Population clustering of structural brain aging and its association with brain development

  1. Haojing Duan
  2. Runye Shi
  3. Jujiao Kang
  4. Tobias Banaschewski
  5. Arun LW Bokde
  6. Christian Büchel
  7. Sylvane Desrivières
  8. Herta Flor
  9. Antoine Grigis
  10. Hugh Garavan
  11. Penny A Gowland
  12. Andreas Heinz
  13. Rüdiger Brühl
  14. Jean-Luc Martinot
  15. Marie-Laure Paillère Martinot
  16. Eric Artiges
  17. Frauke Nees
  18. Dimitri Papadopoulos Orfanos
  19. Luise Poustka
  20. Sarah Hohmann
  21. Nathalie Nathalie Holz
  22. Juliane Fröhner
  23. Michael N Smolka
  24. Nilakshi Vaidya
  25. Henrik Walter
  26. Robert Whelan
  27. Gunter Schumann
  28. Xiaolei Lin  Is a corresponding author
  29. Jianfeng Feng  Is a corresponding author
  1. Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China
  2. Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
  3. School of Data Science, Fudan University, China
  4. Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Germany
  5. Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland
  6. University Medical Centre Hamburg-Eppendorf, Germany
  7. Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, United Kingdom
  8. Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Germany
  9. Department of Psychology, School of Social Sciences, University of Mannheim, Germany
  10. NeuroSpin, CEA, Université Paris-Saclay, France
  11. Departments of Psychiatry and Psychology, University of Vermont, United States
  12. Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, United Kingdom
  13. Department of Psychiatry and Psychotherapy CCM, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
  14. Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Germany
  15. Institut National de la Santé et de la Recherche Médicale, INSERM U1299 "Developmental Trajectories and Psychiatry", Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, France
  16. AP-HP. Sorbonne Université, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, France
  17. Psychiatry Department, EPS Barthélémy Durand, France
  18. Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein, Kiel University, Germany
  19. Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre, Germany
  20. Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Germany
  21. Department of Psychiatry and Neurosciences, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
  22. School of Psychology and Global Brain Health Institute, Trinity College Dublin, Ireland
  23. Centre for Population Neuroscience and Stratified Medicine (PONS Centre), ISTBI, Fudan University, China
  24. Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Germany
  25. Huashan Institute of Medicine, Huashan Hospital affiliated to Fudan University, China
  26. MOE Frontiers Center for Brain Science, Fudan University, China
  27. Zhangjiang Fudan International Innovation Center, China
  28. Department of Computer Science, University of Warwick, United Kingdom

eLife assessment

Duan et al analyzed brain imaging data in UKBK and divided structural brain aging into two groups, revealing that one group is more vulnerable to aging and brain-related diseases compared to the other group. Such subtyping could be valuable and utilized in predicting and diagnosing cognitive decline and neurodegenerative brain disorders in the future. This discovery, supported by solid evidence, harbors a substantial impacts in aging and brain structure and function.

https://doi.org/10.7554/eLife.94970.3.sa0

Abstract

Structural brain aging has demonstrated strong inter-individual heterogeneity and mirroring patterns with brain development. However, due to the lack of large-scale longitudinal neuroimaging studies, most of the existing research focused on the cross-sectional changes of brain aging. In this investigation, we present a data-driven approach that incorporate both cross-sectional changes and longitudinal trajectories of structural brain aging and identified two brain aging patterns among 37,013 healthy participants from UK Biobank. Participants with accelerated brain aging also demonstrated accelerated biological aging, cognitive decline and increased genetic susceptibilities to major neuropsychiatric disorders. Further, by integrating longitudinal neuroimaging studies from a multi-center adolescent cohort, we validated the ‘last in, first out’ mirroring hypothesis and identified brain regions with manifested mirroring patterns between brain aging and brain development. Genomic analyses revealed risk loci and genes contributing to accelerated brain aging and delayed brain development, providing molecular basis for elucidating the biological mechanisms underlying brain aging and related disorders.

Introduction

The structure of the brain undergoes continual changes throughout the entire lifespan, with structural brain alterations intimately linking brain development and brain aging (Fjell and Walhovd, 2010; Shaw et al., 2008). Brain aging is a progressive process that often co-occurs with biological aging and declines of cognitive functions (Elliott et al., 2021; Mattson and Arumugam, 2018; Park and Reuter-Lorenz, 2009), which contribute to the onset and acceleration of neurodegenerative (Mariani et al., 2005) and neuropsychiatric disorders (Kaufmann et al., 2019). Studies on healthy brain aging have revealed significant inter-individual heterogeneity in the patterns of neuroanatomical changes (Raz et al., 2010; Raz and Rodrigue, 2006). Therefore, examining the patterns of structural brain aging and its associations with cognitive decline is of paramount importance in understanding the diverse biological mechanisms of age-related neuropsychiatric disorders.

Despite the fact that there exist large differences between brain development and brain aging (Courchesne et al., 2000), a discernible association between these two processes remains evident. Direct comparisons of brain development and brain aging using structural MRI indicated a ‘last in, first out’ mirroring pattern, where brain regions develop relatively late during adolescence demonstrated accelerated degeneration in older ages (McGinnis et al., 2011; Tamnes et al., 2013). In addition, brain regions with strong mirroring effects showed increased vulnerability to neurodegenerative and neuropsychiatric disorders, including Alzheimer’s disease and schizophrenia (Douaud et al., 2014). However, due to the lack of large-scale longitudinal MRI studies during adolescence and mid-to-late adulthood, validation of the ‘last in, first out’ mirroring hypothesis remains unavailable.

Prior investigations have largely focused on regional and cross-sectional changes of brain aging (Raz and Rodrigue, 2006; Douaud et al., 2014; Suzuki et al., 2019), with relatively few studies exploring longitudinal trajectories of brain aging and its associations with brain development (Raz et al., 2010; Fjell et al., 2015; Nyberg et al., 2023). In this article, we present a data-driven approach to examine the population clustering of longitudinal brain aging trajectories using structure MRI data obtained from 37,013 healthy individuals during mid-to-late adulthood (44–82 years), and explore its association with biological aging, cognitive decline and susceptibilities for neuropsychiatric disorders. Further, mirroring patterns between longitudinal brain development and brain aging are investigated by comparing the region-specific aging / developmental trajectories, and manifestation of the mirroring patterns are investigated across the whole-brain and among participants with different brain aging patterns. Genomic analyses are conducted to reveal risk loci and genes associated with accelerated brain aging and delayed brain development.

Results

Longitudinal trajectories of whole-brain gray matter volume in mid-to-late adulthood define two brain aging patterns

Figure 1 provides the data sources, analytical workflow and research methodology of this study. After the sample selection process (Appendix 1—figure 1, Appendix 1—table 1 and 2), longitudinal grey matter volume (GMV) trajectories in 40 ROIs (33 cortical and 7 subcortical ROIs, see Appendix 1—table 3) were estimated for each of the 37,013 healthy participants in UK Biobank. The first 15 principal components derived from dimensionality reduction via principal component analysis were used in the clustering analysis (see Methods; Alexander-Bloch et al., 2013; Whitwell et al., 2009). Two brain aging patterns were identified, where 18,929 (51.1%) participants with the first brain aging pattern (pattern 1) had higher total GMV at baseline and a slower rate of GMV decrease over time, and the remaining participants with the second pattern (pattern 2) had lower total GMV at baseline and a faster rate of GMV decrease (Figure 2a). Comparing the region-specific rate of GMV decrease, pattern 2 showed a more rapid GMV decrease in medial occipital (lingual gyrus, cuneus, and pericalcarine cortex) and medial temporal (entorhinal cortex, parahippocampal gyrus) regions (Figure 2b and c and Appendix 1—figure 2), which had the largest loadings in the second and third principal components (Appendix 1—table 4). These two patterns can be clearly stratified by both linear and non-linear dimensionality reduction methods, indicating distinct structural differences in brain aging between patterns (Appendix 1—figure 3). Sample characteristics of these 37,013 UK Biobank participants stratified by brain aging patterns are summarized in Appendix 1—table 5. Overall, participants with different brain aging patterns had similar distributions with regard to age, sex, ethnicity, smoking status, Townsend deprivation index (TDI), body mass index (BMI) and years of schooling.

Overview of the study workflow.

(a) Population cohorts (UK Biobank and IMAGEN) and data sources (brain imaging, biological aging biomarkers, cognitive functions, genomic data) involved in this study. (b) Brain aging patterns were identified using longitudinal trajectories of the whole brain GMV, which enabled the capturing of long-term and individualized variations compared to only use cross-sectional data, and associations between brain aging patterns and other measurements (biological aging, cognitive functions and PRS of major neuropsychiatric disorders) were investigated. (c) Mirroring patterns between brain aging and brain development was investigated using z-transformed brain volumetric change map and gene expression analysis.

Global (a) and selected regional (b, c) cortical gray matter volume rate of change among participants with brain aging patterns 1 (red) and 2 (blue).

Rates of volumetric change for total gray matter and each ROI were estimated using GAMM, which incorporates both cross-sectional between-subject variation and longitudinal within-subject variation from 40,921 observations and 37,013 participants. Covariates include sex, assessment center, handedness, ethnic, and ICV. Shaded areas around the fit line denotes 95% CI.

Figure 2—source data 1

Related to Figure 2.

Global (a) and selected regional (b, c) cortical gray matter volume rate of change among participants with brain aging patterns 1 (red) and 2 (blue).

https://cdn.elifesciences.org/articles/94970/elife-94970-fig2-data1-v1.xlsx

Brain aging patterns were significantly associated with biological aging

To explore the relationships between structural brain aging and biological aging, we investigated the distribution of aging biomarkers, such as telomere length and PhenoAge (Levine et al., 2018), across brain aging patterns identified above (Figure 3 and Appendix 1—table 6). Compared to pattern 1, participants in pattern 2 with more rapid GMV decrease had shorter leucocyte telomere length (p=0.009, Cohen’s D=–0.028) and this association remained consistent after adjusting for sex, age, ethnic, BMI, smoking status and alcohol intake frequency (Demanelis et al., 2020). Next, we examined PhenoAge, which was developed as an aging biomarker incorporating composite clinical and biochemical data (Levine et al., 2018), and observed higher PhenoAge among participants with brain aging pattern 2 compared to pattern 1 (p=0.019, Cohen’s D=0.027). Again, the association remained significant after adjusting for sex, age, ethnic, BMI, smoking status, alcohol intake frequency and education years (p=3.05×10–15, Cohen’s D=0.092). Group differences in terms of each individual component of PhenoAge (including albumin, creatinine, glucose, c-reactive protein, lymphocytes percentage, mean corpuscular volume, erythocyte distribution width, alkaline phosphatase and leukocyte count) were also investigated and results were consistent with PhenoAge (Appendix 1—figure 4).

Distributions of biological aging biomarkers (leucocyte telomere length (LTL) and PhenoAge) among participants with brain aging patterns 1 and 2.

Boxes represent the interquartile range (IQR), lines within the boxes indicate the median. Two-sided p values were obtained by comparing LTL or PhenoAge Levine et al., 2018 between brain aging patterns using unadjusted multivariate linear regression models. Results remained significant when adjusting for sex, age, ethnic, BMI, smoking status and alcohol intake frequency in the LTL model Demanelis et al., 2020 and sex, age, ethnic, BMI, smoking status, alcohol frequency and education years in PhenoAge model. Stars indicate statistical significance after Bonferroni correction. ****: p ≤ 0.0001, *: p ≤ 0.05.

Figure 3—source data 1

Related to Figure 3.

Distributions of biological aging biomarkers (leucocyte telomere length (LTL) and PhenoAge) among participants with brain aging patterns 1 and 2.

https://cdn.elifesciences.org/articles/94970/elife-94970-fig3-data1-v1.xlsx

Accelerated brain aging was associated with cognitive decline and increased genetic susceptibilities to attention-deficit/hyperactivity disorder and delayed brain development

Next, we conducted comprehensive comparisons of cognitive functions between participants with different brain aging patterns. In general, those with brain aging pattern 2 (lower baseline total GMV and more rapid GMV decrease) exhibited worse cognitive performances compared to pattern 1. Specifically, brain aging pattern 2 showed lower numbers of correct pairs matching (p=0.006, Cohen’s D=–0.029), worse prospective memory (OR = 0.943, 95% CI [0.891, 0.999]), lower fluid intelligence (p<1.00×10–20, Cohen’s D=–0.102), and worse numeric memory (p=5.97×10–11, Cohen’s D=–0.082). No statistically significant differences were observed in terms of the reaction time (p=0.99) and prospective memory (p=0.052) between these two brain aging patterns after FDR correction. Results were consistent when using models adjusted for sex, age, and socioeconomic status (TDI, education and income; Foster et al., 2018; Townsend et al., 2023; Figure 4). Full results demonstrating the associations between brain aging patterns and cognitive functions are presented in Appendix 1—table 7.

Effect size (Cohen’s D or odds ratio) for comparing the cognitive functions between participants with brain aging patterns 1 and 2.

Results were adjusted such that negative Cohen’s D and Odds Ratio less than 1 indicate worse cognitive performances in brain aging pattern 2 compared to pattern 1. Width of the lines extending from the center point represent 95% confidence interval. Two-sided p values were obtained using both unadjusted and adjusted (for sex, age, and TDI, education and income) multivariate regression models. Stars indicate statistical significance after FDR correction for 11 comparisons. ****: p ≤ 0.0001, ***: p ≤ 0.001, **: p ≤ 0.01, ns: p>0.05.

Figure 4—source data 1

Related to Figure 4.

Effect size (Cohen’s D or odds ratio) for comparing the cognitive functions between participants with brain aging patterns 1 and 2.

https://cdn.elifesciences.org/articles/94970/elife-94970-fig4-data1-v1.xlsx

Having observed cognitive decline among participants with accelerated brain aging pattern, we next investigated whether brain aging patterns were associated with genetic vulnerability to major neuropsychiatric disorders. Since current GWAS are under-powered for attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorders (ASD) and the difficulty in identifying genetic variants was likely due to their polygenic nature, we calculated the corresponding polygenic risk scores (PRS) using multiple p value thresholds. This approach enabled robust investigation of the association between genetic susceptibility of neuropsychiatric disorders and brain imaging phenotypes. PRS for major neuro-developmental disorders including attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorders (ASD), neurodegenerative diseases including Alzheimer’s disease (AD) and Parkinson’s disease (PD), neuropsychiatric disorders including bipolar disorder (BIP), major depressive disorder (MDD), and schizophrenia (SCZ), and delayed structural brain development (GWAS from an unpublished longitudinal neuroimaging study) (Shi et al., 2023) were calculated for each participant using multiple p value thresholds (from 0.005 to 0.5 at intervals of 0.005) and results were then averaged over all thresholds (Figure 5). The primary GWAS datasets used for calculating the PRS were listed in Appendix 1—table 8. Overall, we observed increased genetic susceptibility to ADHD (p=0.040) and delayed brain development (p=1.4810–6) among participants with brain aging pattern 2 after FDR correction, while no statistically significant differences were observed for ASD, AD, PD, BIP, MDD, and SCZ (Figure 5). Details regarding the genetic liability to other common diseases and phenotypes using enhanced PRS from UK Biobank are displayed in Appendix 1tables 9 and 10.

Participants with accelerated brain aging (brain aging pattern 2) had significantly increased genetic liability to ADHD and delayed brain development.

Polygenic risk score (PRS) for ADHD, ASD, AD, PD, BIP, MDD, SCZ and delayed brain development (unpublished GWAS) were calculated at different p-value thresholds from 0.005 to 0.5 at an interval of 0.005. Vertical axis represents negative logarithm of P values comparing PRS in brain aging pattern 2 relative to pattern 1. Red horizontal dashed line indicates FDR corrected p value of 0.05. Colors represent traits and dots within the same color represent different p value thresholds. The trigonometric symbol indicates the average PRS across all p-value thresholds for the same trait.

Figure 5—source data 1

Related to Figure 5.

Participants with accelerated brain aging (brain aging pattern 2) had significantly increased genetic liability to ADHD and delayed brain development.

https://cdn.elifesciences.org/articles/94970/elife-94970-fig5-data1-v1.xlsx

Genome Wide Association Studies (GWAS) identified significant genetic loci associated with accelerated brain aging

Having observed significant associations between brain aging patterns and cognitive performances / genetic liabilities to major neurodevelopmental disorders, we further investigated if there exist genetic variants contributing to individualized brain aging phenotype. We conducted genome-wide association studies (GWAS) using estimated total GMV at 60 years old as the phenotype. This phenotype was derived by adding individual specific deviations to the population averaged total GMV, thus providing additional information compared to studies using only cross-sectional neuroimaging phenotypes.

Six independent single nucleotide polymorphisms (SNPs) were identified at genome-wide significance level (p<5×10–8) (Figure 6) and were subsequently mapped to genes using NCBI, Ensembl and UCSC Genome Browser database (Appendix 1—table 11). Among them, two SNPs (rs10835187 and rs779233904) were also found to be associated with multiple brain imaging phenotypes in previous studies (Smith et al., 2021), such as regional and tissue volume, cortical area and white matter tract measurements. Compared to the GWAS using global gray matter volume as the phenotype, our GWAS revealed additional signal in chromosome 7 (rs7776725), which was mapped to the intron of FAM3C and encodes a secreted protein involved in pancreatic cancer (Grønborg et al., 2006) and Alzheimer’s disease (Liu et al., 2016). This signal was further validated to be associated with specific brain aging mode by another study using a data-driven decomposition approach (Smith et al., 2020). In addition, another significant loci (rs10835187, p=1.11×10–13) is an intergenic variant between gene LGR4-AS1 and LIN7C, and was reported to be associated with bone density and brain volume measurement (Smith et al., 2021; Estrada et al., 2012). LIN7C encodes the Lin-7C protein, which is involved in the localization and stabilization of ion channels in polarized cells, such as neurons and epithelial cells (Bohl et al., 2007; Kaech et al., 1998). Previous study has revealed the association of both allelic and haplotypic variations in the LIN7C gene with ADHD (Lanktree et al., 2008).

Genome-wide association study (GWAS) identified 6 independent SNPs associated with accelerated brain aging.

Total GMV at 60 years old was estimated for each participant using mixed effect models allowing for individualized baseline GMV and GMV change rate, and was used as the phenotype in the GWAS. (a) At genome-wide significance level (p=5×10–8, red dashed line), rs10835187 and rs7776725 loci were identified to be associated with accelerated brain aging. (b) Quantile–quantile plot showed that the most significant p values deviate from the null, suggesting that results are not unduly inflated.

Mirroring patterns between brain aging and brain development

Having observed significant associations between brain aging and genetic susceptibility to neurodevelopmental disorders, we are now interested in examining the mirroring patterns between brain aging and brain development in the whole population, and whether these mirroring patterns were more pronounced in those with accelerated brain aging. Adolescents in the IMAGEN cohort showed more rapid GMV decrease in the frontal and parietal lobes, especially the frontal pole, superior frontal gyrus, rostral middle frontal gyrus, inferior parietal lobule and superior parietal lobule, while those in their mid-to-late adulthood showed more accelerated GMV decrease in the temporal lobe, including medial orbitofrontal cortex, inferior parietal lobule and lateral occipital sulcus (Figure 7a). The mirroring patterns (with slower GMV decrease during brain development and more rapid GMV decrease during brain aging) were particularly prominent in inferior temporal gyrus, caudal anterior cingulate cortex, fusiform cortex, middle temporal gyrus and rostral anterior cingulate cortex (Figure 7b). The regional mirroring patterns became weaker when we focus on late brain aging at age 75 years old, especially in the frontal lobe and cingulate cortex. Further, mirroring patterns were represented more prominently in participants with brain aging pattern 2, where stronger mirroring between brain aging and brain development was observed in frontotemporal area, including lateral occipital sulcus and lingual gyrus (Figure 7c).

The ‘last in, first out’ mirroring patterns between brain development and brain aging.

(a) The annual percentage volume change (APC) was calculated for each ROI and standardized across the whole brain in adolescents (IMAGEN, left) and mid-to-late aged adults (UK Biobank, right), respectively. For adolescents, ROIs of in red indicate delayed structural brain development, while for mid-to-late aged adults, ROIs in blue indicate accelerated structural brain aging. (b) Estimated APC in brain development versus early aging (55 years old, left), and versus late aging (75 years old, right). ROIs in red indicate faster GMV decrease during brain aging and slower GMV decrease during brain development, that is stronger mirroring effects between brain development and brain aging. (c) Mirroring patterns between brain development and brain aging were more manifested in participants with accelerated aging (brain aging pattern 2). The arrows point to ROIs with more pronounced mirroring patterns in each subfigure.

Figure 7—source data 1

Related to Figure 7.

The ‘last in, first out’ mirroring patterns between brain development and brain aging.

https://cdn.elifesciences.org/articles/94970/elife-94970-fig7-data1-v1.xlsx

Gene expression profiles were associated with delayed brain development and accelerated brain aging

The Allen Human Brain Atlas (AHBA) transcriptomic dataset (http://human.brain-map.org) were used to obtain the spatial correlation between gene expression profiles across cortex and structural brain development/aging via partial least square (PLS) regression. The first PLS component explained 24.7% and 53.6% of the GMV change during brain development (estimated at age 15y, rspearman = 0.51, Ppermutation = 0.03) and brain aging (estimated at age 55y, rspearman = 0.49, Ppermutation = 1.510–4), respectively. Seventeen of the 45 genes mapped to GWAS significant SNP were found in AHBA, with LGR4 (rspearman = 0.56, Ppermutation <0.001) significantly associated with delayed brain development and ESR1 (rspearman = 0.53, Ppermutation <0.001) and FAM3C (rspearman = –0.37, Ppermutation = 0.004) significantly associated with accelerated brain aging. BDNF-AS was positively associated with both delayed brain development and accelerated brain aging after spatial permutation test (Appendix 1—table 12 and 13).

Next, we screened the genes based on their contributions and effect directions to the first PLS components in brain development and brain aging. 990 and 2293 genes were identified to be positively associated with brain development and negatively associated with brain aging at FDR corrected p value of 0.005, respectively, representing gene expressions associated with delayed brain development and accelerated brain aging. These genes were then tested for enrichment of GO biological processes and KEGG pathways. Genes associated with delayed brain development showed significant enrichment in ‘regulation of trans-synaptic signaling’, ‘forebrain development’, ‘signal release’ and ‘cAMP signaling pathway’ (Figure 8a), and genes associated with accelerated brain aging showed significant enrichment in ‘macroautophagy’, ‘establishment of protein localization to organelle’, ‘histone modification’, and ‘pathways of neurodegeneration – multiple diseases’ (Figure 8b). Full results of the gene set enrichment analysis were provided in Appendix 1—figure 5. In summary, the analyses from using the databases of GO biological processes and KEGG Pathways indicate synaptic transmission as an important process in the common mechanisms of brain development and aging, and cellular processes (autophagy), as well as the progression of neurodegenerative diseases, are important processes in the mechanisms of brain aging.

Functional enrichment of gene transcripts significantly associated with delayed brain development and accelerated brain aging.

(a) 990 genes were spatially correlated with the first PLS component of delayed structural brain development, and were enriched for trans-synaptic signal regulation, forebrain development, signal release and cAMP signaling pathway. (b) 2293 genes were spatially correlated the first PLS component of accelerated structural brain aging, and were enriched for macroautophagy, pathways of neurodegeneration, establishment of protein localization to organelle and histone modification. Size of the circle represents number of genes in each term and P values were corrected using FDR for multiple comparisons.

Figure 8—source data 1

Related to Figure 8.

Functional enrichment of gene transcripts significantly associated with delayed brain development and accelerated brain aging. Each gene’s contribution to the first PLS component for IMAGEN and UKB.

https://cdn.elifesciences.org/articles/94970/elife-94970-fig8-data1-v1.xlsx
Figure 8—source data 2

Related to Figure 8.

Functional enrichment of gene transcripts significantly associated with delayed brain development and accelerated brain aging. Gene set enrichment for genes spatially positively correlated with the first PLS component in IMAGEN and negatively correlated with the first PLS component in UKB.

https://cdn.elifesciences.org/articles/94970/elife-94970-fig8-data2-v1.xlsx

Discussion

In this study, we adopted a data-driven approach and revealed two distinct brain aging patterns using large-scale longitudinal neuroimaging data in mid-to-late adulthood. Compared to brain aging pattern 1, brain aging pattern 2 were characterized by a faster rate of GMV decrease, accelerated biological aging, cognitive decline, and genetic susceptibility to neurodevelopmental disorders. By integrating longitudinal neuroimaging data from adult and adolescent cohorts, we demonstrated the ‘last in, first out’ mirroring patterns between structural brain aging and brain development, and showed that the mirroring pattern was manifested in the temporal lobe and among participants with accelerated brain aging. Further, genome-wide association studies identified significant genetic loci contributing to accelerated brain aging, while spatial correlation between whole-brain transcriptomic profiles and structural brain aging / development revealed important gene sets associated with both accelerated brain aging and delayed brain development.

Brain aging is closely related to the onset and progression of neurodegenerative and neuropsychiatric disorders. Both neurodegenerative and neuropsychiatric disorders demonstrate strong inter-individual heterogeneity, which prevents the comprehensive understanding of their neuropathology and neurogenetic basis. Therefore, multidimensional investigation into disease subtyping and population clustering of structural brain aging are crucial in elucidating the sources of heterogeneity and neurophysiological basis related to the disease spectrum (Habes et al., 2020). In the last decades, major developments in the subtyping of Alzheimer’s disease, dementia and Parkinson’s disease, have provided new perspectives regarding their clinical diagnosis, treatment, disease progression and prognostics (Habes et al., 2020; Berg et al., 2021; Ferreira et al., 2020). While previous studies of brain aging mostly focused on the cross-sectional differences between cases and healthy controls, we here delineated the structural brain aging patterns among healthy participants using a novel data-driven approach that captured both cross-sectional and longitudinal trajectories of the whole-brain gray matter volume (Feczko et al., 2019; Poulakis et al., 2022). The two brain aging patterns identified using the above approach showed large differences in the rate of change in medial occipitotemporal gyrus, which is involved in vision, word processing, and scene recognition (Bogousslavsky et al., 1987; Epstein et al., 1999; Mechelli et al., 2000). Significant reduction of the gray matter volume and abnormal changes of the functional connectivity in this region were found in subjects with mild cognitive impairment (MCI) and AD, respectively (Chételat et al., 2005; Yao et al., 2010). Previous research on brainAGE (Elliott et al., 2021; Christman et al., 2020) (the difference between chronological age and the age predicted by the machine learning model of brain imaging data) showed that as a biomarker of accelerated brain aging, people with older brainAGE have accelerated biological aging and early signs of cognitive decline, which is consistent with our discoveries in this study. Our results support the establishment of a network connecting brain aging patterns with biological aging profiles involving multi-organ systems throughout the body (Tian et al., 2023). Since structural brain patterns might manifest and diverge decades before cognitive decline (Aljondi et al., 2019), subtyping of brain aging patterns could aid in the early prediction of cognitive decline and severe neurodegenerative and neuropsychiatric disorders.

Mirroring pattern between brain development and brain aging has long been hypothesized by postulating that phylogenetically newer and ontogenetically less precocious brain structures degenerate relatively early (Douaud et al., 2014). Early studies have reported a positive correlation between age-related differences of cortical volumes and precedence of myelination of intracortical fibers (Raz, 2000). Large differences in the patterns of change between adolescent late development and aging in the medial temporal cortex were previously found in studies of brain development and aging patterns (Tamnes et al., 2013). Here, we compared the annual volume change of the whole-brain gray matter during brain development and early / late stages of brain aging, and found that mirroring patterns are predominantly localized to the lateral / medial temporal cortex and the cingulate cortex. These cortical regions characterized by ‘last in, first out’ mirroring patterns showed increased vulnerability to the several neuropsychiatric disorders. For example, regional deficits in the superior temporal gyrus and medial temporal lobe were observed in schizophrenia (Honea et al., 2005), along with morphological abnormalities in the medial occipitotemporal gyrus (Schultz et al., 2010). Children diagnosed with ADHD had lower brain surface area in the frontal, cingulate, and temporal regions (Hoogman et al., 2019). Douaud et al., 2014 revealed a population transmodal network with lifespan trajectories characterized by the mirroring pattern of development and aging. We investigated the genetic susceptibility to individual-level mirroring patterns based on the lasting impact of neurodevelopmental genetic factors on brain (Fjell et al., 2015), demonstrating that those with more rapidly brain aging patterns have a higher risk of delayed development.

Identifying genes contributing to structural brain aging remains a critical step in understanding the molecular changes and biological mechanisms that govern age-related cognitive decline. Several genetic loci have been reported to be associated with brain aging modes and neurocognitive decline, many of which demonstrated global overlap with neuropsychiatric disorders and their related risk factors (Smith et al., 2020; Glahn et al., 2013; Brouwer et al., 2022). Here, we focused on the individual brain aging phenotype by estimating individual deviation from the population averaged total GMV and conducted genome-wide association analysis with this phenotype. Our approach identified six risk SNPs associated with accelerated brain aging, most of which could be further validated by previous studies using population averaged brain aging phenotypes. However, our approach revealed additional genetic signals and demonstrated genetic architecture underlying brain aging patterns overlap with bone density (Estrada et al., 2012; Zheng et al., 2015). In addition, molecular profiling of the aging brain has been thoroughly investigated among patients with neurogenerative diseases, but rarely conducted to shed light on the mirroring patterns among healthy participants. Analysis of the spatial correlation between gene expression profiles and structural brain development / aging further identified genes contributing to delayed brain development and accelerated brain aging. Specifically, expression of gene BDNF-AS was significantly associated with both processes. BDNF-AS is an antisense RNA gene and plays a role in the pathoetiology of non-neoplastic conditions mainly through the mediation of BDNF (Ghafouri-Fard et al., 2021). LGR4 (associated with delayed brain development) and FAM3C (associated with accelerated brain aging) identified in the spatial genetic association analysis also validated our findings in the GWAS.

There are several limitations in the current study that need to be addressed in future research. Firstly, the UK Biobank cohort, which we leveraged to identify population clustering of brain aging patterns, had a limited number of repeated structural MRI scans. Therefore, it remains challenging to obtain robust estimation of the longitudinal whole-brain GMV trajectory at the individual level. As a robustness check, we have calculated both intra-class correlation and variance of both random intercept and age slope to ensure appropriateness of the mixed effect models. Secondly, although aging is driven by numerous hallmarks, we have only investigated the association between brain aging patterns and biological aging in terms of telomere length and blood biochemical markers due to limitations of data access. Other dimensions of aging hallmarks and their relationship with structural brain aging need to be investigated in the future. Thirdly, our genomic analyses were restricted to ‘white British’ participants of European ancestry. The diversity of genomic analyses will continue to improve as the sample sizes of GWAS of non-European ancestry increase. Further, although the gene expression maps from Allen Human Brain Atlas enabled us to gain insights into the spatial coupling between gene expression profiles and mirroring patterns of the brain, the strong inter-individual variation of whole-brain gene expression levels and large temporal span of the human brain samples may lead to the inaccurate correspondence in the observed associations. Finally, we focused on structural MRIs in deriving brain aging patterns in this analysis, future investigations could consider other brain imaging modalities from a multi-dimensional perspective. Nevertheless, our study represents a novel attempt for population clustering of structural brain aging and validated the mirroring pattern hypothesis by leveraging large-scale adolescent and adult cohorts.

Methods

Participants

T1-weighted brain MRI images were obtained from 37,013 individuals aged 44–82 years old from UK Biobank (36,914 participants at baseline visit in 2014+, 4007 participants at the first follow-up visit in 2019+). All participants from UK Biobank provided written informed consent, and ethical approval was granted by the North West Multi-Center Ethics committee (https://www.ukbiobank.ac.uk/learn-more-about-uk-biobank/about-us/ethics) with research ethics committee (REC) approval number 16/NW/0274. Participants were excluded if they were diagnosed with severe psychiatric disorders or neurological diseases using ICD-10 primary and secondary diagnostic codes or from self-reported medical conditions at UK Biobank assessment center (see Appendix 1—tables 1 and 2). Data were obtained under application number 19542. A total of 1529 adolescents with structural MRI images were drawn from the longitudinal project IMAGEN (1463 at age 14, 1377 at age 19, and 1148 at age 23), of which the average number of MRI scans was 2.61 per adolescent. The lMAGEN study was approved by local ethics research committees of King’s College London, University of Nottingham. Trinity College Dublin, University of Heidelberg, Technische Universität Dresden, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, and University Medical Center at the University of Hamburg in compliance with the Declaration of Helsinki (Association, 2013). Informed consent was given by all participants and a parent/guardian of each participant.

MRI acquisition

Quality-controlled T1-weighted neuroimaging data from UK Biobank and IMAGEN were processed using FreeSurfer v6.0. Detailed imaging processing pipeline can be found online for UK Biobank (https://biobank.ctsu.ox.ac.uk/crystal/crystal/docs/brain_mri.pdf) and IMAGEN (https://github.com/imagen2/imagen_mri; Schumann et al., 2010; Imagen, 2020). Briefly, cortical gray matter volume (GMV) from 33 regions in each hemisphere were generated using Desikan–Killiany Atlas (Desikan et al., 2006), and total gray matter volume (TGMV), intracranial volume (ICV) and subcortical volume were derived from ASEG atlas (Fischl et al., 2002 See Appendix 1—table 3). Regional volume was averaged across left and right hemispheres. To avoid deficient segmentation or parcellation, participants with TGMV, ICV or regional GMV beyond 4 standard deviations from the sample mean were considered as outliers and removed from the following analyses.

Identification of longitudinal brain aging patterns

Whole-brain GMV trajectory was estimated for each participant in 40 brain regions of interest (ROIs) (33 cortical regions and 7 subcortical regions), using mixed effect regression model with fixed linear and quadratic age effects, random intercept and random age slope. Covariates include sex, assessment center, handedness, ethnic, and ICV. Models with random intercept and with both random intercept and random age slope were compared using AIC, BIC and evaluation of intra-class correlation (ICC). Results suggested that random age slope model should be chosen for almost all ROIs (Appendix 1—table 14). Deviation of regional GMV from the population average was calculated for each participant at age 60 years and dimensionality reduction was conducted via principal component analysis (PCA). The first 15 principal components explaining approximately 70% of the total variations of regional GMV deviation were used in multivariate k-means clustering. Optimal number of clusters was chosen using both elbow diagram and contour coefficient (Appendix 1—figure 6). Rates of volumetric change for total gray matter and each ROI were estimated using generalized additive mixed effect models (GAMM) with fixed cubic splines of age, random intercept and random age slope, which incorporates both cross-sectional between-subject variation and longitudinal within-subject variation from 40,921 observations and 37,013 participants. Covariates include sex, assessment center, handedness, ethnic, and ICV. We also applied PCA and locally linear embedding (LLE; Roweis and Saul, 2000) to the adjusted GMV ROIs in order to map the high-dimensional imaging-derived phenotypes to a low-dimensional space for stratification visualisation. The GMV of 40 ROIs at baseline were linearly adjusted for sex, assessment center, handedness, ethnic, ICV, and second-degree polynomial in age to be consistent with the whole-brain GMV trajectory model.

Association between brain aging patterns and biological aging, cognitive decline and genetic susceptibilities of neuropsychiatric disorders

Individuals with Z-standardized leucocyte telomere length (Codd et al., 2021) and blood biochemistry (which were used to calculate PhenoAge (Levine et al., 2018) that characterizes biological aging) outside 4 standard deviations from the sample mean were excluded for better quality control. A total of 11 cognitive tests performed on the touchscreen questionnaire were included in the analysis. More information about the cognitive tests is provided in Supplementary Information. Comparisons of biological aging (leucocyte telomere length, PhenoAge) and cognitive function were conducted among participants with different brain aging patterns using both unadjusted and adjusted multivariate regression models with Bonferroni / FDR correction. Polygenic Risk Scores (PRS) were calculated for autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), Alzheimer’s disease (AD), Parkinson’s Disease (PD), bipolar disorder (BIP), major depressive disorder (MDD), schizophrenia (SCZ), and delayed brain development using GWAS summary statistics (Shi et al., 2023) at multiple p value thresholds (from 0.005 to 0.5 at intervals of 0.005, and 1), with higher p value thresholds incorporating larger number of independent SNPs. After quality control of genotype and imaging data, PRSs were generated for 25,861 participants on UK Biobank genotyping data. SNPs were pruned and clumped with a cutoff r2 ≥ 0.1 within a 250 kb window. All calculations were conducted using PRSice v2.3.5 (Choi and O’Reilly, 2019). Enhanced PRS from UK Biobank Genomics for multiple diseases were also tested. Detailed instructions for calculating enhanced PRS in UK Biobank can be found in research of Thompson et al., 2022 Comparisons of neuropsychiatric disorders were conducted among participants with different brain aging patterns using t test with FDR correction. All statistical tests were two-sided.

Genome wide association Study to identify SNPs associated with brain aging patterns

We performed Genome-wide association studies (GWAS) on individual deviations of total GMV relative to the population average at 60 years using PLINK 2.0 (Chang et al., 2015). Variants with missing call rates exceeding 5%, minor allele frequency below 0.5% and imputation INFO score less than 0.8 were filtered out after the genotyping quality control for UK Biobank Imputation V3 dataset. Among the 337,138 unrelated ‘white British’ participants of European ancestry included in our study, 25,861 with recent UK ancestry and accepted genotyping and imaging quality control were included in the GWAS. The analyses were further adjusted for age, age2, sex, assessment center, handedness, ethnic, ICV, and the first 10 genetic principal components. Genome-wide significant SNPs (p<510–8) obtained from the GWAS were clumped by linkage disequilibrium (LD) (r2 <0.1 within a 250 kb window) using UKB release2b White British as the reference panel. We subsequently performed gene-based annotation in FUMA (Watanabe et al., 2017) using genome-wide significant SNPs and SNPs in close LD (r2 ≥ 0.1) using Annotate Variation (ANNOVAR) on Ensemble v102 genes (Wang et al., 2010).

Mirroring patterns between brain aging and brain development

To validate the ‘last in, first out’ mirroring hypothesis, we evaluated the structural association between brain development and brain aging. Longitudinal neuroimaging data from 1529 adolescents in the IMGAEN cohort and 3908 mid-to-late adulthood in the UK Biobank cohort were analyzed. Annual percentage volume change (APC) for each ROI was calculated among individuals with at least two structural MRI scans by subtracting the baseline GMV from follow-up GMV and dividing by the number of years between baseline and follow-up visits. Region-specific APC was regressed on age using smoothing spline with cross validated degree of freedom. Estimated APC for each ROI was obtained at age 15y for adolescents and at age 55y (early aging) and 75y (late aging) for participants in UK Biobank. Region-specific APC during adolescence (or mid-to-late adulthood) was then standardized across all cortical regions to create the brain development (or aging) map. Finally, the brain development map and brain aging map were compared to assess the mirroring pattern for each ROI in the overall population and across different aging subgroups.

Gene expression analysis

The Allen Human Brain Atlas (AHBA) dataset (http://human.brain-map.org), which comprises gene expression measurements in six postmortem adults (age 24–57y) across 83 parcellated brain regions (Hawrylycz et al., 2012; Markello et al., 2021), were used to identify gene expressions significantly associated with structural brain development and aging. The expression profiles of 15,633 genes were averaged across donors to form a 8315,633 transcriptional matrix and partial least squares (PLS) regression was adopted for analyzing the association between regional change rate of gray matter volume and gene expression profiles. Specifically, estimated regional APC at 15 (obtained from IMAGEN cohort) and 55 years old (obtained from UK Biobank) were regressed on the high-dimensional gene expression profiles upon regularization. Associations between the first PLS component and estimated APC during brain development and brain aging were tested by spatial permutation analysis (10,000 times; Váša et al., 2018). Additionally, gene expression profiles of genes mapped to GWAS significant SNP were extracted from AHBA. The association between gene expression profiles of mapped genes and estimated APC during brain development and aging was also tested by spatial permutation analysis. Statistical significance of each gene’s contribution to the first PLS component was tested with standard error calculated using bootstrap (Li et al., 2021; Morgan et al., 2019; Romero-Garcia et al., 2020), and genes significantly associated with delayed brain development and accelerated brain aging were selected. Enrichment of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and gene ontology (GO) of biological processes for these selected genes were analyzed using R package clusterProfiler (Yu et al., 2012). All statistical significances were corrected for multiple testing using FDR.

Code availability

R version 4.2.0 was used to perform statistical analyses. FreeSurfer version 6.0 was used to process neuroimaging data. lme4 1.1 in R version 4.2.0 was used to perform longitudinal data analyses. PRSice version 2.3.5 (https://choishingwan.github.io/PRSice/; Choi and O’Reilly, 2019) was used to calculate the PRS. PLINK 2.0 (https://www.cog-genomics.org/plink/2.0/) and FUMA version 1.5.6 (https://fuma.ctglab.nl/) were used to perform genome-wide association analysis, and ANNOVAR was used to perform gene-based annotation. AHBA microarray expression data were processed using abagen toolbox version 0.1.3 (https://doi.org/10.5281/zenodo.5129257). The rotate_parcellation code used to perform a spatial permutation test of a parcellated cortical map: https://github.com/frantisekvasa/rotate_parcellation (Váša, 2023; Váša et al., 2018). Code for PLS analysis and bootstrapping to estimate PLS weights are available at https://github.com/KirstieJane/NSPN_WhitakerVertes_PNAS2016/tree/master/SCRIPTS (Whitaker, 2016; Whitaker et al., 2016). clusterProfiler 4.6 in R version 4.2.0 was used to analyze gene-set enrichment.

Appendix 1

Population clustering of structural brain aging and its association with brain development.

Supplementary method

Cognitive assessment

Reaction time

This cognitive function test is based on 12 rounds of the card-game 'Snap'. The participant is shown two cards at a time; if both cards are the same, they press a button-box that is on the table in front of them as quickly as possible. The score used for analysis is mean time to correctly identify matches (UK Biobank data field 20023), which is the mean duration to first press of snap-button summed over rounds in which both cards matched.

Numeric memory

The participant was shown a 2-digit number to remember. The number then disappeared and after a short while they were asked to enter the number onto the screen. The number became one digit longer each time they remembered correctly (up to a maximum of 12 digits). This test is available for a subset of participants. The score used for analysis is maximum digits remembered correctly (UK Biobank data field 4282), which is longest number correctly recalled during the numeric memory test.

Fluid intelligence / reasoning

'Fluid intelligence' is defined as the capacity to solve problems that require logic and reasoning ability, independent of acquired knowledge. The participant has 2 min to complete as many questions as possible from the test. This test was incorporated into the touchscreen towards the end of recruitment. The score used for analysis is fluid intelligence score (UK Biobank data field 20016), which is a simple unweighted sum of the number of correct answers given to the 13 fluid intelligence questions. Participants who did not answer all of the questions within the allotted 2 min limit are scored as zero for each of the unattempted questions.

Trail making

The participant was presented with sets of digits/letters in circles scattered around the screen and asked to click on them sequentially according to a specific algorithm. The scores used for analysis are duration to complete numeric path (trail #1) (UK Biobank data field 6348) and duration to complete alphanumeric path (trail #2) (UK Biobank data field 6350).

Matrix pattern completion

The participant was presented with a series of matrix pattern blocks with an element missing and asked to select the element that best completed the pattern from a range of displayed choices. The score used for analysis is number of puzzles correctly solved (UK Biobank data field 6373).

Symbol digit substitution

The participant was presented with one grid linking symbols to single-digit integers and a second grid containing only the symbols. They were then asked to indicate the numbers attached to each of the symbols in the second grid using the first one as a key. The score used for analysis is number of symbol digit matches made correctly (UK Biobank data field 23324).

Tower rearranging

The participant was presented with an illustration of three pegs (towers) on which three differently-colored hoops had been placed. The were then asked to indicate how many moves it would take to re-arrange the hoops into another specific position. The score used for analysis is number of puzzles correct (UK Biobank data field 21004).

Paired associate learning

In the paired associate learning test the participants were shown 12 pairs of words (for 30 s in total) then, after an interval (in which they did a different test), presented with the first word of 10 of these pairs and asked to select the matching second word from a choice of four alternatives. The words were presented in the order: huge, happy, tattered, old, long, red, sulking, pretty, tiny and new. The score used for analysis is number of word pairs correctly associated (UK Biobank data field 21097), which is the number of word pairs correctly associated out of 10 attempts.

Prospective memory

Early in the touchscreen cognitive section, the participant is shown the message "At the end of the games we will show you four colored shapes and ask you to touch the Blue Square. However, to test your memory, we want you to actually touch the Orange Circle instead." The score used for analysis is prospective memory result (UK Biobank data field 20018), which condenses the results of the prospective memory test into 3 groups (“0”: instruction not recalled, either skipped or incorrect, “1”: correct recall on first attempt, “2”: correct recall on second attempt). We divided the test results into two groups for simplicity of analysis: “0” indicating no correct recall, and the combination of “1” and “2” indicating correct recall.

Pairs matching

Participants are asked to memorize the position of as many matching pairs of cards as possible. The cards are then turned face down on the screen and the participant is asked to touch as many pairs as possible in the fewest tries. Multiple rounds were conducted. The first round used 3 pairs of cards and the second 6 pairs of cards. In the pilot phase an additional (i.e. third) round was conducted using six pairs of cards. However this was dropped from the main study as the extra set of results were very similar to the second and not felt to add significant new information. The score used for analysis is number of incorrect matches in round 2 (UK Biobank data field 399.2).

Appendix 1—figure 1
The sample selection workflow.
Appendix 1—figure 2
Estimated rates of change in regional volumes for 33 bilateral brain regions.
Appendix 1—figure 3
Stratification of the identified brain aging patterns using linear and non-linear dimensionality reduction methods.
Appendix 1—figure 4
Effect size for comparing each individual blood biochemical metric (used to calculate the PhenoAge) between participants with brain aging patterns 1 and 2.
Appendix 1—figure 5
Gene set enrichment of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and gene ontology (GO) of biological processes.
Appendix 1—figure 6
Optimal number of clusters was chosen using elbow method (a) and silhouette method (b).
Appendix 1—table 1
ICD-10 primary and secondary diagnostic codes for exclusion criteria.
ConditionCode
Malignant neoplasmC70, C71
DementiaF00, F01, F02, F03, F04
Mental and behavioural disorders due to psychoactive substance useF10-F19
Schizophrenia, schizotypal and delusional disordersF20-F29
Mood [affective] disordersF30-F39
Mental retardationF70-F79
Disorders of psychological developmentF80-F89
Hyperkinetic disordersF90
Inflammatory diseases of the central nervous systemG00-G09
Systemic atrophies primarily affecting the central nervous systemG10, G11, G122, G13
Extrapyramidal and movement disordersG20, G21, G22, G23
Other degenerative diseases of the nervous systemG30-G32
Demyelinating diseases of the central nervous systemG35-G37
Episodic and paroxysmal disordersG40, G41, G45, G46
Infantile cerebral palsyG80
Cerebrovascular diseasesI60-I69
Down’s syndromeQ90
Intracranial injuryS06
Appendix 1—table 2
Self-reported illness codes for exclusion criteria.
Field IDConditionCode
20001Brain cancer/primary malignant brain tumour1032
Meningeal cancer/malignant meningioma1031
20002Benign neuroma1683
Brain abscess/intracranial abscess1245
Brain haemorrhage1491
Cerebral aneurysm1425
Cerebral palsy1433
Chronic/degenerative neurological problem1258
dementia/alzheimers/cognitive impairment1263
Encephalitis1246
Epilepsy1264
Fracture skull/head1626
Head injury1266
Ischaemic stroke1583
Meningioma/benign meningeal tumour1659
Meningitis1247
Motor Neurone Disease1259
Multiple Sclerosis1261
Nervous system infection1244
Neurological injury/trauma1240
Other demyelinating disease (not Multiple Sclerosis)1397
Other neurological problem1434
Parkinson’s Disease1262
Spina Bifida1524
Stroke1081
Subarachnoid haemorrhage1086
Subdural haemorrhage/haematoma1083
Transient ischaemic attack1082
Appendix 1—table 3
Cortical and subcortical brain regions.
Desikan–Killiany Atlas
bankssts
caudal anterior cingulate
caudal middle frontal
cuneus
entorhinal
fusiform
inferior parietal
inferior temporal
isthmus cingulate
lateral occipital
lateral orbitofrontal
lingual
medial orbitofrontal
middle temporal
parahippocampal
paracentral
pars opercularis
pars orbitalis
pars triangularis
pericalcarine
postcentral
posterior cingulate
precentral
precuneus
rostral anterior cingulate
rostral middle frontal
superior frontal
superior parietal
superior temporal
supramarginal
frontal pole
transverse temporal
insula
ASEG Atlas
thalamus proper
caudate
putamen
pallidum
hippocampus
amygdala
accumbens area
Appendix 1—table 4
Loadings matrix for the first 15 principal components.
PC1PC2PC3PC4PC5PC6PC7PC8PC9PC10PC11PC12PC13PC14PC15
Cortical
bankssts0.12–0.250.010.30–0.130.010.090.340.040.05–0.100.16–0.09–0.200.02
caudal.anterior.cingulate0.12–0.06–0.020.130.390.280.13–0.09–0.370.240.050.03–0.04–0.130.07
caudal.middle.frontal0.150.01–0.10–0.110.34–0.210.100.060.03–0.28–0.23–0.200.240.000.25
cuneus0.130.47–0.010.06–0.010.000.070.120.010.070.12–0.05–0.090.08–0.07
entorhinal0.090.070.130.080.100.17–0.53–0.180.060.04–0.19–0.100.130.01–0.04
fusiform0.18–0.030.010.13–0.020.02–0.20–0.020.150.30–0.01–0.28–0.230.080.21
inferior.parietal0.15–0.180.010.39–0.090.010.14–0.040.160.05–0.20–0.02–0.15–0.020.02
inferior.temporal0.16–0.130.010.26–0.010.15–0.04–0.100.14–0.08–0.08–0.250.200.29–0.01
isthmus.cingulate0.150.230.020.11–0.14–0.030.08–0.03–0.23–0.07–0.220.160.430.040.11
lateral.occipital0.160.300.000.12–0.020.020.050.100.160.180.06–0.23–0.230.120.17
lateral.orbitofrontal0.22–0.04–0.08–0.17–0.050.15–0.02–0.150.09–0.02–0.010.40–0.010.220.17
lingual0.120.430.030.14–0.01–0.020.070.11–0.01–0.03–0.100.160.02–0.09–0.08
medial.orbitofrontal0.21–0.06–0.06–0.06–0.050.10–0.01–0.130.18–0.060.130.170.050.01–0.25
middle.temporal0.17–0.17–0.010.27–0.140.100.210.250.09–0.15–0.090.040.070.05–0.11
parahippocampal0.120.090.130.050.060.14–0.460.02–0.020.09–0.220.090.03–0.320.02
paracentral0.19–0.05–0.10–0.070.18–0.23–0.11–0.060.010.03–0.020.06–0.35–0.01–0.35
pars.opercularis0.16–0.02–0.06–0.23–0.190.100.140.01–0.16–0.13–0.43–0.29–0.16–0.060.00
pars.orbitalis0.180.02–0.05–0.17–0.110.240.03–0.170.230.080.010.41–0.140.050.24
pars.triangularis0.160.02–0.03–0.35–0.260.230.13–0.05–0.06–0.06–0.25–0.17–0.21–0.08–0.05
pericalcarine0.100.47–0.010.08–0.010.010.090.13–0.02–0.010.020.08–0.030.03–0.17
postcentral0.20–0.04–0.110.060.16–0.29–0.090.120.080.03–0.090.100.040.010.04
posterior.cingulate0.18–0.08–0.040.020.240.090.08–0.02–0.300.01–0.070.10–0.05–0.12–0.31
precentral0.21–0.01–0.10–0.080.28–0.29–0.090.080.15–0.05–0.150.13–0.060.050.12
precuneus0.200.02–0.020.13–0.24–0.260.01–0.35–0.20–0.040.06–0.040.000.030.07
rostral.anterior.cingulate0.15–0.07–0.070.050.310.210.15–0.05–0.300.130.11–0.060.000.180.15
rostral.middle.frontal0.200.02–0.05–0.050.100.140.22–0.110.17–0.110.24–0.140.19–0.220.14
superior.frontal0.22–0.03–0.09–0.210.14–0.13–0.010.060.20–0.180.06–0.110.020.00–0.16
superior.parietal0.170.02–0.050.14–0.18–0.290.01–0.45–0.160.000.14–0.08–0.08–0.060.04
superior.temporal0.21–0.13–0.02–0.06–0.100.01–0.070.40–0.070.130.24–0.050.02–0.060.08
supramarginal0.17–0.15–0.080.06–0.24–0.23–0.130.03–0.210.040.110.100.14–0.08–0.06
frontal.pole0.160.03–0.100.010.000.160.03–0.130.280.050.28–0.160.26–0.38–0.22
transverse.temporal0.15–0.01–0.10–0.27–0.17–0.10–0.160.25–0.220.190.24–0.100.090.040.16
insula0.19–0.080.01–0.22–0.120.10–0.070.11–0.060.15–0.080.000.190.16–0.23
Subcortical
thalamus.proper0.09–0.020.31–0.080.03–0.080.080.000.02–0.210.110.10–0.19–0.370.33
caudate0.06–0.030.32–0.120.02–0.100.15–0.020.110.31–0.110.090.270.140.12
putamen0.08–0.030.42–0.120.03–0.130.13–0.030.050.22–0.05–0.140.07–0.02–0.17
pallidum0.03–0.030.42–0.070.00–0.130.16–0.060.040.20–0.070.000.04–0.20–0.06
hippocampus0.120.010.330.04–0.020.11–0.200.08–0.16–0.390.090.03–0.11–0.030.05
amygdala0.13–0.030.310.05–0.010.12–0.130.09–0.07–0.350.25–0.09–0.020.24–0.01
accumbens.area0.11–0.050.31–0.020.12–0.070.11–0.040.00–0.030.100.09–0.140.34–0.18
Appendix 1—table 5
Baseline and demographic characteristics for participants in the total population and stratified by brain aging patterns.
Total (n=37,013)Pattern 1 (n=18,929)Pattern 2 (n=18,084)
Age (years), mean (SD)63.9 (7.63)63.9 (7.64)63.8 (7.63)
Female, n (%)19,958 (53.9)10,117 (53.4)9,841 (54.4)
Ethnicity, n (%)
White34,219 (92.5)17,509 (92.5)16,710 (92.4)
Mixed1,137 (3.1)573 (3.0)564 (3.1)
Asian or Asian British1,210 (3.3)612 (3.2)598 (3.3)
Other447 (1.2)235 (1.2)212 (1.2)
Smoking status, n (%)*
Never smoker23,633 (64.4)12,269 (65.4)11,364 (63.4)
Previous smoker12,213 (33.3)6,085 (32.4)6,128 (34.2)
Current smoker833 (2.3)414 (2.2)419 (2.3)
TDI, mean (SD)–1.94 (2.69)–1.97 (2.66)–1.90 (2.71)
BMI (kg/m2), mean (SD)26.4 (4.32)26.3 (4.17)26.5 (4.46)
Years of Schooling, mean (SD)§16.8 (4.32)16.9 (4.29)16.8 (4,35)
  1. TDI = Townsend Deprivation Index, BMI = Body Mass Index.

  2. *

    Missing 334

  3. Missing 36

  4. Missing 1937

  5. §

    Missing 337

Appendix 1—table 6
Associations between results of biological aging biomarkers and subgroups stratified by whole-brain TGMV trajectories.

Cohen’s d measures the standardized difference of means between brain aging pattern 2 and brain aging pattern 1.

Biological aging biomarkersBrain aging pattern 1Brain aging pattern 2
NMean(SD)NMean(SD)
LTL17,6910.083 (0.98)16,8760.055 (0.97)
PhenoAge15,22841.35 (8.17)14,32341.58 (8.32)
Biological aging biomarkersUnadjustedAdjusted
Cohen’s d (95% CI)pP.BonferroniCohen’s d (95% CI)pP.Bonferroni
LTL–0.028 (-0.049,–0.007)0.0090–0.030 (-0.051,–0.009)0.0060.011
PhenoAge0.027 (0.004, 0.050)0.01900.092 (0.070, 0.116)3.05E-156.11E-15
Appendix 1—table 7
Associations between results of cognitive function tests and subgroups stratified by whole-brain TGMV trajectories.

Cohen’s d measures the standardized difference of means between brain aging pattern 2 and brain aging pattern 1.

Cognitive functionsBrain aging pattern 1Brain aging pattern 2
NMean(SD)NMean(SD)
Reaction time17,749–594.70 (108.15)16,831–594.68 (110.50)
Numeric memory13,3506.82 (1.26)12,3466.72 (1.27)
Fluid intelligence17,5806.73 (2.06)16,6126.53 (2.04)
Trail making A13,052–224.50 (84.82)12,036–227.56 (84.85)
Trail making B12,743–563.06 (246.74)11,753–576.55 (260.75)
Matrix pattern completion13,0648.07 (2.11)12,0647.91 (2.14)
Symbol digit substitution13,07719.08 (5.15)12,05618.87 (5.35)
Tower rearranging12,9529.96 (3.20)11,9589.83 (3.23)
Paired associate learning13,1847.01 (2.59)12,1986.88 (2.65)
Prospective memory17,831N/A16,949N/A
Pairs matching17,840–3.58 (2.90)16,956–3.67 (2.94)
Cognitive functionsUnadjustedAdjusted
Cohen’s d (95% CI)PP.FDRCohen’s d (95% CI)PP.FDR
Reaction time0.000 (-0.021, 0.021)0.990.990.006 (-0.016, 0.028)0.610.61
Numeric memory–0.082 (-0.106,–0.057)5.97E-113.28E-10–0.080 (-0.106,–0.055)8.99E-104.95E-09
Fluid intelligence–0.102 (-0.123,–0.080)5.94E-216.54E-20–0.99 (-0.121,–0.077)3.30E-183.63E-17
Trail making A–0.036 (-0.061,–0.011)0.0040.006–0.050 (-0.074,–0.024)1.46E-042.29E-04
Trail making B–0.053 (-0.078,–0.028)3.16E-058.68E-05–0.067 (-0.093,–0.041)6.16E-071.69E-06
Matrix pattern completion–0.076 (-0.101,–0.051)1.84E-096.74E-09–0.078 (-0.104,–0.052)3.67E-091.35E-08
Symbol digit substitution–0.040 (-0.065,–0.015)0.0020.002–0.053 (-0.079,–0.027)6.15E-051.13E-04
Tower rearranging–0.041 (-0.066,–0.016)0.0010.002–0.049 (-0.075,–0.023)2.18E-043.00E-04
Paired associate learning–0.051 (-0.076,–0.027)4.64E-051.02E-04–0.054 (-0.079,–0.028)4.89E-051.08E-04
Prospective memoryOR: 0.943 (0.891, 0.999)0.0470.052OR: 0.940 (0.883, 1.000)0.0520.057
Pairs matching–0.029 (-0.050,–0.008)0.0060.008–0.033 (-0.055,–0.011)0.0030.004
Appendix 1—table 8
Genome-wide association study details.

Loci associated with risk were thresholded at p<5×10–8, then distance-based clumping was used to define independently significant loci.

StudyNumber casesNumber controlsNumber of genome-wide independently significant lociDownload link
Attention deficit hyperactivity disorder Demontis et al., 201920,18335,19112https://figshare.com/ndownloader/files/28169253
Autism spectrum disorder Grove et al., 201918,38127,9695https://figshare.com/ndownloader/files/28169292
Alzheimer’s disease Jansen et al., 201971,880383,37825https://ctg.cncr.nl/software/summary_statistics
Parkinson’s disease Nalls et al., 201937,688, and 18,618 (proxy-cases)1,417,79190https://drive.google.com/file/d/1FZ9UL99LAqyWnyNBxxlx6qOUlfAnublN/view?usp=sharing
Bipolar disorder Mullins et al., 202141,917371,54964https://figshare.com/ndownloader/files/40036705
Major depressive disorder Wray et al., 2018135,458344,90144https://figshare.com/ndownloader/files/39504667
Schizophrenia Trubetskoy et al., 202276,755243,649287https://figshare.com/ndownloader/files/34517828
Delayed Brain Development Shi et al., 20237662 (proxy phenotype, continuous)1https://delayedneurodevelopment.page.link/amTC
Appendix 1—table 9
Polygenic Risk Scores comparisons between two subgroups.

Data supporting these scores were obtained either entirely from external GWAS data (the Standard PRS set). The bold P values reflect significance after FDR correction.

Traitn1n2statisticpp.adjust
AAM18,42917,586–2.2180.0270.080
AMD18,42917,5861.7530.0800.169
AD18,42917,5860.7350.4620.616
AST18,42917,586–0.8610.3890.543
AF18,42917,586–0.1000.9200.945
BD18,42917,5863.5573.75E-040.002
BMI18,42917,586–3.3090.0010.005
CRC18,42917,586–0.5440.5860.703
BC18,42917,586–3.1400.0020.008
CVD18,42917,586–2.1040.0350.091
CED18,42917,5861.0460.2960.484
CAD18,42917,586–1.5880.1120.202
CD18,42917,586–0.0940.9250.945
EOC18,42917,586–2.1830.0290.080
EBMDT18,42917,586–11.343<1.00E-20<1.00E-20
HBA1C_DF18,42917,586–2.9480.0030.013
HEIGHT18,42917,5866.6582.81E-113.37E-10
HDL18,42917,5860.8840.3770.543
HT18,42917,586–3.5394.02E-040.002
IOP18,42917,586–1.6050.1090.202
ISS18,42917,586–2.3830.0170.056
LDL_SF18,42917,586–0.6860.4920.627
MEL18,42917,5862.0250.0430.103
MS18,42917,586–0.0690.9450.945
OP18,42917,58612.029<1.00E-20<1.00E-20
PD18,42917,5861.4560.1450.249
POAG18,42917,586–0.8560.3920.543
PC18,42917,586–0.2400.8100.941
PSO18,42917,586–1.7810.0750.169
RA18,42917,586–2.4370.0150.053
SCZ18,42917,5860.1580.8740.945
SLE18,42917,5861.6950.0900.180
T1D18,42917,5860.6660.5050.627
T2D18,42917,586–5.5233.35E-083.02E-07
UC18,42917,5860.8830.3770.543
VTE18,42917,586–0.1700.8650.945
Appendix 1—table 10
Polygenic Risk Scores comparisons between two subgroups.

Data supporting these scores were obtained external and internal UK Biobank data (the Enhanced PRS set). The bold p values reflect significance after FDR correction.

Traitn1n2statisticpp.adjust
AAM3,4073,409–1.7080.0880.344
AMD3,4073,4090.5470.5840.931
AD3,4073,4090.7560.4500.820
APOEA3,4073,4090.0230.9820.993
APOEB3,4073,4090.1190.9050.968
AST3,4073,4090.1120.9110.968
AF3,4073,4091.3060.1920.600
BD3,4073,4090.5610.5750.931
BMI3,4073,409–0.9760.3290.730
CRC3,4073,4090.9840.3250.730
BC3,4073,409–0.9950.3200.730
CAL3,4073,409–1.7860.0740.326
CVD3,4073,409–0.0090.9930.993
CED3,4073,4091.2800.2000.600
CAD3,4073,4090.2310.8180.961
DOA3,4073,409–0.3260.7450.961
EOC3,4073,409–2.1670.0300.155
EBMDT3,4073,409–6.1111.04E-092.65E-08
EGCR3,4073,4090.4130.6800.961
EGCY3,4073,409–0.2100.8340.961
HBA1C_DF3,4073,4090.1300.8960.968
HEIGHT3,4073,4094.3511.38E-052.35E-04
HDL3,4073,4090.2940.7690.961
HT3,4073,409–0.8840.3770.743
IOP3,4073,409–2.3660.0180.151
ISS3,4073,4090.0660.9470.986
LDL_SF3,4073,409–0.1930.8470.961
MEL3,4073,4092.6590.0080.080
MS3,4073,409–2.2930.0220.151
OTFA3,4073,4090.3180.7500.961
OSFA3,4073,4090.7700.4410.820
OP3,4073,4096.4849.54E-114.87E-09
PD3,4073,4091.0410.2980.730
PDCL3,4073,4090.6630.5070.862
PHG3,4073,4090.3920.6950.961
PFA3,4073,4090.4950.6210.932
POAG3,4073,409–1.0830.2790.730
PC3,4073,4090.6750.5000.862
PSO3,4073,409–2.2340.0260.151
RMNC3,4073,4090.5010.6170.932
RHR3,4073,409–2.8650.0040.053
RA3,4073,4090.2970.7660.961
SCZ3,4073,409–0.8800.3790.743
SGM3,4073,409–0.2240.8230.961
SLE3,4073,4091.4580.1450.493
TCH3,4073,4090.1910.8480.961
TFA3,4073,4090.8920.3720.743
TTG3,4073,4091.2120.2260.640
T1D3,4073,4091.7710.0770.326
T2D3,4073,409–2.2180.0270.151
VTE3,4073,409–1.6130.1070.390
Appendix 1—table 11
Most significant single-variant associations (p < 510-8) detected in the GWAS analyses.

Six independent SNPs at genome-wide significance level were identified by linkage disequilibrium (LD) clumping (r2 < 0.1 within a 250 kb window). The location (chromosome [chr] and base position [bp]), alleles (A1 = effect allele and A2 = other allele), effect (β) and its standard error (β SE) with respect to A1, and association p-values from regression model of the variants are given, along with functional consequences of SNPs on gene by performing ANNOVAR.

SNPA1A2p-valueββ SELocation (chr:bp)Gene symbolPosition relative to gene
rs10835187CT1.70e-14–0.025580.00333311:27505677LGR4, LIN7Cintergenic
rs7776725CT4.47e-13–0.026400.0036447:121033121FAM3Cintronic
rs779233904AACA1.57e-090.020830.0034496:151910404CCDC170intronic
rs2504071TC3.34e-090.019590.0033116:152084862ESR1intronic
17:43553496:A:AATAAAT6.65e-09–0.024720.00426117:43553496PLEKHM1intronic
10:104227791:G:GAGAG1.48e-08–0.018890.00333410:104227791TMEM180intronic
Appendix 1—table 12
Association between gene expression profiles of mapped genes and estimated APC during brain development.

The bold p values reflect significance after the spatial permutation test.

Gene SymbolSpearman's ρp valueP.permutation
ACTR1A0.0960.4400.328
ARHGAP270.0510.6840.445
ARL17B0.0470.7050.452
BDNF-AS0.2560.0380.038
CCDC1700.2680.0300.069
ESR10.0210.8700.483
FAM3C–0.0960.4440.356
KANSL1–0.2620.0340.073
KANSL1-AS10.0670.5940.313
LGR40.5581.78E-062.50E-04
LIN7C0.0360.7750.464
LRRC37A4P–0.2720.0270.148
MAPT0.0240.8460.405
PLEKHM1–0.2760.0250.109
SPPL2C0.1160.3510.189
STH–0.1470.2380.277
SUFU0.4070.0010.028
Appendix 1—table 13
Association between gene expression profiles of mapped genes and estimated APC during brain aging.

The bold p values reflect significance after the spatial permutation test.

Gene SymbolSpearman's ρPvalueP.permutation
ACTR1A–0.2350.0580.052
ARHGAP270.4864.48E-055.50E-04
ARL17B0.0900.4730.240
BDNF-AS0.4903.68E-051.50E-04
CCDC1700.2060.0980.075
ESR10.5326.02E-061.50E-04
FAM3C–0.3660.0030.005
KANSL10.2130.0860.078
KANSL1-AS1–0.2620.0340.059
LGR40.0700.5760.348
LIN7C0.1770.1540.120
LRRC37A4P0.1430.2500.165
MAPT–0.2870.0200.022
PLEKHM10.2020.1040.080
SPPL2C0.2110.0890.059
STH–0.0010.9970.490
SUFU–0.0360.7730.373
Appendix 1—table 14
Model evaluation results using relative measures: AIC, BIC, likelihood ratio test and intra-class correlation (ICC).
modelAICBIClrtestICC_adjustedICC_unadjusted
lmer_intr_thalamus.proper79444.2479591.296.45E-150.88750.3958
lmer_slope_thalamus.proper79382.8979547.246.45E-150.88890.3986
lmer_intr_caudate95845.4895992.542.68E-710.95430.6824
lmer_slope_caudate95524.4995688.842.68E-710.95640.6817
lmer_intr_putamen92106.0692253.122.40E-420.93980.5946
lmer_slope_putamen91918.492082.752.40E-420.94240.5933
lmer_intr_pallidum90500.390647.364.21E-420.88590.5116
lmer_slope_pallidum90313.7690478.124.21E-420.88620.5093
lmer_intr_hippocampus89833.9289980.971.37E-080.93090.5505
lmer_slope_hippocampus89801.7189966.061.37E-080.93290.5505
lmer_intr_amygdala89976.9890124.034.01E-060.86970.4898
lmer_slope_amygdala89956.1390120.484.01E-060.87130.4897
lmer_intr_accumbens.area96936.1997083.241.69E-140.82280.5295
lmer_slope_accumbens.area96876.7797041.121.69E-140.82330.5310
lmer_intr_bankssts97981.9698129.010.0018180.93390.6798
lmer_slope_bankssts97973.3498137.690.0018180.93540.6814
lmer_intr_caudal.anterior.cingulate109133.2109280.30.1315070.86380.7653
lmer_slope_caudal.anterior.cingulate109133.2109297.50.1315070.86440.7659
lmer_intr_caudal.middle.frontal93118.1393265.198.31E-060.92270.5929
lmer_slope_caudal.middle.frontal93098.7493263.098.31E-060.92460.5949
lmer_intr_cuneus101801.4101948.50.0144290.92420.7256
lmer_slope_cuneus101796.9101961.30.0144290.92450.7262
lmer_intr_entorhinal109404.9109551.91.63E-140.80130.6908
lmer_slope_entorhinal109345.4109509.71.63E-140.80370.6928
lmer_intr_fusiform87545.8887692.939.17E-050.91390.5062
lmer_slope_fusiform87531.2887695.639.17E-050.91630.5074
lmer_intr_inferior.parietal89044.4389191.485.89E-140.93740.5564
lmer_slope_inferior.parietal88987.5189151.865.89E-140.94190.5603
lmer_intr_inferior.temporal84066.4784213.525.73E-070.93840.4956
lmer_slope_inferior.temporal84041.7384206.085.73E-070.94050.4977
lmer_intr_isthmus.cingulate92442.1292589.170.1271910.92750.5862
lmer_slope_isthmus.cingulate9244292606.350.1271910.92840.5869
lmer_intr_lateral.occipital89550.489697.450.0039430.91210.5273
lmer_slope_lateral.occipital89543.3389707.680.0039430.91290.5282
lmer_intr_lateral.orbitofrontal86224.1386371.181.29E-080.84660.4323
lmer_slope_lateral.orbitofrontal86191.7986356.141.29E-080.84970.4345
lmer_intr_lingual102605.2102752.20.0412420.91810.7268
lmer_slope_lingual102602.8102767.20.0412420.91810.7272
lmer_intr_medial.orbitofrontal89954.3690101.412.25E-060.78320.4219
lmer_slope_medial.orbitofrontal89932.3590096.72.25E-060.78720.4241
lmer_intr_middle.temporal83331.0183478.060.00190.91770.4615
lmer_slope_middle.temporal83322.4883486.830.00190.91950.4629
lmer_intr_parahippocampal108997.1109144.24.99E-050.86860.7639
lmer_slope_parahippocampal108981.3109145.64.99E-050.86900.7639
lmer_intr_paracentral98058.6398205.680.0156860.86950.5958
lmer_slope_paracentral98054.3298218.670.0156860.87050.5960
lmer_intr_pars.opercularis96829.0596976.11.20E-120.93540.6658
lmer_slope_pars.opercularis96778.1596942.51.20E-120.93960.6698
lmer_intr_pars.orbitalis96989.6197136.671.39E-050.87850.5892
lmer_slope_pars.orbitalis96971.2597135.61.39E-050.88050.5912
lmer_intr_pars.triangularis96637.5896784.634.55E-180.94020.6710
lmer_slope_pars.triangularis96561.7296726.074.55E-180.94390.6748
lmer_intr_pericalcarine105115.91052630.0228410.94290.8190
lmer_slope_pericalcarine105112.4105276.70.0228410.94410.8200
lmer_intr_postcentral91605.691752.650.0005490.87640.5189
lmer_slope_postcentral91594.5991758.940.0005490.87890.5203
lmer_intr_posterior.cingulate96853.7397000.782.21E-190.89130.6014
lmer_slope_posterior.cingulate96771.8296936.172.21E-190.89490.6022
lmer_intr_precentral91186.5991333.642.61E-120.84780.4864
lmer_slope_precentral91137.2591301.62.61E-120.85040.4864
lmer_intr_precuneus84734.5884881.631.23E-080.90880.4708
lmer_slope_precuneus84702.1684866.511.23E-080.91260.4730
lmer_intr_rostral.anterior.cingulate95688.6895835.732.64E-120.90930.6097
lmer_slope_rostral.anterior.cingulate95639.3695803.722.64E-120.91220.6129
lmer_intr_rostral.middle.frontal80873.8481020.892.57E-170.91370.4354
lmer_slope_rostral.middle.frontal80801.4480965.792.57E-170.91910.4395
lmer_intr_superior.frontal79730.679877.651.25E-110.89210.4038
lmer_slope_superior.frontal79684.3879848.731.25E-110.89720.4060
lmer_intr_superior.parietal92895.95930438.55E-070.88990.5487
lmer_slope_superior.parietal92872.0193036.368.55E-070.89340.5512
lmer_intr_superior.temporal86495.2486642.290.0030210.91480.4959
lmer_slope_superior.temporal86487.6486651.990.0030210.91680.4971
lmer_intr_supramarginal87390.3987537.443.67E-130.92630.5221
lmer_slope_supramarginal87337.1287501.473.67E-130.93200.5258
lmer_intr_frontal.pole110426.3110573.41.95E-650.69070.5868
lmer_slope_frontal.pole110132.3110296.71.95E-650.69570.5882
lmer_intr_transverse.temporal102753.2102900.20.0324130.91300.7247
lmer_slope_transverse.temporal102750.3102914.70.0324130.91440.7258
lmer_intr_insula88693.288840.267.61E-140.82630.4416
lmer_slope_insula88636.7988801.147.61E-140.82900.4420

Data availability

The summary statistics of GWAS for individual deviations of total GMV is available at https://doi.org/10.5061/dryad.jh9w0vtmn. All the UK Biobank data used in the study are available at https://www.ukbiobank.ac.uk. The IMAGEN project data are available at https://imagen-project.org (Schumann et al., 2010). GWAS summary statistics used to calculate the PRS are available in the Appendix 1—table 8. Human gene expression data are available in the Allen Human Brain Atlas dataset: https://human.brainmap.org (Hawrylycz et al., 2012). Source data files have been provided for Figures 25, 7 and 8, which contain the numerical data used to generate the figures. Summary statistics of the GWAS for delayed brain development by Shi et al are available at https://delayedneurodevelopment.page.link/amTC.

The following data sets were generated
    1. Duan H
    2. Shi R
    3. Kang J
    (2024) Dryad
    The summary statistics of GWAS for individual deviations of total GMV.
    https://doi.org/10.5061/dryad.jh9w0vtmn
The following previously published data sets were used
    1. Nalls MA
    2. Blauwendraat C
    3. Vallerga CL
    (2019) GWAS Catalog
    ID GCST009325. Parkinson’s disease or first degree relation to individual with Parkinson’s disease.
    1. Jansen IE
    2. Savage JE
    3. Watanabe K
    (2019) Vrije Universiteit Research Drive
    ID l7aiRr1UEgdoJfZ. Summary statistics for Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer's disease risk.
    1. Adams MJ
    (2023) figshare
    MDD2 (MDD2018) GWAS sumstats w/o UKBB.
    https://doi.org/10.6084/m9.figshare.21655784

References

    1. Brouwer RM
    2. Klein M
    3. Grasby KL
    4. Schnack HG
    5. Jahanshad N
    6. Teeuw J
    7. Thomopoulos SI
    8. Sprooten E
    9. Franz CE
    10. Gogtay N
    11. Kremen WS
    12. Panizzon MS
    13. Olde Loohuis LM
    14. Whelan CD
    15. Aghajani M
    16. Alloza C
    17. Alnæs D
    18. Artiges E
    19. Ayesa-Arriola R
    20. Barker GJ
    21. Bastin ME
    22. Blok E
    23. Bøen E
    24. Breukelaar IA
    25. Bright JK
    26. Buimer EEL
    27. Bülow R
    28. Cannon DM
    29. Ciufolini S
    30. Crossley NA
    31. Damatac CG
    32. Dazzan P
    33. de Mol CL
    34. de Zwarte SMC
    35. Desrivières S
    36. Díaz-Caneja CM
    37. Doan NT
    38. Dohm K
    39. Fröhner JH
    40. Goltermann J
    41. Grigis A
    42. Grotegerd D
    43. Han LKM
    44. Harris MA
    45. Hartman CA
    46. Heany SJ
    47. Heindel W
    48. Heslenfeld DJ
    49. Hohmann S
    50. Ittermann B
    51. Jansen PR
    52. Janssen J
    53. Jia T
    54. Jiang J
    55. Jockwitz C
    56. Karali T
    57. Keeser D
    58. Koevoets M
    59. Lenroot RK
    60. Malchow B
    61. Mandl RCW
    62. Medel V
    63. Meinert S
    64. Morgan CA
    65. Mühleisen TW
    66. Nabulsi L
    67. Opel N
    68. de la Foz VOG
    69. Overs BJ
    70. Paillère Martinot ML
    71. Redlich R
    72. Marques TR
    73. Repple J
    74. Roberts G
    75. Roshchupkin GV
    76. Setiaman N
    77. Shumskaya E
    78. Stein F
    79. Sudre G
    80. Takahashi S
    81. Thalamuthu A
    82. Tordesillas-Gutiérrez D
    83. van der Lugt A
    84. van Haren NEM
    85. Wardlaw JM
    86. Wen W
    87. Westeneng HJ
    88. Wittfeld K
    89. Zhu AH
    90. Zugman A
    91. Armstrong NJ
    92. Bonfiglio G
    93. Bralten J
    94. Dalvie S
    95. Davies G
    96. Di Forti M
    97. Ding L
    98. Donohoe G
    99. Forstner AJ
    100. Gonzalez-Peñas J
    101. Guimaraes J
    102. Homuth G
    103. Hottenga JJ
    104. Knol MJ
    105. Kwok JBJ
    106. Le Hellard S
    107. Mather KA
    108. Milaneschi Y
    109. Morris DW
    110. Nöthen MM
    111. Papiol S
    112. Rietschel M
    113. Santoro ML
    114. Steen VM
    115. Stein JL
    116. Streit F
    117. Tankard RM
    118. Teumer A
    119. van ’t Ent D
    120. van der Meer D
    121. van Eijk KR
    122. Vassos E
    123. Vázquez-Bourgon J
    124. Witt SH
    125. IMAGEN Consortium
    126. Adams HHH
    127. Agartz I
    128. Ames D
    129. Amunts K
    130. Andreassen OA
    131. Arango C
    132. Banaschewski T
    133. Baune BT
    134. Belangero SI
    135. Bokde ALW
    136. Boomsma DI
    137. Bressan RA
    138. Brodaty H
    139. Buitelaar JK
    140. Cahn W
    141. Caspers S
    142. Cichon S
    143. Crespo-Facorro B
    144. Cox SR
    145. Dannlowski U
    146. Elvsåshagen T
    147. Espeseth T
    148. Falkai PG
    149. Fisher SE
    150. Flor H
    151. Fullerton JM
    152. Garavan H
    153. Gowland PA
    154. Grabe HJ
    155. Hahn T
    156. Heinz A
    157. Hillegers M
    158. Hoare J
    159. Hoekstra PJ
    160. Ikram MA
    161. Jackowski AP
    162. Jansen A
    163. Jönsson EG
    164. Kahn RS
    165. Kircher T
    166. Korgaonkar MS
    167. Krug A
    168. Lemaitre H
    169. Malt UF
    170. Martinot JL
    171. McDonald C
    172. Mitchell PB
    173. Muetzel RL
    174. Murray RM
    175. Nees F
    176. Nenadić I
    177. Oosterlaan J
    178. Ophoff RA
    179. Pan PM
    180. Penninx B
    181. Poustka L
    182. Sachdev PS
    183. Salum GA
    184. Schofield PR
    185. Schumann G
    186. Shaw P
    187. Sim K
    188. Smolka MN
    189. Stein DJ
    190. Trollor JN
    191. van den Berg LH
    192. Veldink JH
    193. Walter H
    194. Westlye LT
    195. Whelan R
    196. White T
    197. Wright MJ
    198. Medland SE
    199. Franke B
    200. Thompson PM
    201. Hulshoff Pol HE
    (2022) Genetic variants associated with longitudinal changes in brain structure across the lifespan
    Nature Neuroscience 25:421–432.
    https://doi.org/10.1038/s41593-022-01042-4
    1. Estrada K
    2. Styrkarsdottir U
    3. Evangelou E
    4. Hsu Y-H
    5. Duncan EL
    6. Ntzani EE
    7. Oei L
    8. Albagha OME
    9. Amin N
    10. Kemp JP
    11. Koller DL
    12. Li G
    13. Liu C-T
    14. Minster RL
    15. Moayyeri A
    16. Vandenput L
    17. Willner D
    18. Xiao S-M
    19. Yerges-Armstrong LM
    20. Zheng H-F
    21. Alonso N
    22. Eriksson J
    23. Kammerer CM
    24. Kaptoge SK
    25. Leo PJ
    26. Thorleifsson G
    27. Wilson SG
    28. Wilson JF
    29. Aalto V
    30. Alen M
    31. Aragaki AK
    32. Aspelund T
    33. Center JR
    34. Dailiana Z
    35. Duggan DJ
    36. Garcia M
    37. Garcia-Giralt N
    38. Giroux S
    39. Hallmans G
    40. Hocking LJ
    41. Husted LB
    42. Jameson KA
    43. Khusainova R
    44. Kim GS
    45. Kooperberg C
    46. Koromila T
    47. Kruk M
    48. Laaksonen M
    49. Lacroix AZ
    50. Lee SH
    51. Leung PC
    52. Lewis JR
    53. Masi L
    54. Mencej-Bedrac S
    55. Nguyen TV
    56. Nogues X
    57. Patel MS
    58. Prezelj J
    59. Rose LM
    60. Scollen S
    61. Siggeirsdottir K
    62. Smith AV
    63. Svensson O
    64. Trompet S
    65. Trummer O
    66. van Schoor NM
    67. Woo J
    68. Zhu K
    69. Balcells S
    70. Brandi ML
    71. Buckley BM
    72. Cheng S
    73. Christiansen C
    74. Cooper C
    75. Dedoussis G
    76. Ford I
    77. Frost M
    78. Goltzman D
    79. González-Macías J
    80. Kähönen M
    81. Karlsson M
    82. Khusnutdinova E
    83. Koh J-M
    84. Kollia P
    85. Langdahl BL
    86. Leslie WD
    87. Lips P
    88. Ljunggren Ö
    89. Lorenc RS
    90. Marc J
    91. Mellström D
    92. Obermayer-Pietsch B
    93. Olmos JM
    94. Pettersson-Kymmer U
    95. Reid DM
    96. Riancho JA
    97. Ridker PM
    98. Rousseau F
    99. Slagboom PE
    100. Tang NLS
    101. Urreizti R
    102. Van Hul W
    103. Viikari J
    104. Zarrabeitia MT
    105. Aulchenko YS
    106. Castano-Betancourt M
    107. Grundberg E
    108. Herrera L
    109. Ingvarsson T
    110. Johannsdottir H
    111. Kwan T
    112. Li R
    113. Luben R
    114. Medina-Gómez C
    115. Palsson ST
    116. Reppe S
    117. Rotter JI
    118. Sigurdsson G
    119. van Meurs JBJ
    120. Verlaan D
    121. Williams FMK
    122. Wood AR
    123. Zhou Y
    124. Gautvik KM
    125. Pastinen T
    126. Raychaudhuri S
    127. Cauley JA
    128. Chasman DI
    129. Clark GR
    130. Cummings SR
    131. Danoy P
    132. Dennison EM
    133. Eastell R
    134. Eisman JA
    135. Gudnason V
    136. Hofman A
    137. Jackson RD
    138. Jones G
    139. Jukema JW
    140. Khaw K-T
    141. Lehtimäki T
    142. Liu Y
    143. Lorentzon M
    144. McCloskey E
    145. Mitchell BD
    146. Nandakumar K
    147. Nicholson GC
    148. Oostra BA
    149. Peacock M
    150. Pols HAP
    151. Prince RL
    152. Raitakari O
    153. Reid IR
    154. Robbins J
    155. Sambrook PN
    156. Sham PC
    157. Shuldiner AR
    158. Tylavsky FA
    159. van Duijn CM
    160. Wareham NJ
    161. Cupples LA
    162. Econs MJ
    163. Evans DM
    164. Harris TB
    165. Kung AWC
    166. Psaty BM
    167. Reeve J
    168. Spector TD
    169. Streeten EA
    170. Zillikens MC
    171. Thorsteinsdottir U
    172. Ohlsson C
    173. Karasik D
    174. Richards JB
    175. Brown MA
    176. Stefansson K
    177. Uitterlinden AG
    178. Ralston SH
    179. Ioannidis JPA
    180. Kiel DP
    181. Rivadeneira F
    (2012) Genome-wide meta-analysis identifies 56 bone mineral density loci and reveals 14 loci associated with risk of fracture
    Nature Genetics 44:491–501.
    https://doi.org/10.1038/ng.2249
    1. Hoogman M
    2. Muetzel R
    3. Guimaraes JP
    4. Shumskaya E
    5. Mennes M
    6. Zwiers MP
    7. Jahanshad N
    8. Sudre G
    9. Wolfers T
    10. Earl EA
    11. Soliva Vila JC
    12. Vives-Gilabert Y
    13. Khadka S
    14. Novotny SE
    15. Hartman CA
    16. Heslenfeld DJ
    17. Schweren LJS
    18. Ambrosino S
    19. Oranje B
    20. de Zeeuw P
    21. Chaim-Avancini TM
    22. Rosa PGP
    23. Zanetti MV
    24. Malpas CB
    25. Kohls G
    26. von Polier GG
    27. Seitz J
    28. Biederman J
    29. Doyle AE
    30. Dale AM
    31. van Erp TGM
    32. Epstein JN
    33. Jernigan TL
    34. Baur-Streubel R
    35. Ziegler GC
    36. Zierhut KC
    37. Schrantee A
    38. Høvik MF
    39. Lundervold AJ
    40. Kelly C
    41. McCarthy H
    42. Skokauskas N
    43. O’Gorman Tuura RL
    44. Calvo A
    45. Lera-Miguel S
    46. Nicolau R
    47. Chantiluke KC
    48. Christakou A
    49. Vance A
    50. Cercignani M
    51. Gabel MC
    52. Asherson P
    53. Baumeister S
    54. Brandeis D
    55. Hohmann S
    56. Bramati IE
    57. Tovar-Moll F
    58. Fallgatter AJ
    59. Kardatzki B
    60. Schwarz L
    61. Anikin A
    62. Baranov A
    63. Gogberashvili T
    64. Kapilushniy D
    65. Solovieva A
    66. El Marroun H
    67. White T
    68. Karkashadze G
    69. Namazova-Baranova L
    70. Ethofer T
    71. Mattos P
    72. Banaschewski T
    73. Coghill D
    74. Plessen KJ
    75. Kuntsi J
    76. Mehta MA
    77. Paloyelis Y
    78. Harrison NA
    79. Bellgrove MA
    80. Silk TJ
    81. Cubillo AI
    82. Rubia K
    83. Lazaro L
    84. Brem S
    85. Walitza S
    86. Frodl T
    87. Zentis M
    88. Castellanos FX
    89. Yoncheva YN
    90. Haavik J
    91. Reneman L
    92. Conzelmann A
    93. Lesch K-P
    94. Pauli P
    95. Reif A
    96. Tamm L
    97. Konrad K
    98. Oberwelland Weiss E
    99. Busatto GF
    100. Louza MR
    101. Durston S
    102. Hoekstra PJ
    103. Oosterlaan J
    104. Stevens MC
    105. Ramos-Quiroga JA
    106. Vilarroya O
    107. Fair DA
    108. Nigg JT
    109. Thompson PM
    110. Buitelaar JK
    111. Faraone SV
    112. Shaw P
    113. Tiemeier H
    114. Bralten J
    115. Franke B
    (2019) Brain imaging of the cortex in adhd: a coordinated analysis of large-scale clinical and population-based samples
    The American Journal of Psychiatry 176:531–542.
    https://doi.org/10.1176/appi.ajp.2019.18091033
    1. Mullins N
    2. Forstner AJ
    3. O’Connell KS
    4. Coombes B
    5. Coleman JRI
    6. Qiao Z
    7. Als TD
    8. Bigdeli TB
    9. Børte S
    10. Bryois J
    11. Charney AW
    12. Drange OK
    13. Gandal MJ
    14. Hagenaars SP
    15. Ikeda M
    16. Kamitaki N
    17. Kim M
    18. Krebs K
    19. Panagiotaropoulou G
    20. Schilder BM
    21. Sloofman LG
    22. Steinberg S
    23. Trubetskoy V
    24. Winsvold BS
    25. Won H-H
    26. Abramova L
    27. Adorjan K
    28. Agerbo E
    29. Al Eissa M
    30. Albani D
    31. Alliey-Rodriguez N
    32. Anjorin A
    33. Antilla V
    34. Antoniou A
    35. Awasthi S
    36. Baek JH
    37. Bækvad-Hansen M
    38. Bass N
    39. Bauer M
    40. Beins EC
    41. Bergen SE
    42. Birner A
    43. Bøcker Pedersen C
    44. Bøen E
    45. Boks MP
    46. Bosch R
    47. Brum M
    48. Brumpton BM
    49. Brunkhorst-Kanaan N
    50. Budde M
    51. Bybjerg-Grauholm J
    52. Byerley W
    53. Cairns M
    54. Casas M
    55. Cervantes P
    56. Clarke T-K
    57. Cruceanu C
    58. Cuellar-Barboza A
    59. Cunningham J
    60. Curtis D
    61. Czerski PM
    62. Dale AM
    63. Dalkner N
    64. David FS
    65. Degenhardt F
    66. Djurovic S
    67. Dobbyn AL
    68. Douzenis A
    69. Elvsåshagen T
    70. Escott-Price V
    71. Ferrier IN
    72. Fiorentino A
    73. Foroud TM
    74. Forty L
    75. Frank J
    76. Frei O
    77. Freimer NB
    78. Frisén L
    79. Gade K
    80. Garnham J
    81. Gelernter J
    82. Giørtz Pedersen M
    83. Gizer IR
    84. Gordon SD
    85. Gordon-Smith K
    86. Greenwood TA
    87. Grove J
    88. Guzman-Parra J
    89. Ha K
    90. Haraldsson M
    91. Hautzinger M
    92. Heilbronner U
    93. Hellgren D
    94. Herms S
    95. Hoffmann P
    96. Holmans PA
    97. Huckins L
    98. Jamain S
    99. Johnson JS
    100. Kalman JL
    101. Kamatani Y
    102. Kennedy JL
    103. Kittel-Schneider S
    104. Knowles JA
    105. Kogevinas M
    106. Koromina M
    107. Kranz TM
    108. Kranzler HR
    109. Kubo M
    110. Kupka R
    111. Kushner SA
    112. Lavebratt C
    113. Lawrence J
    114. Leber M
    115. Lee H-J
    116. Lee PH
    117. Levy SE
    118. Lewis C
    119. Liao C
    120. Lucae S
    121. Lundberg M
    122. MacIntyre DJ
    123. Magnusson SH
    124. Maier W
    125. Maihofer A
    126. Malaspina D
    127. Maratou E
    128. Martinsson L
    129. Mattheisen M
    130. McCarroll SA
    131. McGregor NW
    132. McGuffin P
    133. McKay JD
    134. Medeiros H
    135. Medland SE
    136. Millischer V
    137. Montgomery GW
    138. Moran JL
    139. Morris DW
    140. Mühleisen TW
    141. O’Brien N
    142. O’Donovan C
    143. Olde Loohuis LM
    144. Oruc L
    145. Papiol S
    146. Pardiñas AF
    147. Perry A
    148. Pfennig A
    149. Porichi E
    150. Potash JB
    151. Quested D
    152. Raj T
    153. Rapaport MH
    154. DePaulo JR
    155. Regeer EJ
    156. Rice JP
    157. Rivas F
    158. Rivera M
    159. Roth J
    160. Roussos P
    161. Ruderfer DM
    162. Sánchez-Mora C
    163. Schulte EC
    164. Senner F
    165. Sharp S
    166. Shilling PD
    167. Sigurdsson E
    168. Sirignano L
    169. Slaney C
    170. Smeland OB
    171. Smith DJ
    172. Sobell JL
    173. Søholm Hansen C
    174. Soler Artigas M
    175. Spijker AT
    176. Stein DJ
    177. Strauss JS
    178. Świątkowska B
    179. Terao C
    180. Thorgeirsson TE
    181. Toma C
    182. Tooney P
    183. Tsermpini E-E
    184. Vawter MP
    185. Vedder H
    186. Walters JTR
    187. Witt SH
    188. Xi S
    189. Xu W
    190. Yang JMK
    191. Young AH
    192. Young H
    193. Zandi PP
    194. Zhou H
    195. Zillich L
    196. Adolfsson R
    197. Agartz I
    198. Alda M
    199. Alfredsson L
    200. Babadjanova G
    201. Backlund L
    202. Baune BT
    203. Bellivier F
    204. Bengesser S
    205. Berrettini WH
    206. Blackwood DHR
    207. Boehnke M
    208. Børglum AD
    209. Breen G
    210. Carr VJ
    211. Catts S
    212. Corvin A
    213. Craddock N
    214. Dannlowski U
    215. Dikeos D
    216. Esko T
    217. Etain B
    218. Ferentinos P
    219. Frye M
    220. Fullerton JM
    221. Gawlik M
    222. Gershon ES
    223. Goes FS
    224. Green MJ
    225. Grigoroiu-Serbanescu M
    226. Hauser J
    227. Henskens F
    228. Hillert J
    229. Hong KS
    230. Hougaard DM
    231. Hultman CM
    232. Hveem K
    233. Iwata N
    234. Jablensky AV
    235. Jones I
    236. Jones LA
    237. Kahn RS
    238. Kelsoe JR
    239. Kirov G
    240. Landén M
    241. Leboyer M
    242. Lewis CM
    243. Li QS
    244. Lissowska J
    245. Lochner C
    246. Loughland C
    247. Martin NG
    248. Mathews CA
    249. Mayoral F
    250. McElroy SL
    251. McIntosh AM
    252. McMahon FJ
    253. Melle I
    254. Michie P
    255. Milani L
    256. Mitchell PB
    257. Morken G
    258. Mors O
    259. Mortensen PB
    260. Mowry B
    261. Müller-Myhsok B
    262. Myers RM
    263. Neale BM
    264. Nievergelt CM
    265. Nordentoft M
    266. Nöthen MM
    267. O’Donovan MC
    268. Oedegaard KJ
    269. Olsson T
    270. Owen MJ
    271. Paciga SA
    272. Pantelis C
    273. Pato C
    274. Pato MT
    275. Patrinos GP
    276. Perlis RH
    277. Posthuma D
    278. Ramos-Quiroga JA
    279. Reif A
    280. Reininghaus EZ
    281. Ribasés M
    282. Rietschel M
    283. Ripke S
    284. Rouleau GA
    285. Saito T
    286. Schall U
    287. Schalling M
    288. Schofield PR
    289. Schulze TG
    290. Scott LJ
    291. Scott RJ
    292. Serretti A
    293. Shannon Weickert C
    294. Smoller JW
    295. Stefansson H
    296. Stefansson K
    297. Stordal E
    298. Streit F
    299. Sullivan PF
    300. Turecki G
    301. Vaaler AE
    302. Vieta E
    303. Vincent JB
    304. Waldman ID
    305. Weickert TW
    306. Werge T
    307. Wray NR
    308. Zwart J-A
    309. Biernacka JM
    310. Nurnberger JI
    311. Cichon S
    312. Edenberg HJ
    313. Stahl EA
    314. McQuillin A
    315. Di Florio A
    316. Ophoff RA
    317. Andreassen OA
    318. HUNT All-In Psychiatry
    (2021) Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology
    Nature Genetics 53:817–829.
    https://doi.org/10.1038/s41588-021-00857-4
  1. Book
    1. Raz N
    (2000)
    Aging of the brain and its impact on cognitive performance: integration of structural and functional findings
    University of Memphis.
  2. Book
    1. Townsend P
    2. Phillimore P
    3. Beattie A
    (2023)
    Health and deprivation: inequality and the north
    Taylor & Francis.
    1. Trubetskoy V
    2. Pardiñas AF
    3. Qi T
    4. Panagiotaropoulou G
    5. Awasthi S
    6. Bigdeli TB
    7. Bryois J
    8. Chen C-Y
    9. Dennison CA
    10. Hall LS
    11. Lam M
    12. Watanabe K
    13. Frei O
    14. Ge T
    15. Harwood JC
    16. Koopmans F
    17. Magnusson S
    18. Richards AL
    19. Sidorenko J
    20. Wu Y
    21. Zeng J
    22. Grove J
    23. Kim M
    24. Li Z
    25. Voloudakis G
    26. Zhang W
    27. Adams M
    28. Agartz I
    29. Atkinson EG
    30. Agerbo E
    31. Al Eissa M
    32. Albus M
    33. Alexander M
    34. Alizadeh BZ
    35. Alptekin K
    36. Als TD
    37. Amin F
    38. Arolt V
    39. Arrojo M
    40. Athanasiu L
    41. Azevedo MH
    42. Bacanu SA
    43. Bass NJ
    44. Begemann M
    45. Belliveau RA
    46. Bene J
    47. Benyamin B
    48. Bergen SE
    49. Blasi G
    50. Bobes J
    51. Bonassi S
    52. Braun A
    53. Bressan RA
    54. Bromet EJ
    55. Bruggeman R
    56. Buckley PF
    57. Buckner RL
    58. Bybjerg-Grauholm J
    59. Cahn W
    60. Cairns MJ
    61. Calkins ME
    62. Carr VJ
    63. Castle D
    64. Catts SV
    65. Chambert KD
    66. Chan RCK
    67. Chaumette B
    68. Cheng W
    69. Cheung EFC
    70. Chong SA
    71. Cohen D
    72. Consoli A
    73. Cordeiro Q
    74. Costas J
    75. Curtis C
    76. Davidson M
    77. Davis KL
    78. de Haan L
    79. Degenhardt F
    80. DeLisi LE
    81. Demontis D
    82. Dickerson F
    83. Dikeos D
    84. Dinan T
    85. Djurovic S
    86. Duan J
    87. Ducci G
    88. Dudbridge F
    89. Eriksson JG
    90. Fañanás L
    91. Faraone SV
    92. Fiorentino A
    93. Forstner A
    94. Frank J
    95. Freimer NB
    96. Fromer M
    97. Frustaci A
    98. Gadelha A
    99. Genovese G
    100. Gershon ES
    101. Giannitelli M
    102. Giegling I
    103. Giusti-Rodríguez P
    104. Godard S
    105. Goldstein JI
    106. González Peñas J
    107. González-Pinto A
    108. Gopal S
    109. Gratten J
    110. Green MF
    111. Greenwood TA
    112. Guillin O
    113. Gülöksüz S
    114. Gur RE
    115. Gur RC
    116. Gutiérrez B
    117. Hahn E
    118. Hakonarson H
    119. Haroutunian V
    120. Hartmann AM
    121. Harvey C
    122. Hayward C
    123. Henskens FA
    124. Herms S
    125. Hoffmann P
    126. Howrigan DP
    127. Ikeda M
    128. Iyegbe C
    129. Joa I
    130. Julià A
    131. Kähler AK
    132. Kam-Thong T
    133. Kamatani Y
    134. Karachanak-Yankova S
    135. Kebir O
    136. Keller MC
    137. Kelly BJ
    138. Khrunin A
    139. Kim S-W
    140. Klovins J
    141. Kondratiev N
    142. Konte B
    143. Kraft J
    144. Kubo M
    145. Kučinskas V
    146. Kučinskiene ZA
    147. Kusumawardhani A
    148. Kuzelova-Ptackova H
    149. Landi S
    150. Lazzeroni LC
    151. Lee PH
    152. Legge SE
    153. Lehrer DS
    154. Lencer R
    155. Lerer B
    156. Li M
    157. Lieberman J
    158. Light GA
    159. Limborska S
    160. Liu C-M
    161. Lönnqvist J
    162. Loughland CM
    163. Lubinski J
    164. Luykx JJ
    165. Lynham A
    166. Macek M Jr
    167. Mackinnon A
    168. Magnusson PKE
    169. Maher BS
    170. Maier W
    171. Malaspina D
    172. Mallet J
    173. Marder SR
    174. Marsal S
    175. Martin AR
    176. Martorell L
    177. Mattheisen M
    178. McCarley RW
    179. McDonald C
    180. McGrath JJ
    181. Medeiros H
    182. Meier S
    183. Melegh B
    184. Melle I
    185. Mesholam-Gately RI
    186. Metspalu A
    187. Michie PT
    188. Milani L
    189. Milanova V
    190. Mitjans M
    191. Molden E
    192. Molina E
    193. Molto MD
    194. Mondelli V
    195. Moreno C
    196. Morley CP
    197. Muntané G
    198. Murphy KC
    199. Myin-Germeys I
    200. Nenadić I
    201. Nestadt G
    202. Nikitina-Zake L
    203. Noto C
    204. Nuechterlein KH
    205. O’Brien NL
    206. O’Neill FA
    207. Oh S-Y
    208. Olincy A
    209. Ota VK
    210. Pantelis C
    211. Papadimitriou GN
    212. Parellada M
    213. Paunio T
    214. Pellegrino R
    215. Periyasamy S
    216. Perkins DO
    217. Pfuhlmann B
    218. Pietiläinen O
    219. Pimm J
    220. Porteous D
    221. Powell J
    222. Quattrone D
    223. Quested D
    224. Radant AD
    225. Rampino A
    226. Rapaport MH
    227. Rautanen A
    228. Reichenberg A
    229. Roe C
    230. Roffman JL
    231. Roth J
    232. Rothermundt M
    233. Rutten BPF
    234. Saker-Delye S
    235. Salomaa V
    236. Sanjuan J
    237. Santoro ML
    238. Savitz A
    239. Schall U
    240. Scott RJ
    241. Seidman LJ
    242. Sharp SI
    243. Shi J
    244. Siever LJ
    245. Sigurdsson E
    246. Sim K
    247. Skarabis N
    248. Slominsky P
    249. So H-C
    250. Sobell JL
    251. Söderman E
    252. Stain HJ
    253. Steen NE
    254. Steixner-Kumar AA
    255. Stögmann E
    256. Stone WS
    257. Straub RE
    258. Streit F
    259. Strengman E
    260. Stroup TS
    261. Subramaniam M
    262. Sugar CA
    263. Suvisaari J
    264. Svrakic DM
    265. Swerdlow NR
    266. Szatkiewicz JP
    267. Ta TMT
    268. Takahashi A
    269. Terao C
    270. Thibaut F
    271. Toncheva D
    272. Tooney PA
    273. Torretta S
    274. Tosato S
    275. Tura GB
    276. Turetsky BI
    277. Üçok A
    278. Vaaler A
    279. van Amelsvoort T
    280. van Winkel R
    281. Veijola J
    282. Waddington J
    283. Walter H
    284. Waterreus A
    285. Webb BT
    286. Weiser M
    287. Williams NM
    288. Witt SH
    289. Wormley BK
    290. Wu JQ
    291. Xu Z
    292. Yolken R
    293. Zai CC
    294. Zhou W
    295. Zhu F
    296. Zimprich F
    297. Atbaşoğlu EC
    298. Ayub M
    299. Benner C
    300. Bertolino A
    301. Black DW
    302. Bray NJ
    303. Breen G
    304. Buccola NG
    305. Byerley WF
    306. Chen WJ
    307. Cloninger CR
    308. Crespo-Facorro B
    309. Donohoe G
    310. Freedman R
    311. Galletly C
    312. Gandal MJ
    313. Gennarelli M
    314. Hougaard DM
    315. Hwu H-G
    316. Jablensky AV
    317. McCarroll SA
    318. Moran JL
    319. Mors O
    320. Mortensen PB
    321. Müller-Myhsok B
    322. Neil AL
    323. Nordentoft M
    324. Pato MT
    325. Petryshen TL
    326. Pirinen M
    327. Pulver AE
    328. Schulze TG
    329. Silverman JM
    330. Smoller JW
    331. Stahl EA
    332. Tsuang DW
    333. Vilella E
    334. Wang S-H
    335. Xu S
    336. Adolfsson R
    337. Arango C
    338. Baune BT
    339. Belangero SI
    340. Børglum AD
    341. Braff D
    342. Bramon E
    343. Buxbaum JD
    344. Campion D
    345. Cervilla JA
    346. Cichon S
    347. Collier DA
    348. Corvin A
    349. Curtis D
    350. Forti MD
    351. Domenici E
    352. Ehrenreich H
    353. Escott-Price V
    354. Esko T
    355. Fanous AH
    356. Gareeva A
    357. Gawlik M
    358. Gejman PV
    359. Gill M
    360. Glatt SJ
    361. Golimbet V
    362. Hong KS
    363. Hultman CM
    364. Hyman SE
    365. Iwata N
    366. Jönsson EG
    367. Kahn RS
    368. Kennedy JL
    369. Khusnutdinova E
    370. Kirov G
    371. Knowles JA
    372. Krebs M-O
    373. Laurent-Levinson C
    374. Lee J
    375. Lencz T
    376. Levinson DF
    377. Li QS
    378. Liu J
    379. Malhotra AK
    380. Malhotra D
    381. McIntosh A
    382. McQuillin A
    383. Menezes PR
    384. Morgan VA
    385. Morris DW
    386. Mowry BJ
    387. Murray RM
    388. Nimgaonkar V
    389. Nöthen MM
    390. Ophoff RA
    391. Paciga SA
    392. Palotie A
    393. Pato CN
    394. Qin S
    395. Rietschel M
    396. Riley BP
    397. Rivera M
    398. Rujescu D
    399. Saka MC
    400. Sanders AR
    401. Schwab SG
    402. Serretti A
    403. Sham PC
    404. Shi Y
    405. St Clair D
    406. Stefánsson H
    407. Stefansson K
    408. Tsuang MT
    409. van Os J
    410. Vawter MP
    411. Weinberger DR
    412. Werge T
    413. Wildenauer DB
    414. Yu X
    415. Yue W
    416. Holmans PA
    417. Pocklington AJ
    418. Roussos P
    419. Vassos E
    420. Verhage M
    421. Visscher PM
    422. Yang J
    423. Posthuma D
    424. Andreassen OA
    425. Kendler KS
    426. Owen MJ
    427. Wray NR
    428. Daly MJ
    429. Huang H
    430. Neale BM
    431. Sullivan PF
    432. Ripke S
    433. Walters JTR
    434. O’Donovan MC
    435. Indonesia Schizophrenia Consortium
    436. PsychENCODE
    437. Psychosis Endophenotypes International Consortium
    438. SynGO Consortium
    439. Schizophrenia Working Group of the Psychiatric Genomics Consortium
    (2022) Mapping genomic loci implicates genes and synaptic biology in schizophrenia
    Nature 604:502–508.
    https://doi.org/10.1038/s41586-022-04434-5
    1. Wray NR
    2. Ripke S
    3. Mattheisen M
    4. Trzaskowski M
    5. Byrne EM
    6. Abdellaoui A
    7. Adams MJ
    8. Agerbo E
    9. Air TM
    10. Andlauer TMF
    11. Bacanu SA
    12. Bækvad-Hansen M
    13. Beekman AFT
    14. Bigdeli TB
    15. Binder EB
    16. Blackwood DRH
    17. Bryois J
    18. Buttenschøn HN
    19. Bybjerg-Grauholm J
    20. Cai N
    21. Castelao E
    22. Christensen JH
    23. Clarke TK
    24. Coleman JIR
    25. Colodro-Conde L
    26. Couvy-Duchesne B
    27. Craddock N
    28. Crawford GE
    29. Crowley CA
    30. Dashti HS
    31. Davies G
    32. Deary IJ
    33. Degenhardt F
    34. Derks EM
    35. Direk N
    36. Dolan CV
    37. Dunn EC
    38. Eley TC
    39. Eriksson N
    40. Escott-Price V
    41. Kiadeh FHF
    42. Finucane HK
    43. Forstner AJ
    44. Frank J
    45. Gaspar HA
    46. Gill M
    47. Giusti-Rodríguez P
    48. Goes FS
    49. Gordon SD
    50. Grove J
    51. Hall LS
    52. Hannon E
    53. Hansen CS
    54. Hansen TF
    55. Herms S
    56. Hickie IB
    57. Hoffmann P
    58. Homuth G
    59. Horn C
    60. Hottenga JJ
    61. Hougaard DM
    62. Hu M
    63. Hyde CL
    64. Ising M
    65. Jansen R
    66. Jin F
    67. Jorgenson E
    68. Knowles JA
    69. Kohane IS
    70. Kraft J
    71. Kretzschmar WW
    72. Krogh J
    73. Kutalik Z
    74. Lane JM
    75. Li Y
    76. Li Y
    77. Lind PA
    78. Liu X
    79. Lu L
    80. MacIntyre DJ
    81. MacKinnon DF
    82. Maier RM
    83. Maier W
    84. Marchini J
    85. Mbarek H
    86. McGrath P
    87. McGuffin P
    88. Medland SE
    89. Mehta D
    90. Middeldorp CM
    91. Mihailov E
    92. Milaneschi Y
    93. Milani L
    94. Mill J
    95. Mondimore FM
    96. Montgomery GW
    97. Mostafavi S
    98. Mullins N
    99. Nauck M
    100. Ng B
    101. Nivard MG
    102. Nyholt DR
    103. O’Reilly PF
    104. Oskarsson H
    105. Owen MJ
    106. Painter JN
    107. Pedersen CB
    108. Pedersen MG
    109. Peterson RE
    110. Pettersson E
    111. Peyrot WJ
    112. Pistis G
    113. Posthuma D
    114. Purcell SM
    115. Quiroz JA
    116. Qvist P
    117. Rice JP
    118. Riley BP
    119. Rivera M
    120. Saeed Mirza S
    121. Saxena R
    122. Schoevers R
    123. Schulte EC
    124. Shen L
    125. Shi J
    126. Shyn SI
    127. Sigurdsson E
    128. Sinnamon GBC
    129. Smit JH
    130. Smith DJ
    131. Stefansson H
    132. Steinberg S
    133. Stockmeier CA
    134. Streit F
    135. Strohmaier J
    136. Tansey KE
    137. Teismann H
    138. Teumer A
    139. Thompson W
    140. Thomson PA
    141. Thorgeirsson TE
    142. Tian C
    143. Traylor M
    144. Treutlein J
    145. Trubetskoy V
    146. Uitterlinden AG
    147. Umbricht D
    148. Van der Auwera S
    149. van Hemert AM
    150. Viktorin A
    151. Visscher PM
    152. Wang Y
    153. Webb BT
    154. Weinsheimer SM
    155. Wellmann J
    156. Willemsen G
    157. Witt SH
    158. Wu Y
    159. Xi HS
    160. Yang J
    161. Zhang F
    162. Arolt V
    163. Baune BT
    164. Berger K
    165. Boomsma DI
    166. Cichon S
    167. Dannlowski U
    168. de Geus ECJ
    169. DePaulo JR
    170. Domenici E
    171. Domschke K
    172. Esko T
    173. Grabe HJ
    174. Hamilton SP
    175. Hayward C
    176. Heath AC
    177. Hinds DA
    178. Kendler KS
    179. Kloiber S
    180. Lewis G
    181. Li QS
    182. Lucae S
    183. Madden PFA
    184. Magnusson PK
    185. Martin NG
    186. McIntosh AM
    187. Metspalu A
    188. Mors O
    189. Mortensen PB
    190. Müller-Myhsok B
    191. Nordentoft M
    192. Nöthen MM
    193. O’Donovan MC
    194. Paciga SA
    195. Pedersen NL
    196. Penninx B
    197. Perlis RH
    198. Porteous DJ
    199. Potash JB
    200. Preisig M
    201. Rietschel M
    202. Schaefer C
    203. Schulze TG
    204. Smoller JW
    205. Stefansson K
    206. Tiemeier H
    207. Uher R
    208. Völzke H
    209. Weissman MM
    210. Werge T
    211. Winslow AR
    212. Lewis CM
    213. Levinson DF
    214. Breen G
    215. Børglum AD
    216. Sullivan PF
    217. eQTLGen
    218. 23andMe
    219. Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium
    (2018) Genome-wide association analyses identify 44 risk variam genomics of parkinson’s disease consortiumnts and refine the genetic architecture of major depression
    Nature Genetics 50:668–681.
    https://doi.org/10.1038/s41588-018-0090-3
    1. Zheng H-F
    2. Forgetta V
    3. Hsu Y-H
    4. Estrada K
    5. Rosello-Diez A
    6. Leo PJ
    7. Dahia CL
    8. Park-Min KH
    9. Tobias JH
    10. Kooperberg C
    11. Kleinman A
    12. Styrkarsdottir U
    13. Liu C-T
    14. Uggla C
    15. Evans DS
    16. Nielson CM
    17. Walter K
    18. Pettersson-Kymmer U
    19. McCarthy S
    20. Eriksson J
    21. Kwan T
    22. Jhamai M
    23. Trajanoska K
    24. Memari Y
    25. Min J
    26. Huang J
    27. Danecek P
    28. Wilmot B
    29. Li R
    30. Chou W-C
    31. Mokry LE
    32. Moayyeri A
    33. Claussnitzer M
    34. Cheng C-H
    35. Cheung W
    36. Medina-Gómez C
    37. Ge B
    38. Chen S-H
    39. Choi K
    40. Oei L
    41. Fraser J
    42. Kraaij R
    43. Hibbs MA
    44. Gregson CL
    45. Paquette D
    46. Hofman A
    47. Wibom C
    48. Tranah GJ
    49. Marshall M
    50. Gardiner BB
    51. Cremin K
    52. Auer P
    53. Hsu L
    54. Ring S
    55. Tung JY
    56. Thorleifsson G
    57. Enneman AW
    58. van Schoor NM
    59. de Groot LC
    60. van der Velde N
    61. Melin B
    62. Kemp JP
    63. Christiansen C
    64. Sayers A
    65. Zhou Y
    66. Calderari S
    67. van Rooij J
    68. Carlson C
    69. Peters U
    70. Berlivet S
    71. Dostie J
    72. Uitterlinden AG
    73. Williams SR
    74. Farber C
    75. Grinberg D
    76. LaCroix AZ
    77. Haessler J
    78. Chasman DI
    79. Giulianini F
    80. Rose LM
    81. Ridker PM
    82. Eisman JA
    83. Nguyen TV
    84. Center JR
    85. Nogues X
    86. Garcia-Giralt N
    87. Launer LL
    88. Gudnason V
    89. Mellström D
    90. Vandenput L
    91. Amin N
    92. van Duijn CM
    93. Karlsson MK
    94. Ljunggren Ö
    95. Svensson O
    96. Hallmans G
    97. Rousseau F
    98. Giroux S
    99. Bussière J
    100. Arp PP
    101. Koromani F
    102. Prince RL
    103. Lewis JR
    104. Langdahl BL
    105. Hermann AP
    106. Jensen J-EB
    107. Kaptoge S
    108. Khaw K-T
    109. Reeve J
    110. Formosa MM
    111. Xuereb-Anastasi A
    112. Åkesson K
    113. McGuigan FE
    114. Garg G
    115. Olmos JM
    116. Zarrabeitia MT
    117. Riancho JA
    118. Ralston SH
    119. Alonso N
    120. Jiang X
    121. Goltzman D
    122. Pastinen T
    123. Grundberg E
    124. Gauguier D
    125. Orwoll ES
    126. Karasik D
    127. Davey-Smith G
    128. AOGC Consortium
    129. Smith AV
    130. Siggeirsdottir K
    131. Harris TB
    132. Zillikens MC
    133. van Meurs JBJ
    134. Thorsteinsdottir U
    135. Maurano MT
    136. Timpson NJ
    137. Soranzo N
    138. Durbin R
    139. Wilson SG
    140. Ntzani EE
    141. Brown MA
    142. Stefansson K
    143. Hinds DA
    144. Spector T
    145. Cupples LA
    146. Ohlsson C
    147. Greenwood CMT
    148. UK10K Consortium
    149. Jackson RD
    150. Rowe DW
    151. Loomis CA
    152. Evans DM
    153. Ackert-Bicknell CL
    154. Joyner AL
    155. Duncan EL
    156. Kiel DP
    157. Rivadeneira F
    158. Richards JB
    (2015) Whole-genome sequencing identifies EN1 as a determinant of bone density and fracture
    Nature 526:112–117.
    https://doi.org/10.1038/nature14878

Article and author information

Author details

  1. Haojing Duan

    1. Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
    2. Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
    Contribution
    Software, Formal analysis, Visualization, Methodology, Writing - original draft, Writing – review and editing
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0009-0004-8659-9591
  2. Runye Shi

    School of Data Science, Fudan University, Shanghai, China
    Contribution
    Data curation, Visualization, Writing – review and editing
    Competing interests
    No competing interests declared
  3. Jujiao Kang

    1. Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
    2. Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  4. Tobias Banaschewski

    Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    Dr Banaschewski served in an advisory or consultancy role for eye level, Infectopharm, Lundbeck, Medice, Neurim Pharmaceuticals, Oberberg GmbH, Roche, and Takeda. He received conference support or speaker's fee by Janssen, Medice and Takeda. He received royalities from Hogrefe, Kohlhammer, CIP Medien, Oxford University Press; the presentwork is unrelated to these relationships
  5. Arun LW Bokde

    Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  6. Christian Büchel

    University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    Reviewing editor, eLife
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-1965-906X
  7. Sylvane Desrivières

    Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
    Contribution
    Data curation, Funding acquisition, Writing – review and editing
    Competing interests
    No competing interests declared
  8. Herta Flor

    1. Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
    2. Department of Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  9. Antoine Grigis

    NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  10. Hugh Garavan

    Departments of Psychiatry and Psychology, University of Vermont, Burlington, United States
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  11. Penny A Gowland

    Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, Nottingham, United Kingdom
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  12. Andreas Heinz

    Department of Psychiatry and Psychotherapy CCM, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  13. Rüdiger Brühl

    Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Berlin, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-0111-5996
  14. Jean-Luc Martinot

    Institut National de la Santé et de la Recherche Médicale, INSERM U1299 "Developmental Trajectories and Psychiatry", Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
    Contribution
    Data curation, Funding acquisition, Writing – review and editing
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-0136-0388
  15. Marie-Laure Paillère Martinot

    1. Institut National de la Santé et de la Recherche Médicale, INSERM U1299 "Developmental Trajectories and Psychiatry", Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
    2. AP-HP. Sorbonne Université, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, Paris, France
    Contribution
    Data curation, Funding acquisition, Writing – review and editing
    Competing interests
    No competing interests declared
  16. Eric Artiges

    1. Institut National de la Santé et de la Recherche Médicale, INSERM U1299 "Developmental Trajectories and Psychiatry", Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
    2. Psychiatry Department, EPS Barthélémy Durand, Etampes, France
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  17. Frauke Nees

    1. Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
    2. Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
    3. Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  18. Dimitri Papadopoulos Orfanos

    NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-1242-8990
  19. Luise Poustka

    Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre, Göttingen, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    Dr Poustka served in an advisory or consultancy role for Roche and Viforpharm and received speaker's fee by Shire. She received royalties from Hogrefe, Kohlhammer and Schattauer. The present work is unrelated to the above grants and relationships
  20. Sarah Hohmann

    Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  21. Nathalie Nathalie Holz

    Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  22. Juliane Fröhner

    Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  23. Michael N Smolka

    Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-5398-5569
  24. Nilakshi Vaidya

    Department of Psychiatry and Neurosciences, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  25. Henrik Walter

    Department of Psychiatry and Psychotherapy CCM, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
    Contribution
    Data curation, Writing – review and editing
    Competing interests
    No competing interests declared
  26. Robert Whelan

    School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
    Contribution
    Data curation, Funding acquisition, Writing – review and editing
    Competing interests
    No competing interests declared
  27. Gunter Schumann

    1. Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
    2. Department of Psychiatry and Neurosciences, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
    3. Centre for Population Neuroscience and Stratified Medicine (PONS Centre), ISTBI, Fudan University, Shanghai, China
    4. Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
    Contribution
    Data curation, Funding acquisition, Writing – review and editing
    Competing interests
    No competing interests declared
  28. Xiaolei Lin

    1. School of Data Science, Fudan University, Shanghai, China
    2. Huashan Institute of Medicine, Huashan Hospital affiliated to Fudan University, Shanghai, China
    Contribution
    Conceptualization, Funding acquisition, Methodology, Writing - original draft, Writing – review and editing
    For correspondence
    xiaoleilin@fudan.edu.cn
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-2463-1272
  29. Jianfeng Feng

    1. Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
    2. Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
    3. School of Data Science, Fudan University, Shanghai, China
    4. Centre for Population Neuroscience and Stratified Medicine (PONS Centre), ISTBI, Fudan University, Shanghai, China
    5. MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
    6. Zhangjiang Fudan International Innovation Center, Shanghai, China
    7. Department of Computer Science, University of Warwick, Warwick, United Kingdom
    Contribution
    Conceptualization, Funding acquisition, Writing – review and editing
    For correspondence
    jianfeng64@gmail.com
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-5987-2258

Funding

National Natural Science Foundation of China (No.82304241)

  • Xiaolei Lin

National Key Research and Development Program of China (No.2019YFA0709502)

  • Jianfeng Feng

National Key Research and Development Program of China (No.2018YFC1312904)

  • Jianfeng Feng

Shanghai Municipal Science and Technology Major Project (No.2018SHZDZX01)

  • Jianfeng Feng

ZJ Lab

  • Jianfeng Feng

Shanghai Center for Brain Science and Brain-Inspired Technology

  • Jianfeng Feng

111 project (No.B18015)

  • Jianfeng Feng

European Union FP6 Integrated Project IMAGEN (LSHM-CT- 2007-037286)

  • Gunter Schumann

Horizon 2020 ERC Advanced Grant 'STRATIFY' (695313)

  • Gunter Schumann

Human Brain Project (HBP SGA 2,785907)

  • Gunter Schumann

Human Brain Project (HBP SGA 3,945539)

  • Gunter Schumann

Medical Research Council Grant 'c-VEDA' (MR/N000390/1)

  • Gunter Schumann

National Institutes of Health (R01DA049238)

  • Gunter Schumann

National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London

  • Gunter Schumann

Deutsche Forschungsgemeinschaft (SM 80/7-2)

  • Gunter Schumann

Medical Research Foundation and Medical Research Council (MR/R00465X/1)

  • Sylvane Desrivières

National Institutes of Health (ENIGMA 5U54EB020403-05)

  • Sylvane Desrivières

National Institutes of Health (U54 EB020403)

  • Sylvane Desrivières

ANR (ANR-12-SAMA-0004 AAPG2019 - GeBra)

  • Jean-Luc Martinot

Eranet Neuron (AF12-NEUR0008-01 - WM2NA)

  • Jean-Luc Martinot

Fondation de France (00081242)

  • Jean-Luc Martinot

Fondation pour la Recherche Médicale (DPA20140629802)

  • Jean-Luc Martinot

Mission Interministérielle de Lutte-contre-les-Drogues-et-les-Conduites-Addictives (MILDECA)

  • Jean-Luc Martinot

Assistance-Publique-Hôpitaux-de-Paris and INSERM (interface grant)

  • Marie-Laure Paillère Martinot

Paris Sud University IDEX 2012

  • Jean-Luc Martinot

Fondation de l'Avenir (AP-RM-17-013)

  • Marie-Laure Paillère Martinot

Fédération pour la Recherche sur le Cerveau

  • Gunter Schumann

National Institutes of Health, Science Foundation Ireland (16/ERCD/3797)

  • Robert Whelan

NSFC (82150710554)

  • Gunter Schumann

environMENTAL (grant no: 101057429)

  • Gunter Schumann

Bundesministeriumfür Bildung und Forschung (01GS08152)

  • Gunter Schumann

Bundesministeriumfür Bildung und Forschung (01EV0711)

  • Gunter Schumann

Forschungsnetz AERIAL (01EE1406A)

  • Gunter Schumann

Forschungsnetz AERIAL (01EE1406B)

  • Gunter Schumann

Forschungsnetz IMAC-Mind (01GL1745B)

  • Gunter Schumann

Deutsche Forschungsgemeinschaft (SFB 940)

  • Gunter Schumann

Deutsche Forschungsgemeinschaft (TRR 265)

  • Gunter Schumann

Deutsche Forschungsgemeinschaft (NE 1383/14-1)

  • Gunter Schumann

Medical Research Foundation and Medical Research Council (MR/S020306/1)

  • Sylvane Desrivières

National Institutes of Health (ENIGMA 1R56AG058854-01)

  • Sylvane Desrivières

Eranet (ANR-18-NEUR00002-01 - ADORe)

  • Jean-Luc Martinot

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Acknowledgements

This research used the UK Biobank Resource under application number 19542. We thank all participants and researchers from the UK Biobank. We thank the IMAGEN Consortium for providing the discover data. This work received support from the following sources: the National Nature Science Foundation of China (No.82304241 [to XL]), National Key R&D Program of China (No.2019YFA0709502 [to JF], No.2018YFC1312904 [to JF]), Shanghai Municipal Science and Technology Major Project (No.2018SHZDZX01 [to JF], ZJ Lab [to JF], and Shanghai Center for Brain Science and Brain-Inspired Technology [to JF]), the 111 Project (No.B18015 [to JF]), the European Union-funded FP6 Integrated Project IMAGEN (Reinforcement-related behaviour in normal brain function and psychopathology) (LSHM-CT- 2007–037286 [to GS]), the Horizon 2020 funded ERC Advanced Grant ‘STRATIFY’ (Brain network based stratification of reinforcement-related disorders) (695313 [to GS]), Human Brain Project (HBP SGA 2, 785907, and HBP SGA 3, 945539 [to GS]), the Medical Research Council Grant 'c-VEDA’ (Consortium on Vulnerability to Externalizing Disorders and Addictions) (MR/N000390/1 [to GS]), the National Institute of Health (NIH) (R01DA049238 [to GS], A decentralized macro and micro gene-by-environment interaction analysis of substance use behavior and its brain biomarkers), the National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, the Bundesministeriumfür Bildung und Forschung (BMBF grants 01GS08152; 01EV0711 [to GS]; Forschungsnetz AERIAL 01EE1406A, 01EE1406B; Forschungsnetz IMAC-Mind 01GL1745B [to GS]), the Deutsche Forschungsgemeinschaft (DFG grants SM 80/7–2, SFB 940, TRR 265, NE 1383/14–1 [to GS]), the Medical Research Foundation and Medical Research Council (grants MR/R00465X/1 and MR/S020306/1 [to SD]), the National Institutes of Health (NIH) funded ENIGMA (grants 5U54EB020403-05 and 1R56AG058854-01 [to SD]), NSFC grant 82150710554 [to GS] and European Union funded project ‘environMENTAL’, grant no: 101057429 [to GS]. Further support was provided by grants from: - the ANR (ANR-12-SAMA-0004, AAPG2019 - GeBra [to JLM]), the Eranet Neuron (AF12-NEUR0008-01 - WM2NA; and ANR-18-NEUR00002-01 - ADORe [to JLM]), the Fondation de France (00081242 [to J.-L.M.]), the Fondation pour la Recherche Médicale (DPA20140629802 [to JLM]), the Mission Interministérielle de Lutte-contre-les-Drogues-et-les-Conduites-Addictives (MILDECA [to JLM]), the Assistance-Publique-Hôpitaux-de-Paris and INSERM (interface grant [to MLPM]), Paris Sud University IDEX 2012 [to J.-LM], the Fondation de l’Avenir (grant AP-RM-17–013 [to MLPM]), the Fédération pour la Recherche sur le Cerveau [to GS]; the National Institutes of Health, Science Foundation Ireland (16/ERCD/3797 [to RW]) and by NIH Consortium grant U54 EB020403 [to SD], supported by a cross-NIH alliance that funds Big Data to Knowledge Centres of Excellence. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Ethics

All participants from UK Biobank provided written informed consent, and ethical approval was granted by the North West Multi-Center Ethics committee (https://www.ukbiobank.ac.uk/learn-more-about-uk-biobank/about-us/ethics) with research ethics committee (REC) approval number 16/NW/0274. The lMAGEN study was approved by local ethics research committees of King's College London, University of Nottingham. Trinity College Dublin, University of Heidelberg, Technische Universität Dresden, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, and University Medical Center at the University of Hamburg in compliance with the Declaration of Helsinki (https://doi.org/10.1001/jama.2013.281053). Informed consent was given by all participants and a parent/guardian of each participant.

Version history

  1. Sent for peer review:
  2. Preprint posted:
  3. Reviewed Preprint version 1:
  4. Reviewed Preprint version 2:
  5. Version of Record published:

Cite all versions

You can cite all versions using the DOI https://doi.org/10.7554/eLife.94970. This DOI represents all versions, and will always resolve to the latest one.

Copyright

© 2024, Duan et al.

This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 543
    views
  • 26
    downloads
  • 0
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Haojing Duan
  2. Runye Shi
  3. Jujiao Kang
  4. Tobias Banaschewski
  5. Arun LW Bokde
  6. Christian Büchel
  7. Sylvane Desrivières
  8. Herta Flor
  9. Antoine Grigis
  10. Hugh Garavan
  11. Penny A Gowland
  12. Andreas Heinz
  13. Rüdiger Brühl
  14. Jean-Luc Martinot
  15. Marie-Laure Paillère Martinot
  16. Eric Artiges
  17. Frauke Nees
  18. Dimitri Papadopoulos Orfanos
  19. Luise Poustka
  20. Sarah Hohmann
  21. Nathalie Nathalie Holz
  22. Juliane Fröhner
  23. Michael N Smolka
  24. Nilakshi Vaidya
  25. Henrik Walter
  26. Robert Whelan
  27. Gunter Schumann
  28. Xiaolei Lin
  29. Jianfeng Feng
(2024)
Population clustering of structural brain aging and its association with brain development
eLife 13:RP94970.
https://doi.org/10.7554/eLife.94970.3

Share this article

https://doi.org/10.7554/eLife.94970

Further reading

    1. Evolutionary Biology
    2. Genetics and Genomics
    Hanna Tutaj, Katarzyna Tomala ... Ryszard Korona
    Research Article

    The loss of a single chromosome in a diploid organism halves the dosage of many genes and is usually accompanied by a substantial decrease in fitness. We asked whether this decrease simply reflects the joint damage caused by individual gene dosage deficiencies. We measured the fitness effects of single heterozygous gene deletions in yeast and combined them for each chromosome. This predicted a negative growth rate, that is, lethality, for multiple monosomies. However, monosomic strains remained alive and grew as if much (often most) of the damage caused by single mutations had disappeared, revealing an exceptionally large and positive epistatic component of fitness. We looked for functional explanations by analyzing the transcriptomes. There was no evidence of increased (compensatory) gene expression on the monosomic chromosomes. Nor were there signs of the cellular stress response that would be expected if monosomy led to protein destabilization and thus cytotoxicity. Instead, all monosomic strains showed extensive upregulation of genes encoding ribosomal proteins, but in an indiscriminate manner that did not correspond to their altered dosage. This response did not restore the stoichiometry required for efficient biosynthesis, which probably became growth limiting, making all other mutation-induced metabolic defects much less important. In general, the modular structure of the cell leads to an effective fragmentation of the total mutational load. Defects outside the module(s) currently defining fitness lose at least some of their relevance, producing the epiphenomenon of positive interactions between individually negative effects.

    1. Genetics and Genomics
    Yi-Ting Tsai, Nogayhan Seymen ... Mohammad M Karimi
    Research Article

    Retrotransposons (RTEs) have been postulated to reactivate with age and contribute to aging through activated innate immune response and inflammation. Here, we analyzed the relationship between RTE expression and aging using published transcriptomic and methylomic datasets of human blood. Despite no observed correlation between RTE activity and chronological age, the expression of most RTE classes and families except short interspersed nuclear elements (SINEs) correlated with biological age-associated gene signature scores. Strikingly, we found that the expression of SINEs was linked to upregulated DNA repair pathways in multiple cohorts. We also observed DNA hypomethylation with aging and the significant increase in RTE expression level in hypomethylated RTEs except for SINEs. Additionally, our single-cell transcriptomic analysis suggested a role for plasma cells in aging mediated by RTEs. Altogether, our multi-omics analysis of large human cohorts highlights the role of RTEs in biological aging and suggests possible mechanisms and cell populations for future investigations.