www.fgks.org   »   [go: up one dir, main page]

Skip to main content

Significance

The phenotypic age of the human brain, as revealed via deep learning of anatomic magnetic resonance images, reflects patterns of structural change related to cognitive decline. Our interpretable deep learning estimates that the brain ages more accurately than any other approaches to date. Furthermore, compared to chronological age, our inferred brain ages are significantly more strongly associated with early signs of Alzheimer’s disease. Maps conveying the importance of each brain region for estimating brain age reveal differences in patterns of neurological aging between males and females and between persons with and without cognitive impairment. These findings provide insight into early identification of persons at high risk of Alzheimer’s disease.

Abstract

The gap between chronological age (CA) and biological brain age, as estimated from magnetic resonance images (MRIs), reflects how individual patterns of neuroanatomic aging deviate from their typical trajectories. MRI-derived brain age (BA) estimates are often obtained using deep learning models that may perform relatively poorly on new data or that lack neuroanatomic interpretability. This study introduces a convolutional neural network (CNN) to estimate BA after training on the MRIs of 4,681 cognitively normal (CN) participants and testing on 1,170 CN participants from an independent sample. BA estimation errors are notably lower than those of previous studies. At both individual and cohort levels, the CNN provides detailed anatomic maps of brain aging patterns that reveal sex dimorphisms and neurocognitive trajectories in adults with mild cognitive impairment (MCI, N = 351) and Alzheimer’s disease (AD, N = 359). In individuals with MCI (54% of whom were diagnosed with dementia within 10.9 y from MRI acquisition), BA is significantly better than CA in capturing dementia symptom severity, functional disability, and executive function. Profiles of sex dimorphism and lateralization in brain aging also map onto patterns of neuroanatomic change that reflect cognitive decline. Significant associations between BA and neurocognitive measures suggest that the proposed framework can map, systematically, the relationship between aging-related neuroanatomy changes in CN individuals and in participants with MCI or AD. Early identification of such neuroanatomy changes can help to screen individuals according to their AD risk.

Get full access to this article

Purchase, subscribe or recommend this article to your librarian.

Data, Materials, and Software Availability

3D-CNN software is available from https://github.com/irimia-laboratory/USC_BA_estimator MRI data are publicly available from ADNI (https://adni.loni.usc.edu/), UKBB (https://www.ukbiobank.ac.uk/), CamCAN (https://www.cam-can.org/), and HCP (https://www.humanconnectome.org/). There are no relevant accession codes required to access these data, and the authors had no special access privileges that others would not have to the data obtained from any of these databases. Data used in the preparation of this article were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database (https://adni.loni.usc.edu/). The ADNI was launched in 2003 as a public–private partnership, led by Principal Investigator Michael W. Weiner, MD. The primary goal of ADNI has been to test whether serial MRI, positron emission tomography (PET), other biological markers, and clinical and neuropsychological assessment can be combined to measure the progression of mild cognitive impairment (MCI) and early Alzheimer’s disease (AD).

Acknowledgments

A.I. is grateful to Kenneth H. Buetow, Caleb E. Finch, Margaret Gatz, and Mara Mather for useful discussions. The authors acknowledge Alexander S. Maher for assistance with data retrieval. P.E., A.A., N.F.C., R.J.M., N.N.C., and A.I. gratefully acknowledge the support from the (NIH) under grant R01 NS 100973, the US Department of Defense (DoD) under award W81-XWH-1810413, the James J. & Sue Femino Foundation, a Hanson-Thorell Research Scholarship, the Undergraduate Research Associate Program (URAP), and the Center for Undergraduate Research in Viterbi Engineering (CURVE) at the University of Southern California. C.Y., M.C., X.C., and P.B. gratefully acknowledge the support from the NSF under CAREER award CPS/CNS-1453860 and grants CCF-1837131, MCB-1936775, CNS-1932620, and CMMI-1936624, from the Okawa Foundation, from the Defense Advanced Research Projects Agency (DARPA) under a Young Faculty Award and a Director Award under grant N66001-17-1-4044, from a 2021 USC Stevens Center Technology Advancement Grant (TAG) award, from an Intel Faculty Award and from a Northrop Grumman grant. P.M.T. gratefully acknowledges the support from the NIH under grants R01 AG 058854, U01 AG 068057, R01 AG 060610, and P41 EB 015922. Data collection and sharing for this project was funded by the Alzheimer’s Disease Neuroimaging Initiative (ADNI) through NIH grant U01 AG 024904 and DoD ADNI through DoD award number W81-XWH-1220012. ADNI is funded by the National Institute on Aging, the National Institute of Biomedical Imaging and Bioengineering, and generous contributions from the following: AbbVie, Alzheimer’s Association; Alzheimer’s Drug Discovery Foundation; Araclon Biotech; BioClinica, Inc.; Biogen; Bristol-Myers Squibb Company; CereSpir, Inc.; Cogstate; Eisai Inc.; Elan Pharmaceuticals, Inc.; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche Ltd and its affiliated company Genentech, Inc.; Fujirebio; GE Healthcare; IXICO Ltd.; Janssen Alzheimer Immunotherapy Research & Development, LLC.; Johnson & Johnson Pharmaceutical Research & Development, LLC.; Lumosity; Lundbeck; Merck & Co., Inc.; Meso Scale Diagnostics, LLC.; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer Inc.; 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 NIH (http://www.fnih.org). The ADNI 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. Data for this study were provided, in part, by the Human Connectome Project, WUMinn Consortium (Principal Investigators: David Van Essen and Kamil Ugurbil; 1 U54 MH 091657) funded by the 16 NIH Institutes and Centers that support the NIH Blueprint for Neuroscience Research and by the McDonnell Center for Systems Neuroscience at Washington University. Research reported in this publication was also supported by the National Institute on Aging of the NIH under award U01 AG 052564. CamCAN funding was provided by the UK Biotechnology and Biological Sciences Research Council (grant number BB/H008217/1), together with support from the UK Medical Research Council and the University of Cambridge, UK. This research has been conducted using the UK Biobank Resource, under application numbers 11559 and 47656. The views, opinions, and/or findings contained in this article are those of the authors and should not be interpreted as representing the official views or policies, either expressed or implied by the NIH, DARPA, DoD, NSF, or any other entity acknowledged here. The funding sources had no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; and in the decision to submit the article for publication.

Author contributions

P.B. and A.I. designed research; C.Y. and A.A. performed research; C.Y., M.C., and A.A. contributed new reagents/analytic tools; C.Y., P.I., M.C., A.A., N.F.C., R.J.M., N.N.C., and X.C. analyzed data; A.N. data used in research was sourced from the ADNI repository; and C.Y., P.I., A.A., N.F.C., P.M.T., P.B., and A.I. wrote the paper.

Competing interest

The authors have research support to disclose, P.M.T. discloses research grant support from Biogen, Inc. for work unrelated to this study.

Supporting Information

Appendix 01 (PDF)
Dataset S01 (DOCX)

References

1
A. Amgalan et al., Brain age estimation reveals older adults’ accelerated senescence after traumatic brain injury. GeroScience pages 1–17 (2022).
2
C. Lopez-Otin, M. A. Blasco, L. Partridge, M. Serrano, G. Kroemer, The hallmarks of aging. Cell 153, 1194–1217 (2013).
3
A. Irimia, C. M. Torgerson, S.-Y. Matthew Goh, J. D. Van Horn, Statistical estimation of physiological brain age as a descriptor of senescence rate during adulthood. Brain Imaging Behav. 9, 678–689 (2015).
4
I. Beheshti, S. Nugent, O. Potvin, S. Duchesne, Bias-adjustment in neuroimaging-based brain age frameworks: A robust scheme. NeuroImage: Clin. 24, 102063 (2019).
5
J. H. Cole et al., Brain age predicts mortality. Mol. Psychiatry 23, 1385–1392 (2018a).
6
R. J. Massett et al., Regional neuroanatomic effects on brain age inferred using magnetic resonance imaging and ridge regression. J. Gerontol.: Series A (2022).
7
K. Ning et al., Improving brain age estimates with deep learning leads to identification of novel genetic factors associated with brain aging. Neurobiol. Aging 105, 199–204 (2021).
8
N. Opel et al., Brain structural abnormalities in obesity: Relation to age, genetic risk, and common psychiatric disorders. Mol. Psychiatry 26, 4839–4852 (2021).
9
J. H. Cole et al., Increased brain-predicted aging in treated HIV disease. Neurology 88, 1349–1357 (2017a).
10
J. H. Cole et al., No evidence for accelerated aging-related brain pathology in treated human immunodeficiency virus: Longitudinal neuroimaging results from the comorbidity in relation to AIDS (COBRA) project. Clin. Infect. Dis. 66, 1899–1909 (2018b).
11
A. Irimia, Cross-sectional volumes and trajectories of the human brain, gray matter, white matter and cerebrospinal fluid in 9473 typically aging adults. Neuroinf. 19, 347–366 (2021).
12
A. Irimia, A. S. Maher, N. N. Chaudhari, N. F. Chowdhury, E. B. Jacobs, Acute cognitive deficits after traumatic brain injury predict Alzheimer’s disease-like degradation of the human default mode network. Gerosci. 42, 1411–1429 (2020).
13
A. Salih et al., Brain age estimation at tract group level and its association with daily life measures, cardiac risk factors and genetic variants. Sci. Rep. 11, 1–14 (2021).
14
W. Y. Isaac Tseng, Y. Chin Hsu, Te. Wei Kao, Brain age difference at baseline predicts clinical dementia rating change in approximately two years. J. Alzheimer’s Dis. 1–15 (2022).
15
P. Imms, H. C. Chui, A. Irimia, Alzheimer’s disease after mild traumatic brain injury. Aging 14, 5292 (2022).
16
J. H. Cole et al., Predicting brain age with deep learning from raw imaging data results in a reliable and heritable biomarker. NeuroImage 163, 115–124 (2017b).
17
H. Jiang et al., Predicting brain age of healthy adults based on structural MRI parcellation using convolutional neural networks. Front. Neurol. 10, 1346 (2020).
18
C. Rudin, Stop explaining black box machine learning models for high stakes decisions and use interpretable models instead. Nat. Mach. Intell. 1, 206–215 (2019).
19
L. Fisch et al., Predicting chronological age from structural neuroimaging: The predictive analytics competition 2019. Front. Psychiatry 12 (2021).
20
H. Peng, W. Gong, C. F. Beckmann, A. Vedaldi, S. M. Smith, Accurate brain age prediction with lightweight deep neural networks. Med. Image Anal. 68, 101871 (2021).
21
W. Gong, C. F. Beckmann, A. Vedaldi, S. M. Smith, H. Peng, Optimising a simple fully convolutional network for accurate brain age prediction in the PAC 2019 challenge. Front. Psychiatry 12 (2021).
22
S. Borson et al., Improving dementia care: The role of screening and detection of cognitive impairment. Alzheimers Dementia 9, 151–159 (2013).
23
C. Marcus, E. Mena, R. M. Subramaniam, Brain PET in the diagnosis of Alzheimer’s disease. Clin. Nucl. Med. 39, e413 (2014).
24
J. José Vaquero, P. Kinahan, Positron emission tomography: Current challenges and opportunities for technological advances in clinical and preclinical imaging systems. Ann. Rev. Biomed. Eng. 17, 385–414 (2015).
25
J. Wrigglesworth et al., Brain-predicted age difference is associated with cognitive processing in later-life. Neurobiol. Aging 109, 195–203 (2022).
26
L. E. Korthauer et al., Brain-behavior investigation of potential cognitive markers of Alzheimer’s disease in middle age: A multi-modal imaging study. Brain Imaging Behav. pages 1–8 (2021).
27
J. Wang et al., Gray matter age prediction as a biomarker for risk of dementia. Proc. Natl. Acad. Sci. 116, 21213–21218 (2019).
28
F. Biondo et al., Brain-age is associated with progression to dementia in memory clinic patients. NeuroImage: Clin. 36, 103175 (2022).
29
C. Gaser et al., Brainage in mild cognitive impaired patients: predicting the conversion to alzheimer’s disease. PloS one 8, e67346 (2013).
30
R. C. Petersen et al., Alzheimer’s disease neuroimaging initiative (ADNI): Clinical characterization. Neurology 74, 201–209 (2010).
31
J. H. Cole, R. E. Marioni, S. E. Harris, I. J. Deary, Brain age and other bodily : Implications for neuropsychiatry. Mol. Psychiatry 24, 266–281 (2019).
32
D. Brennan, T. Wu, J. Fan, Morphometrical brain markers of sex difference. Cereb. Cortex 31, 3641–3649 (2021).
33
L. Kogler et al., Sex differences in the functional connectivity of the amygdalae in association with cortisol. NeuroImage 134, 410–423 (2016).
34
A. Veronica Witte, M. Savli, A. Holik, S. Kasper, R. Lanzenberger, Regional sex differences in grey matter volume are associated with sex hormones in the young adult human brain. NeuroImage 49, 1205–1212 (2010).
35
G. F. Wooten, L. J. Currie, V. E. Bovbjerg, J. K. Lee, J. Patrie, Are men at greater risk for Parkinson’s disease than women?J. Neurol., Neuros. Psychiatry 75, 637–639 (2004).
36
T. Liu et al., The effects of age and sex on cortical sulci in the elderly. NeuroImage 51, 19–27 (2010).
37
J. A. Kaye, C. DeCarli, J. S. Luxenberg, S. I. Rapoport, The significance of age-related enlargement of the cerebral ventricles in healthy men and women measured by quantitative computed X-ray tomography. J. Am. Geriatr. Soc. 40, 225–231 (1992).
38
M. Hirnstein, K. Hugdahl, M. Hausmann, Cognitive sex differences and hemispheric asymmetry: A critical review of 40 years of research. Laterality: Asymmetries of Body, Brain and Cognition 24, 204–252 (2019).
39
R. Cabeza, Hemispheric asymmetry reduction in older adults: The HAROLD model. Psychol. Aging 17, 85 (2002).
40
S. Liu, J. Seidlitz, J. D. Blumenthal, L. S. Clasen, A. Raznahan, Integrative structural, functional, and transcriptomic analyses of sex-biased brain organization in humans. Proc. Natl. Acad. Sci. 117, 18788–18798 (2020).
41
E. R. Sowell et al., Sex differences in cortical thickness mapped in 176 healthy individuals between 7 and 87 years of age. Cereb. Cortex 17, 1550–1560 (2007).
42
M. W. Bondi et al., Neuropsychological criteria for mild cognitive impairment improves diagnostic precision, biomarker associations, and progression rates. J. Alzheimer’s Dis. 42, 275–289 (2014).
43
L. Christine Löwe, C. Gaser, K. Franke, Alzheimer’s Disease Neuroimaging Initiative, The effect of the ApoE genotype on individual BrainAGE in normal aging, mild cognitive impairment, and Alzheimer’s disease. PloS One 11, e0157514 (2016).
44
H. I. L. Jacobs, M. P. J. Van Boxtel, J. Jolles, F. R. J. Verhey, H. B. M. Uylings, Parietal cortex matters in Alzheimer’s disease: An overview of structural, functional and metabolic findings. Neurosci. Biobehav. Rev. 36, 297–309 (2012).
45
A. M. Fjell et al., What is normal in normal aging? Effects of aging, amyloid and Alzheimer’s disease on the cerebral cortex and the hippocampusProgr. Neurobiol. 117, 20–40 (2014).
46
G. S. Day et al., Tau PET binding distinguishes patients with early-stage posterior cortical atrophy from amnestic Alzheimer’s disease dementia. Alzheimer Dis. Ass. Dis. 31, 87 (2017).
47
H. Braak, E. V. A. Braak, Staging of Alzheimer’s disease-related neurofibrillary changes. Neurobiol. Aging 16, 271–278 (1995).
48
B. Couvy-Duchesne et al., Ensemble learning of convolutional neural network, support vector machine, and best linear unbiased predictor for brain age prediction: Aramis contribution to the predictive analytics competition 2019 challenge. Front. Psychiatry page 1451 (2020).
49
C. Yuan Kuo et al., Improving individual brain age prediction using an ensemble deep learning framework. Front. Psychiatry 12 (2021).
50
P. Besson, T. Parrish, A. K. Katsaggelos, S. Kathleen Bandt, Geometric deep learning on brain shape predicts sex and age. Comput. Med. Imaging Graphics 91, 101939 (2021).
51
F. Liem et al., Predicting brain-age from multimodal imaging data captures cognitive impairment. Neuroimage 148, 179–188 (2017).
52
J. Lee et al., Deep learning-based brain age prediction in normal aging and dementia. Nature Aging https://doi.org/10.1038/s43587-022-00219-7 (2022).
53
E. H. Leonardsen et al., Deep neural networks learn general and clinically relevant representations of the ageing brain. NeuroImage 256, 119210. (2022).
54
R. Li, M. Singh, Sex differences in cognitive impairment and Alzheimer’s disease. Front. Neuroendocrinol. 35, 385–403 (2014).
55
S. He, P. Ellen Grant, Y. Ou, Global-local transformer for brain age estimation. IEEE Trans. Med. Imaging 41, 213–224 (2021).
56
D. A. Wood et al., Accurate brain-age models for routine clinical mri examinations. NeuroImage 249, 118871 (2022).
57
H. Lee et al., Estimating and accounting for the effect of MRI scanner changes on longitudinal whole-brain volume change measurements. NeuroImage 184, 555–565 (2019).
58
J. Stine Elam et al., The Human Connectome Project: A retrospective. NeuroImage 244, 118543 (2021).
59
D. C. Van Essen et al., The Human Connectome Project: A data acquisition perspective. NeuroImage 62, 2222–2231 (2012).
60
F. Alfaro-Almagro et al., Image processing and quality control for the first 10,000 brain imaging datasets from UK Biobank. NeuroImage 166, 400–424 (2018).
61
J. R. Taylor et al., The Cambridge Centre for Ageing and Neuroscience (Cam-CAN) data repository: Structural and functional MRI, MEG, and cognitive data from a cross-sectional adult lifespan sample. NeuroImage 144, 262–269 (2017).
62
L. Quitania Park et al., Confirmatory factor analysis of the ADNI neuropsychological battery. Brain Imaging Behav. 6, 528–539 (2012).
63
B. Fischl, Freesurfer. NeuroImage 62, 774–781 (2012).
64
Z. Zhang, Improved Adam optimizer for deep neural networks. In 2018 IEEE/ACM 26th International Symposium on Quality of Service (IWQoS) pages 1–2. IEEE (2018).
65
T. T. Le et al., A nonlinear simulation framework supports adjusting for age when analyzing BrainAGE. Front. Aging Neurosci. page 317 (2018).
66
M. S. Treder et al., Correlation constraints for regression models: Controlling bias in brain age prediction. Front. Psychiatry page 25 (2021).
67
K. Kamnitsas et al., Efficient multi-scale 3d cnn with fully connected crf for accurate brain lesion segmentation. Med. image Anal. 36, 61–78 (2017).
68
P. Baldi, P. J. Sadowski, Understanding dropout. Adv. Neural Inf. Processing Sys. 26 (2013).
69
Y. Benjamini, Y. Hochberg, Controlling the false discovery rate: A practical and powerful approach to multiple testing. J. R. Stat. Soc. Series B (Methodol.) 57, 289–300 (1995). http://www.jstor.org/stable/2346101.
70
K. Simonyan, A. Vedaldi, A. Zisserman, Deep inside convolutional networks: Visualising image classification models and saliency maps. arXiv preprint arXiv:1312.6034, (2013).
71
M. F. Glasser, D. C. Van Essen, Mapping human cortical areas in vivo based on myelin content as revealed by t1- and t2-weighted mri. J. Neurosci. pages 11597–11616.
72
S. O. Mahoney, N. F. Chowdhury, V. Ngo, A. Irimia, Mild traumatic brain injury results in significant and lasting cortical demyelination. Front. Neurol. 13, 854396 (2022).
73
W. Zhang, L. Zhan, P. Thompson, Y. Wang, Deep representation learning for multimodal brain networks in International Conference on Medical Image Computing and Computer-Assisted Intervention. (Springer, pp. 613–624. 2020).

Information & Authors

Information

Published in

Go to Proceedings of the National Academy of Sciences
Go to Proceedings of the National Academy of Sciences
Proceedings of the National Academy of Sciences
Vol. 120 | No. 2
January 10, 2023
PubMed: 36595679

Classifications

Data, Materials, and Software Availability

3D-CNN software is available from https://github.com/irimia-laboratory/USC_BA_estimator MRI data are publicly available from ADNI (https://adni.loni.usc.edu/), UKBB (https://www.ukbiobank.ac.uk/), CamCAN (https://www.cam-can.org/), and HCP (https://www.humanconnectome.org/). There are no relevant accession codes required to access these data, and the authors had no special access privileges that others would not have to the data obtained from any of these databases. Data used in the preparation of this article were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database (https://adni.loni.usc.edu/). The ADNI was launched in 2003 as a public–private partnership, led by Principal Investigator Michael W. Weiner, MD. The primary goal of ADNI has been to test whether serial MRI, positron emission tomography (PET), other biological markers, and clinical and neuropsychological assessment can be combined to measure the progression of mild cognitive impairment (MCI) and early Alzheimer’s disease (AD).

Submission history

Received: August 26, 2022
Accepted: November 10, 2022
Published online: January 3, 2023
Published in issue: January 10, 2023

Keywords

  1. brain age
  2. cognitive impairment
  3. Alzheimer’s disease
  4. deep learning

Acknowledgments

A.I. is grateful to Kenneth H. Buetow, Caleb E. Finch, Margaret Gatz, and Mara Mather for useful discussions. The authors acknowledge Alexander S. Maher for assistance with data retrieval. P.E., A.A., N.F.C., R.J.M., N.N.C., and A.I. gratefully acknowledge the support from the (NIH) under grant R01 NS 100973, the US Department of Defense (DoD) under award W81-XWH-1810413, the James J. & Sue Femino Foundation, a Hanson-Thorell Research Scholarship, the Undergraduate Research Associate Program (URAP), and the Center for Undergraduate Research in Viterbi Engineering (CURVE) at the University of Southern California. C.Y., M.C., X.C., and P.B. gratefully acknowledge the support from the NSF under CAREER award CPS/CNS-1453860 and grants CCF-1837131, MCB-1936775, CNS-1932620, and CMMI-1936624, from the Okawa Foundation, from the Defense Advanced Research Projects Agency (DARPA) under a Young Faculty Award and a Director Award under grant N66001-17-1-4044, from a 2021 USC Stevens Center Technology Advancement Grant (TAG) award, from an Intel Faculty Award and from a Northrop Grumman grant. P.M.T. gratefully acknowledges the support from the NIH under grants R01 AG 058854, U01 AG 068057, R01 AG 060610, and P41 EB 015922. Data collection and sharing for this project was funded by the Alzheimer’s Disease Neuroimaging Initiative (ADNI) through NIH grant U01 AG 024904 and DoD ADNI through DoD award number W81-XWH-1220012. ADNI is funded by the National Institute on Aging, the National Institute of Biomedical Imaging and Bioengineering, and generous contributions from the following: AbbVie, Alzheimer’s Association; Alzheimer’s Drug Discovery Foundation; Araclon Biotech; BioClinica, Inc.; Biogen; Bristol-Myers Squibb Company; CereSpir, Inc.; Cogstate; Eisai Inc.; Elan Pharmaceuticals, Inc.; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche Ltd and its affiliated company Genentech, Inc.; Fujirebio; GE Healthcare; IXICO Ltd.; Janssen Alzheimer Immunotherapy Research & Development, LLC.; Johnson & Johnson Pharmaceutical Research & Development, LLC.; Lumosity; Lundbeck; Merck & Co., Inc.; Meso Scale Diagnostics, LLC.; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer Inc.; 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 NIH (http://www.fnih.org). The ADNI 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. Data for this study were provided, in part, by the Human Connectome Project, WUMinn Consortium (Principal Investigators: David Van Essen and Kamil Ugurbil; 1 U54 MH 091657) funded by the 16 NIH Institutes and Centers that support the NIH Blueprint for Neuroscience Research and by the McDonnell Center for Systems Neuroscience at Washington University. Research reported in this publication was also supported by the National Institute on Aging of the NIH under award U01 AG 052564. CamCAN funding was provided by the UK Biotechnology and Biological Sciences Research Council (grant number BB/H008217/1), together with support from the UK Medical Research Council and the University of Cambridge, UK. This research has been conducted using the UK Biobank Resource, under application numbers 11559 and 47656. The views, opinions, and/or findings contained in this article are those of the authors and should not be interpreted as representing the official views or policies, either expressed or implied by the NIH, DARPA, DoD, NSF, or any other entity acknowledged here. The funding sources had no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; and in the decision to submit the article for publication.
Author Contributions
P.B. and A.I. designed research; C.Y. and A.A. performed research; C.Y., M.C., and A.A. contributed new reagents/analytic tools; C.Y., P.I., M.C., A.A., N.F.C., R.J.M., N.N.C., and X.C. analyzed data; A.N. data used in research was sourced from the ADNI repository; and C.Y., P.I., A.A., N.F.C., P.M.T., P.B., and A.I. wrote the paper.
Competing Interest
The authors have research support to disclose, P.M.T. discloses research grant support from Biogen, Inc. for work unrelated to this study.

Notes

This article is a PNAS Direct Submission.

Authors

Affiliations

Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
Roy J. Massett
Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
Corwin D. Denney Research Center, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
Corwin D. Denney Research Center, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
Imaging Genetics Center, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90033
Department of Quantitative & Computational Biology, Dana & David Dornsife College of Arts & Sciences, University of Southern California, Los Angeles, CA 90089
Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
Department of Psychiatry, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
Department of Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
Corwin D. Denney Research Center, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
Department of Quantitative & Computational Biology, Dana & David Dornsife College of Arts & Sciences, University of Southern California, Los Angeles, CA 90089
the Alzheimer’s Disease Neuroimaging Initiative3

Notes

2To whom correspondence may be addressed. Email: [email protected].
1C.Y., P.I., M.C., A.A., N.F.C., P.B., and A.I. contributed equally to this work.
3A complete list of the Alzheimer’s Disease Neuroimaging Initiative can be found in the SI Appendix.

Metrics & Citations

Metrics

Note: The article usage is presented with a three- to four-day delay and will update daily once available. Due to ths delay, usage data will not appear immediately following publication. Citation information is sourced from Crossref Cited-by service.


Citation statements

Altmetrics

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

View Options

Get Access

Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login

Recommend to a librarian

Recommend PNAS to a Librarian

Purchase options

Purchase this article to get full access to it.

Single Article Purchase

Anatomically interpretable deep learning of brain age captures domain-specific cognitive impairment
Proceedings of the National Academy of Sciences
  • Vol. 120
  • No. 2

View options

PDF format

Download this article as a PDF file

DOWNLOAD PDF

Media

Figures

Tables

Other

Share

Share

Share article link

Share on social media