney, NSW, Australia,Neuroscience Research Australia, Sydney, NSW,
Australia
| | - Frans Henskens
- Priority Research Center for Health Behaviour, The
University of Newcastle, Newcastle, NSW, Australia,Hunter Medical Research Institute, Newcastle, NSW,
Australia,School of Medicine and Public Health, The University of
Newcastle, Newcastle, NSW, Australia
| | | | - Bryan J Mowry
- Queensland Brain Institute, The University of Queensland,
Brisbane, QLD, Australia,Queensland Centre for Mental Health Research, The
University of Queensland, Brisbane, QLD, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, University of Melbourne
& Melbourne Health, Melbourne, VIC, Australia,Florey Institute of Neuroscience and Mental Health,
University of Melbourne, VIC, Australia
| | - Yann Quidé
- School of Psychiatry, University of New South Wales,
Sydney, NSW, Australia,Neuroscience Research Australia, Sydney, NSW,
Australia
| | - Ulrich Schall
- Priority Research Centres for Brain & Mental Health
and Grow Up Well, The University of Newcastle, Newcastle, NSW, Australia,Hunter Medical Research Institute, Newcastle, NSW,
Australia
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, The
University of Newcastle, Newcastle, NSW, Australia,Hunter Medical Research Institute, Newcastle, NSW,
Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The
University of Newcastle, Newcastle, NSW, Australia,Hunter Medical Research Institute, Newcastle, NSW,
Australia
| | - Marc Seal
- Murdoch Children's Research Institute, Melbourne,
VIC, Australia
| | - Paul A Tooney
- School of Biomedical Sciences and Pharmacy, The
University of Newcastle, Newcastle, NSW, Australia,Hunter Medical Research Institute, Newcastle, NSW,
Australia,Priority Research Centre for Brain & Mental Health,
The University of Newcastle, Newcastle, NSW, Australia
| | - Paul E Rasser
- Priority Research Centre for Brain & Mental Health,
The University of Newcastle, Newcastle, NSW, Australia
| | - Gavin Cooper
- Priority Research Centre for Brain & Mental Health,
The University of Newcastle, Newcastle, NSW, Australia
| | - Cynthia Shannon Weickert
- School of Psychiatry, University of New South Wales,
Sydney, NSW, Australia,Neuroscience Research Australia, Sydney, NSW,
Australia
| | - Thomas W Weickert
- School of Psychiatry, University of New South Wales,
Sydney, NSW, Australia,Neuroscience Research Australia, Sydney, NSW,
Australia
| | - Elliot Hong
- Maryland Psychiatric Research Center, University of
Maryland School of Medicine, Baltimore, MD, USA
| | - Peter Kochunov
- Maryland Psychiatric Research Center, University of
Maryland School of Medicine, Baltimore, MD, USA
| | - Raquel E Gur
- Department of Psychiatry, University of Pennsylvania,
Philadelphia, PA, USA
| | - Ruben C Gur
- Department of Psychiatry, University of Pennsylvania,
Philadelphia, PA, USA
| | - Judith M Ford
- Department of Psychiatry, University of California, San
Francisco, San Francisco, CA, USA,San Francisco VA Medical Center, San Francisco, CA,
USA
| | - Fabio Macciardi
- Department of Psychiatry and Human Behavior, University of
California, Irvine, Irvine, CA, USA
| | - Daniel H Mathalon
- Department of Psychiatry, University of California, San
Francisco, San Francisco, CA, USA,San Francisco VA Medical Center, San Francisco, CA,
USA
| | - Steven G Potkin
- Department of Psychiatry and Human Behavior, University of
California, Irvine, Irvine, CA, USA
| | - Adrian Preda
- Department of Psychiatry and Human Behavior, University of
California, Irvine, Irvine, CA, USA
| | - Fengmei Fan
- Psychiatry Research Center, Beijing Huilongguan Hospital,
Beijing, China
| | - Stefan Ehrlich
- Division of Psychological and Social Medicine and
Developmental Neurosciences, Faculty of Medicine, TU Dresden, Germany, Dresden,
Germany,Massachusetts General Hospital/ Harvard Medical School,
Athinoula A. Martinos Center for Biomedical Imaging, Psychiatric Neuroimaging
Research Program
| | | | - Lieuwe De Haan
- Department of psychiatry, Academic Medical Center,
University of Amsterdam, Amsterdam, The Netherlands
| | - Dick J Veltman
- Department of Psychiatry, Vrije Universiteit Medical
Center, Amsterdam, The Netherlands
| | - Francesca Assogna
- Laboratory of Neuropsychiatry, Department of Clinical and
Behavioral Neurology, Istituto Di Ricovero e Cura a Carattere Scientifico Santa
Lucia Foundation, Rome, Italy,Centro Fermi - Museo Storico della Fisica e Centro Studi
e Ricerche “Enrico Fermi”, Rome, Italy
| | - Nerisa Banaj
- Laboratory of Neuropsychiatry, Department of Clinical and
Behavioral Neurology, Istituto Di Ricovero e Cura a Carattere Scientifico Santa
Lucia Foundation, Rome, Italy
| | - Pietro de Rossi
- Laboratory of Neuropsychiatry, Department of Clinical and
Behavioral Neurology, Istituto Di Ricovero e Cura a Carattere Scientifico Santa
Lucia Foundation, Rome, Italy,Dipartimento di Neuroscienze, Salute Mentale e Organi di
Senso (NESMOS) Department, Faculty of Medicine and Psychology, University
“Sapienza” of Rome, Rome, Italy,Department of Neurology and Psychiatry, Sapienza
University of Rome, Rome, Italy
| | - Mariangela Iorio
- Laboratory of Neuropsychiatry, Department of Clinical and
Behavioral Neurology, Istituto Di Ricovero e Cura a Carattere Scientifico Santa
Lucia Foundation, Rome, Italy
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, Department of Clinical and
Behavioral Neurology, Istituto Di Ricovero e Cura a Carattere Scientifico Santa
Lucia Foundation, Rome, Italy,Centro Fermi - Museo Storico della Fisica e Centro Studi
e Ricerche “Enrico Fermi”, Rome, Italy
| | - Gianfranco Spalletta
- Laboratory of Neuropsychiatry, Department of Clinical and
Behavioral Neurology, Istituto Di Ricovero e Cura a Carattere Scientifico Santa
Lucia Foundation, Rome, Italy,Beth K. and Stuart C. Yudofsky Division of
Neuropsychiatry, Menninger Department of Psychiatry and Behavioral Sciences, Baylor
College of Medicine, Houston, Tx USA
| | - Edith Pomarol-Clotet
- Fundación para la Investigación y Docencia
Maria Angustias Giménez (FIDMAG) Germanes Hospitalaries Research Foundation,
Barcelona, Spain,Centro Investigación Biomédica en Red de
Salud Mental (CIBERSAM), Barcelona, Spain
| | - Sinead Kelly
- Department of Psychiatry, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, MA, USA,Psychiatry Neuroimaging Laboratory, Brigham and
Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simone Ciufolini
- Department of Psychosis Studies, Institute of Psychiatry,
Psychology and Neuroscience, King's College London, London, United
Kingdom
| | - Joaquim Radua
- Department of Clinical Neuroscience, Centre for
Psychiatric Research, Karolinska Institutet, Stockholm, Sweden,Fundación para la Investigación y Docencia
Maria Angustias Giménez (FIDMAG) Germanes Hospitalaries Research Foundation,
Barcelona, Spain,Centro Investigación Biomédica en Red de
Salud Mental (CIBERSAM), Barcelona, Spain,Department of Psychosis Studies, Institute of Psychiatry,
Psychology and Neuroscience, King's College London, London, United
Kingdom,Institut d'Investigacions Biomediques August Pi i
Sunyer (IDIBAPS), Barcelona, Spain
| | - Robin Murray
- Department of Psychosis Studies, Institute of Psychiatry,
Psychology and Neuroscience, King's College London, London, United
Kingdom
| | - Tiago Reis Marques
- Department of Psychosis Studies, Institute of Psychiatry,
Psychology and Neuroscience, King's College London, London, United
Kingdom
| | - Andrew Simmons
- Department of Psychosis Studies, Institute of Psychiatry,
Psychology and Neuroscience, King's College London, London, United
Kingdom
| | | | | | | | | | - Kathryn I Alpert
- Department of Psychiatry and Behavioral Sciences,
Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Alessandro Bertolino
- Department of Basic Medical Science, Neuroscience and
Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Aurora Bonvino
- Istituto Di Ricovero e Cura a Carattere Scientifico Casa
Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Annabella Di Giorgio
- Istituto Di Ricovero e Cura a Carattere Scientifico Casa
Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Emma Neilson
- Division of Psychiatry, University of Edinburgh,
Edinburgh, United Kingdom
| | | | - Je-Yeon Yun
- Seoul National University Hospital, Seoul, Republic of
Korea,Yeongeon Student Support Center, Seoul National
University College of Medicine, Seoul, Republic of Korea
| | - Dara M Cannon
- Centre for Neuroimaging & Cognitive Genomics (NICOG),
Clinical Neuroimaging Laboratory, National Centre for Biomedical Engineering Galway
Neuroscience Centre, College of Medicine Nursing and Health Sciences, National
University of Ireland Galway, H91 TK33 Galway, Ireland
| | | | | | - Tolibjohn Akhadov
- Children's Clinical and Research Institute of
Emergency Surgery and Trauma, Moscow, Russia
| | | | - Helena Fatouros-Bergman
- Centre for Psychiatry Research, Department of Clinical
Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm
County Council, Stockholm, Sweden
| | - Lena Flyckt
- Centre for Psychiatry Research, Department of Clinical
Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm
County Council, Stockholm, Sweden
| | | | - Pedro GP Rosa
- Laboratory of Psychiatric Neuroimaging (LIM 21),
Department of Psychiatry, Faculty of Medicine, University of São Paulo,
São Paulo, Brazil,Center for Interdisciplinary Research on Applied
Neurosciences (NAPNA), University of São Paulo, São Paulo,
Brazil
| | - Mauricio H Serpa
- Laboratory of Psychiatric Neuroimaging (LIM 21),
Department of Psychiatry, Faculty of Medicine, University of São Paulo,
São Paulo, Brazil,Center for Interdisciplinary Research on Applied
Neurosciences (NAPNA), University of São Paulo, São Paulo,
Brazil
| | - Marcus V Zanetti
- Laboratory of Psychiatric Neuroimaging (LIM 21),
Department of Psychiatry, Faculty of Medicine, University of São Paulo,
São Paulo, Brazil,Center for Interdisciplinary Research on Applied
Neurosciences (NAPNA), University of São Paulo, São Paulo,
Brazil
| | - Cyril Hoschl
- National Institute of Mental Health, Klecany, Czech
Republic
| | - Antonin Skoch
- National Institute of Mental Health, Klecany, Czech
Republic,MR Unit, Department of Diagnostic and Interventional
Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech
Republic
| | - Filip Spaniel
- National Institute of Mental Health, Klecany, Czech
Republic
| | - David Tomecek
- National Institute of Mental Health, Klecany, Czech
Republic,Institute of Computer Science, Czech Academy of
Sciences, Prague, Czech Republic,Faculty of Electrical Engineering, Czech Technical
University in Prague, Prague, Czech Republic
| | - Andrew M McIntosh
- Division of Psychiatry, University of Edinburgh,
Edinburgh, United Kingdom,Centre for Cognitive Ageing and Cognitive Epidemiology,
University of Edinburgh, Edinburgh, United Kingdom
| | - Heather C Whalley
- Division of Psychiatry, University of Edinburgh,
Edinburgh, United Kingdom
| | - Christian Knöchel
- Department of Psychiatry, Psychosomatic Medicine and
Psychotherapy, University Hospital Frankfurt, Goethe University Frankfurt,
Frankfurt, Germany
| | - Viola Oertel-Knöchel
- Department of Psychiatry, Psychosomatic Medicine and
Psychotherapy, University Hospital Frankfurt, Goethe University Frankfurt,
Frankfurt, Germany
| | - Fleur M Howells
- University of Cape Town Dept of Psychiatry, Groote
Schuur Hospital (J2), Cape Town South Africa
| | - Dan J Stein
- University of Cape Town Dept of Psychiatry, Groote
Schuur Hospital (J2), Cape Town South Africa,Medical Research Council Unit on Risk & Resilience
in Mental Disorders, Department of Psychiatry, University of Cape Town, Cape Town,
South Africa
| | - Henk S Temmingh
- University of Cape Town Dept of Psychiatry, Groote
Schuur Hospital (J2), Cape Town South Africa
| | - Anne Uhlmann
- University of Cape Town Dept of Psychiatry, Groote
Schuur Hospital (J2), Cape Town South Africa,MRC Unit on Risk & Resilience in Mental Disorders,
Department of Psychiatry, Stellenbosch University, Cape Town, South Africa
| | - Carlos Lopez-Jaramillo
- Research Group in Psychiatry, Department of Psychiatry,
Faculty of Medicine, Universidad de Antioquia, Medellin, Colombia
| | - Danai Dima
- Department of Psychology, City, University of London,
London, United Kingdom,Department of Neuroimaging, IOPPN, King's College
London, London, United Kingdom
| | - Joshua I Faskowitz
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging
& Informatics Institute, Keck School of Medicine of the University of Southern
California, Marina del Rey, CA, USA
| | - Boris A Gutman
- Department of Biomedical Engineering, Illinois Institute
of Technology, Chicago, Illinois
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging
& Informatics Institute, Keck School of Medicine of the University of Southern
California, Marina del Rey, CA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging
& Informatics Institute, Keck School of Medicine of the University of Southern
California, Marina del Rey, CA, USA
| | - Jessica A Turner
- Mind Research Network, Albuquerque, NM, USA,Imaging Genetics and Neuroinformatics Lab, Department of
Psychology, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
48
|
Klein M, Walters RK, Demontis D, Stein JL, Hibar DP, Adams HH, Bralten J, Roth Mota N, Schachar R, Sonuga-Barke E, Mattheisen M, Neale BM, Thompson PM, Medland SE, Børglum AD, Faraone SV, Arias-Vasquez A, Franke B. Genetic Markers of ADHD-Related Variations in Intracranial Volume. Am J Psychiatry 2019; 176:228-238. [PMID: 30818988 PMCID: PMC7780894 DOI: 10.1176/appi.ajp.2018.18020149] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Attention deficit hyperactivity disorder (ADHD) is a common and highly heritable neurodevelopmental disorder with a complex pathophysiology. Intracranial volume (ICV) and volumes of the nucleus accumbens, amygdala, caudate nucleus, hippocampus, and putamen are smaller in people with ADHD compared with healthy individuals. The authors investigated the overlap between common genetic variation associated with ADHD risk and these brain volume measures to identify underlying biological processes contributing to the disorder. METHODS The authors combined genome-wide association results from the largest available studies of ADHD (N=55,374) and brain volumes (N=11,221-24,704), using a set of complementary methods to investigate overlap at the level of global common variant genetic architecture and at the single variant level. RESULTS Analyses revealed a significant negative genetic correlation between ADHD and ICV (rg=-0.22). Meta-analysis of single variants revealed two significant loci of interest associated with both ADHD risk and ICV; four additional loci were identified for ADHD and volumes of the amygdala, caudate nucleus, and putamen. Exploratory gene-based and gene-set analyses in the ADHD-ICV meta-analytic data showed association with variation in neurite outgrowth-related genes. CONCLUSIONS This is the first genome-wide study to show significant genetic overlap between brain volume measures and ADHD, both on the global and the single variant level. Variants linked to smaller ICV were associated with increased ADHD risk. These findings can help us develop new hypotheses about biological mechanisms by which brain structure alterations may be involved in ADHD disease etiology.
Collapse
Affiliation(s)
- Marieke Klein
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Raymond K. Walters
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Ditte Demontis
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Jason L. Stein
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Derrek P. Hibar
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Hieab H. Adams
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Janita Bralten
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Nina Roth Mota
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Russell Schachar
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Edmund Sonuga-Barke
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Manuel Mattheisen
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Benjamin M. Neale
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Paul M. Thompson
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Sarah E. Medland
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Anders D. Børglum
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Stephen V. Faraone
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Alejandro Arias-Vasquez
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Barbara Franke
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| |
Collapse
|
49
|
Weinstein G, Davis-Plourde KL, Conner S, Himali JJ, Beiser AS, Lee A, Rawlings AM, Sedaghat S, Ding J, Moshier E, van Duijn CM, Beeri MS, Selvin E, Ikram MA, Launer LJ, Haan MN, Seshadri S. Association of metformin, sulfonylurea and insulin use with brain structure and function and risk of dementia and Alzheimer's disease: Pooled analysis from 5 cohorts. PLoS One 2019; 14:e0212293. [PMID: 30768625 PMCID: PMC6377188 DOI: 10.1371/journal.pone.0212293] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 01/30/2019] [Indexed: 12/12/2022] Open
Abstract
Objective To determine whether classes of diabetes medications are associated with cognitive health and dementia risk, above and beyond their glycemic control properties. Research design and methods Findings were pooled from 5 population-based cohorts: the Framingham Heart Study, the Rotterdam Study, the Atherosclerosis Risk in Communities (ARIC) Study, the Aging Gene-Environment Susceptibility-Reykjavik Study (AGES) and the Sacramento Area Latino Study on Aging (SALSA). Differences between users and non-users of insulin, metformin and sulfonylurea were assessed in each cohort for cognitive and brain MRI measures using linear regression models, and cognitive decline and dementia/AD risk using mixed effect models and Cox regression analyses, respectively. Findings were then pooled using meta-analytic techniques, including 3,590 individuals with diabetes for the prospective analysis. Results After adjusting for potential confounders including indices of glycemic control, insulin use was associated with increased risk of new-onset dementia (pooled HR (95% CI) = 1.58 (1.18, 2.12);p = 0.002) and with a greater decline in global cognitive function (β = -0.014±0.007;p = 0.045). The associations with incident dementia remained similar after further adjustment for renal function and excluding persons with diabetes whose treatment was life-style change only. Insulin use was not related to cognitive function nor to brain MRI measures. No significant associations were found between metformin or sulfonylurea use and outcomes of brain function and structure. There was no evidence of significant between-study heterogeneity. Conclusions Despite its advantages in controlling glycemic dysregulation and preventing complications, insulin treatment may be associated with increased adverse cognitive outcomes possibly due to a greater risk of hypoglycemia.
Collapse
Affiliation(s)
- Galit Weinstein
- School of Public Health, University of Haifa, Haifa, Israel
- * E-mail:
| | - Kendra L. Davis-Plourde
- Framingham Heart Study, Framingham, MA, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
| | - Sarah Conner
- Framingham Heart Study, Framingham, MA, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
| | - Jayandra J. Himali
- Framingham Heart Study, Framingham, MA, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States of America
| | - Alexa S. Beiser
- Framingham Heart Study, Framingham, MA, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States of America
| | - Anne Lee
- Department of Epidemiology & Biostatistics, University of California, San Francisco, California, United States of America
| | - Andreea M. Rawlings
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Sanaz Sedaghat
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Jie Ding
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
| | - Erin Moshier
- Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Michal S. Beeri
- Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Radiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
| | - Mary N. Haan
- Department of Epidemiology & Biostatistics, University of California, San Francisco, California, United States of America
| | - Sudha Seshadri
- Framingham Heart Study, Framingham, MA, United States of America
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States of America
| |
Collapse
|
50
|
Alexander-Bloch AF, Mathias SR, Fox PT, Olvera RL, Göring HHH, Duggirala R, Curran JE, Blangero J, Glahn DC. Human Cortical Thickness Organized into Genetically-determined Communities across Spatial Resolutions. Cereb Cortex 2019; 29:106-118. [PMID: 29190330 PMCID: PMC6676978 DOI: 10.1093/cercor/bhx309] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/19/2017] [Indexed: 12/13/2022] Open
Abstract
The cerebral cortex may be organized into anatomical genetic modules, communities of brain regions with shared genetic influences via pleiotropy. Such modules could represent novel phenotypes amenable to large-scale gene discovery. This modular structure was investigated with network analysis of in vivo MRI of extended pedigrees, revealing a "multiscale" structure where smaller and larger modules exist simultaneously and in partially overlapping fashion across spatial scales, in contrast to prior work suggesting a specific number of cortical thickness modules. Inter-regional genetic correlations, gene co-expression patterns and computational models indicate that two simple organizational principles account for a large proportion of the apparent complexity in the network of genetic correlations. First, regions are strongly genetically correlated with their homologs in the opposite cerebral hemisphere. Second, regions are strongly genetically correlated with nearby regions in the same hemisphere, with an initial steep decrease in genetic correlation with anatomical distance, followed by a more gradual decline. Understanding underlying organizational principles of genetic influence is a critical step towards a mechanistic model of how specific genes influence brain anatomy and mediate neuropsychiatric risk.
Collapse
Affiliation(s)
| | - Samuel R Mathias
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, USA
| | - Peter T Fox
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - Rene L Olvera
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - Harold H H Göring
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - Ravi Duggirala
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - Joanne E Curran
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - David C Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, USA
| |
Collapse
|