Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Brain aging patterns in a large and diverse cohort of 49,482 individuals

Abstract

Brain aging process is influenced by various lifestyle, environmental and genetic factors, as well as by age-related and often coexisting pathologies. Magnetic resonance imaging and artificial intelligence methods have been instrumental in understanding neuroanatomical changes that occur during aging. Large, diverse population studies enable identifying comprehensive and representative brain change patterns resulting from distinct but overlapping pathological and biological factors, revealing intersections and heterogeneity in affected brain regions and clinical phenotypes. Herein, we leverage a state-of-the-art deep-representation learning method, Surreal-GAN, and present methodological advances and extensive experimental results elucidating brain aging heterogeneity in a cohort of 49,482 individuals from 11 studies. Five dominant patterns of brain atrophy were identified and quantified for each individual by respective measures, R-indices. Their associations with biomedical, lifestyle and genetic factors provide insights into the etiology of observed variances, suggesting their potential as brain endophenotypes for genetic and lifestyle risks. Furthermore, baseline R-indices predict disease progression and mortality, capturing early changes as supplementary prognostic markers. These R-indices establish a dimensional approach to measuring aging trajectories and related brain changes. They hold promise for precise diagnostics, especially at preclinical stages, facilitating personalized patient management and targeted clinical trial recruitment based on specific brain endophenotypic expression and prognosis.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Surreal-GAN disentangles brain aging heterogeneity through a dimensional representation approach.
Fig. 2: Surreal-GAN identifies five dimensions of brain aging.
Fig. 3: R-indices are associated with chronic diseases, and MCI/Dementia progression, and the risk of mortality.
Fig. 4: Associations between R-indices and lifestyle, cognition and CSF/plasma biomarkers.
Fig. 5: Five R-indices were associated with 73 genomic loci.
Fig. 6: The R-indices can have broad implications for healthcare.

Similar content being viewed by others

Data availability

All derived R-indices in this study are available at Supplementary Data 12, indexed by participant ID in the iSTAGING study. Additional raw imaging and clinical data used in this study were provided by several individual studies via data-sharing agreements, which do not include permission for us to further share the data. Investigators must apply to the source data providers to access additional data and match their subject IDs to those used in this study under the current protocol (primarily for UKBB). Data from ADNI are available from the ADNI database (adni.loni.usc.edu) upon registration and compliance with the data usage agreement. Data from the UKBB are available upon request from the UKBB website (https://www.ukbiobank.ac.uk/). Data from the BLSA study are available upon request at https://www.blsa.nih.gov/how-apply. Data from the AIBL study are available upon request at https://aibl.org.au/. Data from the OASIS study are available upon request at https://www.oasis-brains.org/. Data requests for BIOCARD, PENN, WRAP, CARDIA, SHIP and WHIMS datasets should be directed to M.S.A., D.A.W., S.C.J., L.J.L., K.W. and M.A.E., respectively. As soon as access to the source studies is obtained, investigators can match our derived R-indices to the rest of the data from these studies. Further assistance in matching the R-indices can be requested from the corresponding author, C.D., at Christos.Davatzikos@pennmedicine.upenn.edu, with responses typically provided within 2 weeks. Moreover, we are actively following protocols to upload our derived measures to the UKBB and ADNI websites, making them directly accessible to investigators who obtain access to those studies. The pretrained model for deriving R-indices in this study is available at https://github.com/zhijian-yang/SurrealGAN/blob/main/pretrained_models/brain_aging_5rindices/. Researchers can derive R-indices on their own datasets by following the data processing pipeline outlined in Method 9 and the model application process in Method 4, along with the example script on the same GitHub repository. The GWAS summary statistics are publicly available at https://labs-laboratory.com/medicine.

Code availability

The software Surreal-GAN is available as a published PyPI package. Detailed information about software installation, usage and license can be found at https://pypi.org/project/SurrealGAN/0.1.1/. Custom code can be found at https://github.com/zhijian-yang/SurrealGAN. The deep-learning models are currently being integrated into NiChart (neuroimagingchart.com), enabling researchers to quickly derive R-indices for out-of-domain structural brain MRI scans.

References

  1. Peters, R. Ageing and the brain. Postgrad. Med J. 82, 84–88 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Davatzikos, C., Xu, F., An, Y., Fan, Y. & Resnick, S. M. Longitudinal progression of Alzheimer’s-like patterns of atrophy in normal older adults: the SPARE-AD index. Brain 132, 2026–2035 (2009).

    PubMed  PubMed Central  Google Scholar 

  3. Dubois, B. et al. Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimers Dement. 12, 292–323 (2016).

    PubMed  Google Scholar 

  4. Davatzikos, C. Machine learning in neuroimaging: progress and challenges. Neuroimage 197, 652–656 (2019).

    PubMed  Google Scholar 

  5. Tian, Y. E. et al. Heterogeneous aging across multiple organ systems and prediction of chronic disease and mortality. Nat. Med. 29, 1221–1231 (2023).

    CAS  PubMed  Google Scholar 

  6. Habes, M. et al. Advanced brain aging: relationship with epidemiologic and genetic risk factors, and overlap with Alzheimer disease atrophy patterns. Transl. Psychiatry 6, e775 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Yang, Z. et al. A deep learning framework identifies dimensional representations of Alzheimer’s disease from brain structure. Nat. Commun. 12, 7065 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhang, X. et al. Bayesian model reveals latent atrophy factors with dissociable cognitive trajectories in Alzheimer’s disease. Proc. Natl Acad. Sci. USA 113, E6535–e6544 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Wen, J. et al. Multi-scale semi-supervised clustering of brain images: deriving disease subtypes. Med. Image Anal. 75, 102304 (2022).

    PubMed  Google Scholar 

  10. Yang, Z., Wen, J. & Davatzikos, C. Surreal-GAN: Semi-Supervised Representation Learning via GAN for uncovering heterogeneous disease-related imaging patterns. International Conference on Learning Representations (ICLR, 2022).

  11. Habes, M. et al. The brain chart of aging: Machine-learning analytics reveals links between brain aging, white matter disease, amyloid burden, and cognition in the iSTAGING consortium of 10,216 harmonized MR scans. Alzheimers Dement. 17, 89–102 (2021).

    CAS  PubMed  Google Scholar 

  12. Cox, S. R. et al. Ageing and brain white matter structure in 3,513 UK Biobank participants. Nat. Commun. 7, 13629 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Hedden, T. & Gabrieli, J. D. E. Insights into the ageing mind: a view from cognitive neuroscience. Nat. Rev. Neurosci. 5, 87–96 (2004).

    CAS  PubMed  Google Scholar 

  14. Wang, M. C., Shah, N. S., Carnethon, M. R., O’Brien, M. J. & Khan, S. S. Age at diagnosis of diabetes by race and ethnicity in the United States from 2011 to 2018. JAMA Intern. Med. 181, 1537–1539 (2021).

    PubMed  PubMed Central  Google Scholar 

  15. Huang, X., Lee, K., Wang, M. C., Shah, N. S. & Khan, S. S. Age at diagnosis of hypertension by race and ethnicity in the US from 2011 to 2020. JAMA Cardiol. 7, 986–987 (2022).

    PubMed  PubMed Central  Google Scholar 

  16. Abbott, A. Dementia: a problem for our age. Nature 475, S2–S4 (2011).

    CAS  PubMed  Google Scholar 

  17. Dwyer, D. B. et al. Psychosis brain subtypes validated in first-episode cohorts and related to illness remission: results from the PHENOM consortium. Mol. Psychiatry 28, 2008–2017 (2023).

    PubMed  PubMed Central  Google Scholar 

  18. Stark, K. & Massberg, S. Interplay between inflammation and thrombosis in cardiovascular pathology. Nat. Rev. Cardiol. 18, 666–682 (2021).

    PubMed  PubMed Central  Google Scholar 

  19. Rose-John, S., Winthrop, K. & Calabrese, L. The role of IL-6 in host defence against infections: immunobiology and clinical implications. Nat. Rev. Rheumatol. 13, 399–409 (2017).

    CAS  PubMed  Google Scholar 

  20. Dutta, G., Barber, D. S., Zhang, P., Doperalski, N. J. & Liu, B. Involvement of dopaminergic neuronal cystatin C in neuronal injury-induced microglial activation and neurotoxicity. J. Neurochem. 122, 752–763 (2012).

    CAS  PubMed  Google Scholar 

  21. Buniello, A. et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 47, D1005–d1012 (2019).

    CAS  PubMed  Google Scholar 

  22. Zhao, B. et al. Genome-wide association analysis of 19,629 individuals identifies variants influencing regional brain volumes and refines their genetic co-architecture with cognitive and mental health traits. Nat. Genet. 51, 1637–1644 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhao, B. et al. Large-scale GWAS reveals genetic architecture of brain white matter microstructure and genetic overlap with cognitive and mental health traits (n = 17,706). Mol. Psychiatry 26, 3943–3955 (2021).

    PubMed  Google Scholar 

  24. Seshadri, S. et al. Genetic correlates of brain aging on MRI and cognitive test measures: a genome-wide association and linkage analysis in the Framingham study. BMC Med. Genet. 8, S15 (2007).

    PubMed  PubMed Central  Google Scholar 

  25. Leonardsen, E. H. et al. Genetic architecture of brain age and its causal relations with brain and mental disorders. Mol. Psychiatry 28, 3111–3120 (2023).

    PubMed  PubMed Central  Google Scholar 

  26. Wen, J. et al. The genetic architecture of multimodal human brain age. Nat. Commun. 15, 2604 (2024).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chauhan, G. et al. Association of Alzheimer’s disease GWAS loci with MRI markers of brain aging. Neurobiol. Aging 36, 1765.e1767–1765.e1716 (2015).

    Google Scholar 

  28. Binnewies, J. et al. Lifestyle-related risk factors and their cumulative associations with hippocampal and total grey matter volume across the adult lifespan: a pooled analysis in the European Lifebrain consortium. Brain Res. Bull. 200, 110692 (2023).

    PubMed  Google Scholar 

  29. Fotuhi, M., Do, D. & Jack, C. Modifiable factors that alter the size of the hippocampus with ageing. Nat. Rev. Neurol. 8, 189–202 (2012).

    CAS  PubMed  Google Scholar 

  30. Kapasi, A., DeCarli, C. & Schneider, J. A. Impact of multiple pathologies on the threshold for clinically overt dementia. Acta Neuropathol. 134, 171–186 (2017).

    PubMed  PubMed Central  Google Scholar 

  31. Savva, G. M. et al. Age, neuropathology, and dementia. N. Engl. J. Med. 360, 2302–2309 (2009).

    CAS  PubMed  Google Scholar 

  32. Dong, A. et al. Heterogeneity of neuroanatomical patterns in prodromal Alzheimer’s disease: links to cognition, progression and biomarkers. Brain 140, 735–747 (2017).

    PubMed  Google Scholar 

  33. Young, A. L. et al. Uncovering the heterogeneity and temporal complexity of neurodegenerative diseases with subtype and stage inference. Nat. Commun. 9, 4273 (2018).

    PubMed  PubMed Central  Google Scholar 

  34. Chand, G. B. et al. Two distinct neuroanatomical subtypes of schizophrenia revealed using machine learning. Brain 143, 1027–1038 (2020).

    PubMed  PubMed Central  Google Scholar 

  35. Wen, J. et al. Characterizing heterogeneity in neuroimaging, cognition, clinical symptoms, and genetics among patients with late-life depression. JAMA Psychiatry 79, 464–474 (2022).

    PubMed  PubMed Central  Google Scholar 

  36. Eavani, H. et al. Heterogeneity of structural and functional imaging patterns of advanced brain aging revealed via machine learning methods. Neurobiol. Aging 71, 41–50 (2018).

    PubMed  PubMed Central  Google Scholar 

  37. Habes, M. et al. White matter hyperintensities and imaging patterns of brain ageing in the general population. Brain 139, 1164–1179 (2016).

    PubMed  PubMed Central  Google Scholar 

  38. Skampardoni, I. et al. Genetic and clinical correlates of AI-based brain aging patterns in cognitively unimpaired individuals. JAMA Psychiatry https://doi.org/10.1001/jamapsychiatry.2023.5599 (2024).

    Article  PubMed  Google Scholar 

  39. Moonen, J. E. F. et al. Race, sex, and mid-life changes in brain health: Cardia MRI substudy. Alzheimers Dement. 18, 2428–2437 (2022).

    PubMed  Google Scholar 

  40. Nasrallah, I. M. et al. Association of intensive vs standard blood pressure control with magnetic resonance imaging biomarkers of Alzheimer disease: secondary analysis of the SPRINT MIND randomized trial. JAMA Neurol. 78, 568–577 (2021).

    PubMed  Google Scholar 

  41. Schneider, J. A., Arvanitakis, Z., Bang, W. & Bennett, D. A. Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology 69, 2197–2204 (2007).

    PubMed  Google Scholar 

  42. Pagani, E. et al. Regional brain atrophy evolves differently in patients with multiple sclerosis according to clinical phenotype. Am. J. Neuroradiol. 26, 341–346 (2005).

    PubMed  PubMed Central  Google Scholar 

  43. Cagol, A. et al. Association of brain atrophy with disease progression independent of relapse activity in patients with relapsing multiple sclerosis. JAMA Neurol. 79, 682–692 (2022).

    PubMed  Google Scholar 

  44. Schoonheim, M. M., Broeders, T. A. A. & Geurts, J. J. G. The network collapse in multiple sclerosis: An overview of novel concepts to address disease dynamics. Neuroimage Clin. 35, 103108 (2022).

    PubMed  PubMed Central  Google Scholar 

  45. Lee, C. U. et al. Fusiform gyrus volume reduction in first-episode schizophrenia: a magnetic resonance imaging study. Arch. Gen. Psychiatry 59, 775–781 (2002).

    PubMed  Google Scholar 

  46. Onitsuka, T. et al. Middle and inferior temporal gyrus gray matter volume abnormalities in chronic schizophrenia: an MRI study. Am. J. Psychiatry 161, 1603–1611 (2004).

    PubMed  PubMed Central  Google Scholar 

  47. Tremblay, C. et al. Brain atrophy progression in Parkinson’s disease is shaped by connectivity and local vulnerability. Brain Commun. 3, fcab269 (2021).

    PubMed  PubMed Central  Google Scholar 

  48. Kaur, A. et al. Structural and functional alterations of the temporal lobe in schizophrenia: a literature review. Cureus 12, e11177 (2020).

    PubMed  PubMed Central  Google Scholar 

  49. Gogolla, N. The insular cortex. Curr. Biol. 27, R580–r586 (2017).

    CAS  PubMed  Google Scholar 

  50. Oppenheimer, S. M., Gelb, A., Girvin, J. P. & Hachinski, V. C. Cardiovascular effects of human insular cortex stimulation. Neurology 42, 1727–1732 (1992).

    CAS  PubMed  Google Scholar 

  51. Fink, J. N. et al. Insular cortex infarction in acute middle cerebral artery territory stroke: predictor of stroke severity and vascular lesion. Arch. Neurol. 62, 1081–1085 (2005).

    PubMed  Google Scholar 

  52. Craig, A. D. How do you feel — now? The anterior insula and human awareness. Nat. Rev. Neurosci. 10, 59–70 (2009).

    CAS  PubMed  Google Scholar 

  53. Paulus, M. P. & Stein, M. B. An insular view of anxiety. Biol. Psychiatry 60, 383–387 (2006).

    PubMed  Google Scholar 

  54. Brosch, K. et al. Reduced hippocampal gray matter volume is a common feature of patients with major depression, bipolar disorder, and schizophrenia spectrum disorders. Mol. Psychiatry 27, 4234–4243 (2022).

    PubMed  PubMed Central  Google Scholar 

  55. Alexandros Lalousis, P. et al. Transdiagnostic structural neuroimaging features in depression and psychosis: a systematic review. Neuroimage Clin. 38, 103388 (2023).

    PubMed  PubMed Central  Google Scholar 

  56. Ribe, A. R. et al. Long-term risk of dementia in persons with schizophrenia: a danish population-based cohort study. JAMA Psychiatry 72, 1095–1101 (2015).

    PubMed  Google Scholar 

  57. de Lau, L. M. L., Schipper, C. M. A., Hofman, A., Koudstaal, P. J. & Breteler, M. M. B. Prognosis of Parkinson disease: risk of dementia and mortality: the rotterdam study. Arch. Neurol. 62, 1265–1269 (2005).

    PubMed  Google Scholar 

  58. Alzheimer’s Association. 2016 Alzheimer’s disease facts and figures. Alzheimers Dement. https://doi.org/10.1016/j.jalz.2016.03.001 (2016).

  59. Ten Kate, M. et al. Atrophy subtypes in prodromal Alzheimer’s disease are associated with cognitive decline. Brain 141, 3443–3456 (2018).

    PubMed  PubMed Central  Google Scholar 

  60. Jack, C. R. Jr. et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 14, 535–562 (2018).

    PubMed  Google Scholar 

  61. Schneider, J. A. & Bennett, D. A. Where vascular meets neurodegenerative disease. Stroke 41, S144–S146 (2010).

    PubMed  PubMed Central  Google Scholar 

  62. Smith, A. D. Imaging the progression of Alzheimer pathology through the brain. Proc. Natl Acad. Sci. USA 99, 4135–4137 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Daviet, R. et al. Associations between alcohol consumption and gray and white matter volumes in the UK Biobank. Nat. Commun. 13, 1175 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Topiwala, A., Ebmeier, K. P., Maullin-Sapey, T. & Nichols, T. E. Alcohol consumption and MRI markers of brain structure and function: Cohort study of 25,378 UK Biobank participants. Neuroimage Clin. 35, 103066 (2022).

    PubMed  PubMed Central  Google Scholar 

  65. Karama, S. et al. Cigarette smoking and thinning of the brain’s cortex. Mol. Psychiatry 20, 778–785 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Elbejjani, M. et al. Cigarette smoking and gray matter brain volumes in middle age adults: the CARDIA Brain MRI sub-study. Transl. Psych. https://doi.org/10.1038/s41398-019-0401-1 (2019).

  67. Kang, J. et al. Increased brain volume from higher cereal and lower coffee intake: shared genetic determinants and impacts on cognition and metabolism. Cereb. Cortex 32, 5163–5174 (2022).

    PubMed  PubMed Central  Google Scholar 

  68. de Lange, A. G. et al. Population-based neuroimaging reveals traces of childbirth in the maternal brain. Proc. Natl Acad. Sci. USA 116, 22341–22346 (2019).

    PubMed  PubMed Central  Google Scholar 

  69. Aanes, S., Bjuland, K. J., Skranes, J. & Løhaugen, G. C. Memory function and hippocampal volumes in preterm born very-low-birth-weight (VLBW) young adults. Neuroimage 105, 76–83 (2015).

    PubMed  Google Scholar 

  70. Rodrigues, D. et al. Chronic stress causes striatal disinhibition mediated by SOM-interneurons in male mice. Nat. Commun. 13, 7355 (2022).

    PubMed  PubMed Central  Google Scholar 

  71. Admon, R. et al. Striatal hypersensitivity during stress in remitted individuals with recurrent depression. Biol. Psychiatry 78, 67–76 (2015).

    PubMed  Google Scholar 

  72. Lou, C. et al. Leveraging machine learning predictive biomarkers to augment the statistical power of clinical trials with baseline magnetic resonance imaging. Brain Commun. 3, fcab264 (2021).

    PubMed  PubMed Central  Google Scholar 

  73. Goodfellow, I. et al. Generative adversarial networks. Adv. Neural Inf. Process. Syst. https://doi.org/10.1145/3422622 (2014).

  74. Chen, X. et al. InfoGAN: Interpretable Representation Learning by Information Maximizing Generative Adversarial Nets (NIPS, 2016).

  75. Petersen, R. C. et al. Alzheimer’s Disease Neuroimaging Initiative (ADNI): clinical characterization. Neurology 74, 201–209 (2010).

    PubMed  PubMed Central  Google Scholar 

  76. Miller, K. L. et al. Multimodal population brain imaging in the UK Biobank prospective epidemiological study. Nat. Neurosci. 19, 1523–1536 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Resnick, S. M. et al. One-year age changes in MRI brain volumes in older adults. Cereb. Cortex 10, 464–472 (2000).

    CAS  PubMed  Google Scholar 

  78. Resnick, S. M., Pham, D. L., Kraut, M. A., Zonderman, A. B. & Davatzikos, C. Longitudinal magnetic resonance imaging studies of older adults: a shrinking brain. J. Neurosci. 23, 295–301 (2003).

    Google Scholar 

  79. Ellis, K. A. et al. The Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging: methodology and baseline characteristics of 1112 individuals recruited for a longitudinal study of Alzheimer’s disease. Int. Psychogeriatr. 21, 672–687 (2009).

    PubMed  Google Scholar 

  80. Soldan, A. et al. Relationship of medial temporal lobe atrophy, APOE genotype, and cognitive reserve in preclinical Alzheimer’s disease. Hum. Brain Mapp. 36, 2826–2841 (2015).

    PubMed  PubMed Central  Google Scholar 

  81. LaMontagne, P. J. et al. OASIS-3: longitudinal neuroimaging, clinical, and cognitive dataset for normal aging and Alzheimer disease. Preprint at medRxiv https://doi.org/10.1101/2019.12.13.19014902 (2019).

  82. Johnson, S. C. et al. The Wisconsin registry for Alzheimer’s prevention: a review of findings and current directions. Alzheimers Dement. 10, 130–142 (2018).

    Google Scholar 

  83. Friedman, G. D. et al. CARDIA: study design, recruitment, and some characteristics of the examined subjects. J. Clin. Epidemiol. 41, 1105–1116 (1988).

    CAS  PubMed  Google Scholar 

  84. Völzke, H. et al. Cohort profile: the study of health in Pomerania. Int. J. Epidemiol. 40, 294–307 (2011).

    PubMed  Google Scholar 

  85. Coker, L. H. et al. Postmenopausal hormone therapy and subclinical cerebrovascular disease: the WHIMS-MRI Study. Neurology 72, 125–134 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Sled, J. G., Zijdenbos, A. P. & Evans, A. C. A nonparametric method for automatic correction of intensity nonuniformity in MRI data. IEEE Trans. Med. Imaging 17, 87–97 (1998).

    CAS  PubMed  Google Scholar 

  87. Doshi, J., Erus, G., Ou, Y., Gaonkar, B. & Davatzikos, C. Multi-atlas skull-stripping. Acad. Radiol. 20, 1566–1576 (2013).

    PubMed  Google Scholar 

  88. Doshi, J. et al. MUSE: multi-atlas region segmentation utilizing ensembles of registration algorithms and parameters, and locally optimal atlas selection. NeuroImage 127, 186–195 (2016).

    PubMed  Google Scholar 

  89. Davatzikos, C., Genc, A., Xu, D. & Resnick, S. M. Voxel-based morphometry using the RAVENS maps: methods and validation using simulated longitudinal atrophy. Neuroimage 14, 1361–1369 (2001).

    CAS  PubMed  Google Scholar 

  90. Ou, Y., Sotiras, A., Paragios, N. & Davatzikos, C. DRAMMS: Deformable registration via attribute matching and mutual-saliency weighting. Med. Image Anal. 15, 622–639 (2011).

    PubMed  Google Scholar 

  91. Ronneberger, O., Fischer, P. & Brox, T. U-Net: convolutional networks for biomedical image segmentation. in Medical Image Computing and Computer-Assisted Intervention – MICCAI 2015. (eds Navab, N. et al) 234–241 (Springer, 2015).

  92. Pomponio, R. et al. Harmonization of large MRI datasets for the analysis of brain imaging patterns throughout the lifespan. Neuroimage 208, 116450 (2020).

    PubMed  Google Scholar 

  93. Hansson, O. et al. CSF biomarkers of Alzheimer’s disease concord with amyloid-β PET and predict clinical progression: a study of fully automated immunoassays in BioFINDER and ADNI cohorts. Alzheimers Dement. 14, 1470–1481 (2018).

    PubMed  Google Scholar 

  94. Crane, P. K. et al. Development and assessment of a composite score for memory in the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Brain Imaging Behav. 6, 502–516 (2012).

    PubMed  PubMed Central  Google Scholar 

  95. Gibbons, L. E. et al. A composite score for executive functioning, validated in Alzheimer’s Disease Neuroimaging Initiative (ADNI) participants with baseline mild cognitive impairment. Brain Imaging Behav. 6, 517–527 (2012).

    PubMed  PubMed Central  Google Scholar 

  96. Choi, S. E. et al. Development and validation of language and visuospatial composite scores in ADNI. Alzheimers Dement. 6, e12072 (2020).

    Google Scholar 

  97. Abraham, A. et al. Machine learning for neuroimaging with scikit-learn. Front. Neuroinform. 8, 14 (2014).

    PubMed  PubMed Central  Google Scholar 

  98. Manichaikul, A. et al. Robust relationship inference in genome-wide association studies. Bioinformatics 26, 2867–2873 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Price, A. L., Zaitlen, N. A., Reich, D. & Patterson, N. New approaches to population stratification in genome-wide association studies. Nat. Rev. Genet. 11, 459–463 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Abraham, G., Qiu, Y. & Inouye, M. FlashPCA2: principal component analysis of Biobank-scale genotype datasets. Bioinformatics 33, 2776–2778 (2017).

    CAS  PubMed  Google Scholar 

  101. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Watanabe, K., Taskesen, E., van Bochoven, A. & Posthuma, D. Functional mapping and annotation of genetic associations with FUMA. Nat. Commun. 8, 1826 (2017).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Data used in this study are part of the iSTAGING study (representative, C. Davatzikos), the Preclinical AD Consortium (M. S. Albert), ADNI (M. W. Weiner) and the BLSA (S. M. Resnick). The iSTAGING consortium is a multi-institutional effort funded by the National Institute on Aging (NIA) by RF1 AG054409. The BLSA neuroimaging study is funded by the Intramural Research Program, NIA, National Institutes of Health (NIH) and by HHSN271201600059C. The BIOCARD study is in part supported by NIH grant U19-AG033655. SHIP is part of the Community Medicine Research net of the University of Greifswald, which is funded by the Federal Ministry of Education and Research (grant nos. 01ZZ9603, 01ZZ0103 and 01ZZ0403), the Ministry of Cultural Affairs and the Social Ministry of the Federal State of Mecklenburg-West Pomerania. MRI scans in SHIP-START and SHIP-TREND have been supported by a joint grant from Siemens Healthineers and the Federal State of Mecklenburg-West Pomerania. The Women’s Health Initiative was funded by the National Heart, Lung and Blood Institute of the NIH, US Department of Health and Human Services. Contracts HHSN268200464221C and N01-WH-4-4221 provided additional support. The WHIMS (M.A.E.) was funded in part by Wyeth Pharmaceuticals. The HANDLS study is supported by the NIA Intramural Research Program, NIH, Project ZIA-AG000513. The HANDLS Scan substudy is supported by NIH grants 1RO1AG034161, 2P30AG028747-14S1 and 1R56AG064088-01A1. The HABS-HD project is funded by grants from the NIA: R01AG054073 and R01AG058533. HABS-HD multiple principal investigators were S. E. O’Bryant, K. Yaffe, A. Toga, R. Rissman and L. Johnson; and the HABS-HD investigators were M. Braskie, K. King, J. R Hall, M. Petersen, R. Palmer, R. Barber, Y. Shi, F. Zhang, R. Nandy, R. McColl, D. Mason, B. Christian, N. Philips, S. Large, J. Lee, B. Vardarajan, M. Rivera Mindt, A. Cheema, L. Barnes, M. Mapstone, A. Cohen, A. Kind, O. Okonkwo, R. Vintimilla, Z. Zhou, M. Donohue, R. Raman, M. Borzage, M. Mielke, B. Ances, G. Babulal, J. Llibre-Guerra, C. Hill and R. Vig. Other supporting grants include 5U01AG068057-02 and 1U24AG074855-01. S.R.H. and J.C.M. are supported by multiple grants and contracts from NIH. A.A. was supported by grants 191026 and 206795 from the Swiss National Science Foundation. R.T.S. was supported by grants R01MH123550 and R01MH112847. J.C.M. was supported by NIH ACS grant, P01AG026276. Y.F. was supported by grant R01AG066650. M.H. was supported by the National Institute of Health (NIH) grant number 1R01AG080821. This research has been conducted using the UKBB Resource under application no. 35148. Data used in the preparation of this article were in part obtained from the ADNI database (adni.loni.usc.edu). As such, the investigators within the ADNI contributed to the design and implementation of ADNI and/or provided data but did not participate in the analysis or writing of this report. A complete listing of ADNI investigators can be found at http://adni.loni.usc.edu/wpcontent/uploads/how_to_apply/ADNI_Acknowledgement_List.pdf. ADNI is funded by the NIA, the National Institute of Biomedical Imaging and Bioengineering and through generous contributions from the following: AbbVie, Alzheimer’s Association; Alzheimer’s Drug Discovery Foundation; Araclon Biotech; BioClinica; Biogen; Bristol-Myers Squibb; CereSpir; Cogstate; Eisai; Elan Pharmaceuticals; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche and its affiliated company Genentech; Fujirebio; GE Healthcare; IXICO; Janssen Alzheimer Immunotherapy Research & Development; Johnson & Johnson Pharmaceutical Research & Development; Lumosity; Lundbeck; Merck & Co; Meso Scale Diagnostics; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer; Piramal Imaging; Servier; Takeda Pharmaceutical Company; and Transition Therapeutics. The Canadian Institutes of Health Research is providing funds to support ADNI clinical sites in Canada. Private sector contributions are facilitated by the Foundation for the National Institutes of Health (www.fnih.org). The grantee organization is the Northern California Institute for Research and Education, and the study is coordinated by the Alzheimer’s Therapeutic Research Institute at the University of Southern California. ADNI data are disseminated by the Laboratory for Neuro Imaging at the University of Southern California. Z.Y. had full access to all the data in the study and is responsible for the integrity of the data and the accuracy of the data analysis.

Author information

Authors and Affiliations

Authors

Contributions

Study concept and design was by Z.Y. and C.D. Model development was by Z.Y. Data interpretation was by Z.Y., J.W., I.M.N., P.A.L., N.K., S.M.R., H.S. and C.D. Drafting of the manuscript was by Z.Y., J.W., I.M.N. and C.D. Statistical analysis was by Z.Y. Genetic analysis was by J.W. Data collection and processing was by Z.Y., J.W., G.E., S.T.G., R.M., E.M., Y.C., D.S., A.A., P.P., K.W., H.J.G., R.B., S.F., D.T., M.B., Y.A., D.Y., D.S.M., P.L., T.L.S.B., S.R.H., T.R.A., S.R.W., M.K.E., A.B.Z., L.J.L., A.S., M.A.E., C.L.M., P.M., J.F., A.W.T., S.O.B., M.M.C., S.V., S.C.J., J.C.M., M.S.A., K.Y., H.V., L.F., R.N.B., D.A.W., S.M.R. and C.D. Critical revision of the manuscript for important intellectual content was by Z.Y., J.W., G.E., S.T.G., R.M., E.M., Y.C., D.S., A.A., P.P., K.W., H.J.G., R.B., S.F., D.T., M.B., Y.A., D.Y., D.S.M., P.L., T.L.S.B., S.R.H., T.R.A., S.R.W., M.K.E., A.B.Z., L.J.L., A.S., M.A.E., C.L.M., P.M., J.F., A.W.T., S.O.B., M.M.C., S.V., S.C.J., J.C.M., M.S.A., K.Y., H.V., L.F., R.N.B., R.T.S., Y.F., M.H., P.A.L., N.K., D.A.W., S.M.R., H.S., I.M.N. and C.D.

Corresponding author

Correspondence to Christos Davatzikos.

Ethics declarations

Competing interests

H.J.G. has received travel grants and speaker’s honoraria from Fresenius Medical Care, Neuraxpharm, Servier and Janssen Cilag as well as research funding from Fresenius Medical Care. R.T.S. received consulting income from Octave Bioscience and has received compensation for scientific reviewing from the American Medical Association. T.L.S.B. has received investigator-initiated research funding from the NIH, the Alzheimer’s Association, the Foundation at Barnes-Jewish Hospital, Siemens Healthineers and Avid Radiopharmaceuticals (a wholly owned subsidiary of Eli Lilly and Company). She participates as a site investigator in clinical trials sponsored by Eli Lilly and Company, Biogen, Eisai, Jaansen and Roche. She has served as a paid and unpaid consultant to Eisai, Siemens, Biogen, Janssen and Bristol-Myers Squibb. The other authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks Cristina Granziera and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Lorenzo Righetto, in collaboration with the Nature Medicine team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Figs. 1–3 and Supplementary Tables 1–3.

Reporting summary

Supplementary Data 1–11

Supplementary data, including detailed description of data and statistical test results.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, Z., Wen, J., Erus, G. et al. Brain aging patterns in a large and diverse cohort of 49,482 individuals. Nat Med (2024). https://doi.org/10.1038/s41591-024-03144-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41591-024-03144-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing