1
|
Oh K, Heo DW, Mulyadi AW, Jung W, Kang E, Lee KH, Suk HI. A quantitatively interpretable model for Alzheimer's disease prediction using deep counterfactuals. Neuroimage 2025; 309:121077. [PMID: 39954872 DOI: 10.1016/j.neuroimage.2025.121077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/19/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
Deep learning (DL) for predicting Alzheimer's disease (AD) has provided timely intervention in disease progression yet still demands attentive interpretability to explain how their DL models make definitive decisions. Counterfactual reasoning has recently gained increasing attention in medical research because of its ability to provide a refined visual explanatory map. However, such visual explanatory maps based on visual inspection alone are insufficient unless we intuitively demonstrate their medical or neuroscientific validity via quantitative features. In this study, we synthesize the counterfactual-labeled structural MRIs using our proposed framework and transform it into a gray matter density map to measure its volumetric changes over the parcellated region of interest (ROI). We also devised a lightweight linear classifier to boost the effectiveness of constructed ROIs, promoted quantitative interpretation, and achieved comparable predictive performance to DL methods. Throughout this, our framework produces an "AD-relatedness index" for each ROI. It offers an intuitive understanding of brain status for an individual patient and across patient groups concerning AD progression.
Collapse
Affiliation(s)
- Kwanseok Oh
- Department of Artificial Intelligence, Korea University, Seoul 02841, Republic of Korea
| | - Da-Woon Heo
- Department of Artificial Intelligence, Korea University, Seoul 02841, Republic of Korea
| | - Ahmad Wisnu Mulyadi
- Department of Brain and Cognitive Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Wonsik Jung
- Department of Brain and Cognitive Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Eunsong Kang
- Department of Brain and Cognitive Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Kun Ho Lee
- Gwangju Alzheimer's & Related Dementia Cohort Research Center, Chosun University, Gwangju 61452, Republic of Korea; Department of Biomedical Science and Gwangju Alzheimer's & Related Dementia Cohort Research Center, Chosun University, Gwangju 61452, Republic of Korea; Korea Brain Research Institute, Daegu 41062, Republic of Korea.
| | - Heung-Il Suk
- Department of Artificial Intelligence, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
2
|
Kumar S, Earnest T, Yang B, Kothapalli D, Aschenbrenner AJ, Hassenstab J, Xiong C, Ances B, Morris J, Benzinger TLS, Gordon BA, Payne P, Sotiras A. Analyzing heterogeneity in Alzheimer disease using multimodal normative modeling on imaging-based ATN biomarkers. Alzheimers Dement 2025; 21:e70143. [PMID: 40235115 PMCID: PMC12000228 DOI: 10.1002/alz.70143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/17/2025]
Abstract
INTRODUCTION Previous studies have applied normative modeling on a single neuroimaging modality to investigate Alzheimer disease (AD) heterogeneity. We employed a deep learning-based multimodal normative framework to analyze individual-level variation across ATN (amyloid-tau-neurodegeneration) imaging biomarkers. METHODS We selected cross-sectional discovery (n = 665) and replication cohorts (n = 430) with available T1-weighted magnetic resonance imaging (MRI), amyloid, and tau positron emission tomography (PET). Normative modeling estimated individual-level abnormal deviations in amyloid-positive individuals compared to amyloid-negative controls. Regional abnormality patterns were mapped at different clinical group levels to assess intra-group heterogeneity. An individual-level disease severity index (DSI) was calculated using both the spatial extent and magnitude of abnormal deviations across ATN. RESULTS Greater intra-group heterogeneity in ATN abnormality patterns was observed in more severe clinical stages of AD. Higher DSI was associated with worse cognitive function and increased risk of disease progression. DISCUSSION Subject-specific abnormality maps across ATN reveal the heterogeneous impact of AD on the brain. HIGHLIGHTS Normative modeling examined AD heterogeneity across multimodal imaging biomarkers. Heterogeneity in spatial patterns of gray matter atrophy, amyloid, and tau burden. Higher within-group heterogeneity for AD patients at advanced dementia stages. Patient-specific metric summarized extent of neurodegeneration and neuropathology. Metric is a marker of poor brain health and can monitor risk of disease progression.
Collapse
Affiliation(s)
- Sayantan Kumar
- Department of Computer Science and EngineeringWashington University in St LouisSaint LouisMissouriUSA
- Institute for InformaticsData Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Tom Earnest
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Braden Yang
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Deydeep Kothapalli
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | | | - Jason Hassenstab
- Department of NeurologyWashington University School of MedicineSt louisMissouriUSA
| | - Chengie Xiong
- Institute for InformaticsData Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Beau Ances
- Department of NeurologyWashington University School of MedicineSt louisMissouriUSA
| | - John Morris
- Department of NeurologyWashington University School of MedicineSt louisMissouriUSA
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Brian A. Gordon
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Philip Payne
- Department of Computer Science and EngineeringWashington University in St LouisSaint LouisMissouriUSA
- Institute for InformaticsData Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Aristeidis Sotiras
- Institute for InformaticsData Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | | |
Collapse
|
3
|
Xu H, Mu S, Bao J, Davatzikos C, Shou H, Shen L. High-dimensional mediation analysis reveals the mediating role of physical activity patterns in genetic pathways leading to AD-like brain atrophy. BioData Min 2025; 18:24. [PMID: 40128806 PMCID: PMC11931790 DOI: 10.1186/s13040-025-00432-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 02/07/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex disorder that affects multiple biological systems including cognition, behavior and physical health. Unfortunately, the pathogenic mechanisms behind AD are not yet clear and the treatment options are still limited. Despite the increasing number of studies examining the pairwise relationships between genetic factors, physical activity (PA), and AD, few have successfully integrated all three domains of data, which may help reveal mechanisms and impact of these genomic and phenomic factors on AD. We use high-dimensional mediation analysis as an integrative framework to study the relationships among genetic factors, PA and AD-like brain atrophy quantified by spatial patterns of brain atrophy. RESULTS We integrate data from genetics, PA and neuroimaging measures collected from 13,425 UK Biobank samples to unveil the complex relationship among genetic risk factors, behavior and brain signatures in the contexts of aging and AD. Specifically, we used a composite imaging marker, Spatial Pattern of Abnormality for Recognition of Early AD (SPARE-AD) that characterizes AD-like brain atrophy, as an outcome variable to represent AD risk. Through GWAS, we identified single nucleotide polymorphisms (SNPs) that are significantly associated with SPARE-AD as exposure variables. We employed conventional summary statistics and functional principal component analysis to extract patterns of PA as mediators. After constructing these variables, we utilized a high-dimensional mediation analysis method, Bayesian Mediation Analysis (BAMA), to estimate potential mediating pathways between SNPs, multivariate PA signatures and SPARE-AD. BAMA incorporates Bayesian continuous shrinkage prior to select the active mediators from a large pool of candidates. We identified a total of 22 mediation pathways, indicating how genetic variants can influence SPARE-AD by altering physical activity. By comparing the results with those obtained using univariate mediation analysis, we demonstrate the advantages of high-dimensional mediation analysis methods over univariate mediation analysis. CONCLUSION Through integrative analysis of multi-omics data, we identified several mediation pathways of physical activity between genetic factors and SPARE-AD. These findings contribute to a better understanding of the pathogenic mechanisms of AD. Moreover, our research demonstrates the potential of the high-dimensional mediation analysis method in revealing the mechanisms of disease.
Collapse
Affiliation(s)
- Hanxiang Xu
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Family Medicine and Public Health, University of California, San Diego, CA, 92093, USA
| | - Shizhuo Mu
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Christos Davatzikos
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
4
|
Govindarajan ST, Mamourian E, Erus G, Abdulkadir A, Melhem R, Doshi J, Pomponio R, Tosun D, Bilgel M, An Y, Sotiras A, Marcus DS, LaMontagne P, Benzinger TLS, Espeland MA, Masters CL, Maruff P, Launer LJ, Fripp J, Johnson SC, Morris JC, Albert MS, Bryan RN, Resnick SM, Habes M, Shou H, Wolk DA, Nasrallah IM, Davatzikos C. Machine learning reveals distinct neuroanatomical signatures of cardiovascular and metabolic diseases in cognitively unimpaired individuals. Nat Commun 2025; 16:2724. [PMID: 40108173 PMCID: PMC11923046 DOI: 10.1038/s41467-025-57867-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
Comorbid cardiovascular and metabolic risk factors (CVM) differentially impact brain structure and increase dementia risk, but their specific magnetic resonance imaging signatures (MRI) remain poorly characterized. To address this, we developed and validated machine learning models to quantify the distinct spatial patterns of atrophy and white matter hyperintensities related to hypertension, hyperlipidemia, smoking, obesity, and type-2 diabetes mellitus at the patient level. Using harmonized MRI data from 37,096 participants (45-85 years) in a large multinational dataset of 10 cohort studies, we generated five in silico severity markers that: i) outperformed conventional structural MRI markers with a ten-fold increase in effect sizes, ii) captured subtle patterns at sub-clinical CVM stages, iii) were most sensitive in mid-life (45-64 years), iv) were associated with brain beta-amyloid status, and v) showed stronger associations with cognitive performance than diagnostic CVM status. Integrating personalized measurements of CVM-specific brain signatures into phenotypic frameworks could guide early risk detection and stratification in clinical studies.
Collapse
Affiliation(s)
| | - Elizabeth Mamourian
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Centre for Artificial Intelligence, ZHAW School of Engineering, Winterthur, Switzerland
| | - Randa Melhem
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jimit Doshi
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymond Pomponio
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Aristeidis Sotiras
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark A Espeland
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Colin L Masters
- Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Maruff
- Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Lenore J Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mohamad Habes
- Biggs Alzheimer's Institute, University of Texas San Antonio Health Science Center, San Antonio, TX, USA
| | - Haochang Shou
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Karlsson L, Vogel J, Arvidsson I, Åström K, Strandberg O, Seidlitz J, Bethlehem RAI, Stomrud E, Ossenkoppele R, Ashton NJ, Zetterberg H, Blennow K, Palmqvist S, Smith R, Janelidze S, La Joie R, Rabinovici GD, Binette AP, Mattsson‐Carlgren N, Hansson O. Machine learning prediction of tau-PET in Alzheimer's disease using plasma, MRI, and clinical data. Alzheimers Dement 2025; 21:e14600. [PMID: 39985487 PMCID: PMC11846480 DOI: 10.1002/alz.14600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/24/2025]
Abstract
INTRODUCTION Tau positron emission tomography (PET) is a reliable neuroimaging technique for assessing regional load of tau pathology in the brain, but its routine clinical use is limited by cost and accessibility barriers. METHODS We thoroughly investigated the ability of various machine learning models to predict clinically useful tau-PET composites (load and laterality index) from low-cost and non-invasive features, for example, clinical variables, plasma biomarkers, and structural magnetic resonance imaging (MRI). RESULTS Models including plasma biomarkers yielded the most accurate predictions of tau-PET burden (best model: R-squared = 0.66-0.69), with especially high contribution from plasma phosphorylated tau-217 (p-tau217). MRI variables were the best predictors of asymmetric tau load between the two hemispheres (best model: R-squared = 0.28-0.42). The models showed high generalizability to external test cohorts with data collected at multiple sites. Through a proof-of-concept two-step classification workflow, we also demonstrated possible model translations to a clinical setting. DISCUSSION This study highlights the promising and limiting aspects of using machine learning to predict tau-PET from scalable cost-effective variables, with findings relevant for clinical settings and future research. HIGHLIGHTS Accessible variables showed potential in estimating tau tangle load and distribution. Plasma phosphorylated tau-217 (p-tau217) and magnetic resonance imaging (MRI) were the best predictors of different tau-PET (positron emission tomography) composites. Machine learning models demonstrated high generalizability across AD cohorts.
Collapse
Affiliation(s)
- Linda Karlsson
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
| | - Jacob Vogel
- Department of Clinical SciencesSciLifeLabLund UniversityLundSweden
| | - Ida Arvidsson
- Centre for Mathematical SciencesLund UniversityLundSweden
| | - Kalle Åström
- Centre for Mathematical SciencesLund UniversityLundSweden
| | - Olof Strandberg
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
| | - Jakob Seidlitz
- Penn/CHOP Lifespan Brain InstituteUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Child and Adolescent Psychiatry and Behavioral ScienceThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Institute for Translational Medicine and TherapeuticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Erik Stomrud
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Rik Ossenkoppele
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceAmsterdam UMCAmsterdamthe Netherlands
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Institute of PsychiatryPsychology and NeuroscienceMaurice Wohl Institute Clinical NeuroscienceKing's College LondonLondonUK
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative Diseases5/FBuilding 5E5 Science Park East AvenueHong Kong Science ParkClear Water BayHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Paris Brain InstituteICMPitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiAnhuiP.R. China
| | - Sebastian Palmqvist
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Ruben Smith
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Shorena Janelidze
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
| | - Renaud La Joie
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Gil D. Rabinovici
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoSan FranciscoCaliforniaUSA
| | - Alexa Pichet Binette
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
| | - Niklas Mattsson‐Carlgren
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
| |
Collapse
|
6
|
Yi D, Byun MS, Park J, Kim J, Jung G, Ahn H, Lee J, Lee Y, Kim YK, Kang KM, Sohn C, Liu S, Huang Y, Saykin AJ, Lee DY, Nho K. Tau pathway-based gene analysis on PET identifies CLU and FYN in a Korean cohort. Alzheimers Dement 2025; 21:e14416. [PMID: 39625110 PMCID: PMC11848168 DOI: 10.1002/alz.14416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION The influence of genetic variation on tau protein aggregation, a key factor in Alzheimer's disease (AD), remains not fully understood. We aimed to identify novel genes associated with brain tau deposition using pathway-based candidate gene association analysis in a Korean cohort. METHODS We analyzed data for 146 older adults from the well-established Korean AD continuum cohort (Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease; KBASE). Fifteen candidate genes related to both tau pathways and AD were selected. Association analyses were performed using PLINK: A tool set for whole-genome association and population-based linkage analyses (PLINK) on tau deposition measured by 18F-AV-1451 positron emission tomography (PET) scans, with additional voxel-wise analysis conducted using Statistical Parametric Mapping 12 (SPM12). RESULTS CLU and FYN were significantly associated with tau deposition, with the most significant single-nucleotide polymorphisms (SNPs) being rs149413552 and rs57650567, respectively. These SNPs were linked to increased tau across key brain regions and showed additive effects with apolipoprotein E (APOE) ε4. DISCUSSION CLU and FYN may play specific roles in tau pathophysiology, offering potential targets for biomarkers and therapies. HIGHLIGHTS Gene-based analysis identified CLU and FYN as associated with tau deposition on positron emission tomography (PET). CLU rs149413552 and FYN rs57650567 were associated with brain tau deposition. rs149413552 and rs57650567 were associated with structural brain atrophy. CLU rs149413552 was associated with immediate verbal memory. CLU and FYN may play specific roles in tau pathophysiology.
Collapse
Affiliation(s)
- Dahyun Yi
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSouth Korea
| | - Min Soo Byun
- Department of NeuropsychiatrySeoul National University HospitalSeoulSouth Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSouth Korea
| | - Jong‐Ho Park
- Precision Medicine CenterSeoul National University Bundang HospitalSeongnam‐siGyeonggi‐doSouth Korea
| | - Jong‐Won Kim
- Department of Laboratory Medicine and GeneticsSamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
| | - Gijung Jung
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSouth Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive ScienceSeoul National University College of HumanitiesGwanak‐guSeoulSouth Korea
| | - Jun‐Young Lee
- Department of PsychiatrySeoul National University Boramae Medical Center, Dongjak‐guSeoulSouth Korea
| | - Yun‐Sang Lee
- Department of Nuclear MedicineSeoul National University College of MedicineJongro‐guSeoulSouth Korea
| | - Yu Kyeong Kim
- Department of Nuclear MedicineSeoul National University Boramae Medical Center, Dongjak‐guSeoulSouth Korea
| | - Koung Mi Kang
- Department of RadiologySeoul National University Hospital, Jongro‐guSeoulSouth Korea
- Department of RadiologySeoul National University College of Medicine, Jongro‐guSeoulSouth Korea
| | - Chul‐Ho Sohn
- Department of RadiologySeoul National University Hospital, Jongro‐guSeoulSouth Korea
| | - Shiwei Liu
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Yen‐Ning Huang
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Andrew J. Saykin
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Dong Young Lee
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSouth Korea
- Department of NeuropsychiatrySeoul National University HospitalSeoulSouth Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSouth Korea
| | - Kwangsik Nho
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | | |
Collapse
|
7
|
Duggan MR, Paterson C, Lu Y, Biegel H, Dark HE, Cordon J, Bilgel M, Kaneko N, Shibayama M, Kato S, Furuichi M, Waga I, Hiraga K, Katsuno M, Nishita Y, Otsuka R, Davatzikos C, Erus G, Loupy K, Simpson M, Lewis A, Moghekar A, Palta P, Gottesman RF, Resnick SM, Coresh J, Williams SA, Walker KA. The Dementia SomaSignal Test (dSST): A plasma proteomic predictor of 20-year dementia risk. Alzheimers Dement 2025; 21:e14549. [PMID: 39936291 PMCID: PMC11851157 DOI: 10.1002/alz.14549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 02/13/2025]
Abstract
INTRODUCTION There is an unmet need for tools to quantify dementia risk during its multi-decade preclinical/prodromal phase, given that current biomarkers predict risk over shorter follow-up periods and are specific to Alzheimer's disease. METHODS Using high-throughput proteomic assays and machine learning techniques in the Atherosclerosis Risk in Communities study (n = 11,277), we developed the Dementia SomaSignal Test (dSST). RESULTS In addition to outperforming existing plasma biomarkers, the dSST predicted mid-life dementia risk over a 20-year follow-up across two independent cohorts with different ethnic backgrounds (areas under the curve [AUCs]: dSST 0.68-0.70, dSST+age 0.75-0.81). In a separate cohort, the dSST was associated with longitudinal declines across multiple cognitive domains, accelerated brain atrophy, and elevated measures of neuropathology (as evidenced by positron emission tomography and plasma biomarkers). DISCUSSION The dSST is a cost-effective, scalable, and minimally invasive protein-based prognostic aid that can quantify risk up to two decades before dementia onset. HIGHLIGHTS The Dementia SomaSignal Test (dSST) predicts 20-year dementia risk across two independent cohorts. dSST outperforms existing plasma biomarkers in predicting multi-decade dementia risk. dSST predicts cognitive decline and accelerated brain atrophy in a third cohort. dSST is a prognostic aid that can predict dementia risk over two decades.
Collapse
Grants
- U01HL096812 NHLBI, NIA, NINDS, NIDCD
- U01 HL096812 NHLBI NIH HHS
- 75N92022D00002 NHLBI NIH HHS
- U01 HL096917 NHLBI NIH HHS
- U01 HL096902 NHLBI NIH HHS
- U01HL096902 NHLBI, NIA, NINDS, NIDCD
- 75N92022D00004 NHLBI NIH HHS
- U01HL096917 NHLBI, NIA, NINDS, NIDCD
- U01HL096814 NHLBI, NIA, NINDS, NIDCD
- U01 HL096814 NHLBI NIH HHS
- 75N92022D00003 NHLBI NIH HHS
- 75N92022D00005 NHLBI NIH HHS
- Intramural Research Program (IRP) of the National Institute on Aging (NIA)
- 75N92022D00001 NHLBI NIH HHS
- National Center for Geriatrics and Gerontology
- Nagoya University
- U01HL096899 NHLBI, NIA, NINDS, NIDCD
- NEC Solution Innovators Limited
- U01 HL096899 NHLBI NIH HHS
- National Center for Geriatrics and Gerontology
- Nagoya University
Collapse
Affiliation(s)
- Michael R. Duggan
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Clare Paterson
- Department of Clinical and Research DevelopmentStandard BioToolsBoulderColoradoUSA
| | - Yifei Lu
- Department of EpidemiologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Hannah Biegel
- Department of Clinical and Research DevelopmentStandard BioToolsBoulderColoradoUSA
| | - Heather E. Dark
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Jenifer Cordon
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Murat Bilgel
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Naoto Kaneko
- Innovation LaboratoryNEC Solution Innovators Limited, TokyoKoto‐kuJapan
| | - Masaki Shibayama
- Innovation LaboratoryNEC Solution Innovators Limited, TokyoKoto‐kuJapan
| | - Shintaro Kato
- Innovation LaboratoryNEC Solution Innovators Limited, TokyoKoto‐kuJapan
- FonesLife Proteomics LaboratoryFonesLife Corporation, Chuo CityTokyoJapan
| | - Makio Furuichi
- Innovation LaboratoryNEC Solution Innovators Limited, TokyoKoto‐kuJapan
- FonesLife Proteomics LaboratoryFonesLife Corporation, Chuo CityTokyoJapan
| | - Iwao Waga
- Innovation LaboratoryNEC Solution Innovators Limited, TokyoKoto‐kuJapan
- FonesLife Proteomics LaboratoryFonesLife Corporation, Chuo CityTokyoJapan
- Well‐being Design Institute for HealthTohoku UniversityAoba‐kuSendaiJapan
| | - Keita Hiraga
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaAichiJapan
| | - Masahisa Katsuno
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaAichiJapan
- Department of Clinical Research EducationNagoya University Graduate School of MedicineNagoyaAichiJapan
| | - Yukiko Nishita
- Department of Epidemiology of AgingNational Center for Geriatrics and GerontologyObuAichiJapan
| | - Rei Otsuka
- Department of Epidemiology of AgingNational Center for Geriatrics and GerontologyObuAichiJapan
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Kelsey Loupy
- Department of Clinical and Research DevelopmentStandard BioToolsBoulderColoradoUSA
| | - Melissa Simpson
- Department of Clinical and Research DevelopmentStandard BioToolsBoulderColoradoUSA
| | - Alexandria Lewis
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Abhay Moghekar
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Priya Palta
- Department of NeurologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Rebecca F. Gottesman
- Stroke BranchNational Institute of Neurological Disorders and StrokeBethesdaMarylandUSA
| | - Susan M. Resnick
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Josef Coresh
- Departments of Population Health and MedicineNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Stephen A. Williams
- Department of Clinical and Research DevelopmentStandard BioToolsBoulderColoradoUSA
| | - Keenan A. Walker
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
8
|
Joynes CM, Bilgel M, An Y, Moghekar AR, Ashton NJ, Kac PR, Karikari TK, Blennow K, Zetterberg H, Thambisetty M, Ferrucci L, Resnick SM, Walker KA. Sex differences in the trajectories of plasma biomarkers, brain atrophy, and cognitive decline relative to amyloid onset. Alzheimers Dement 2025; 21:e14405. [PMID: 39609257 PMCID: PMC11775450 DOI: 10.1002/alz.14405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/30/2024]
Abstract
INTRODUCTION The factors that influence the progression of Alzheimer's disease (AD) after individuals become amyloid-positive are poorly understood. This study examines how sex influences the longitudinal trajectories of plasma AD and neurodegenerative biomarkers in the years following a person's estimated onset of amyloid-β. METHODS Linear mixed-effects modeling investigated overall and sex-specific longitudinal trajectories of plasma biomarkers, brain volumes, and cognition relative to the estimated age of amyloid onset in a cohort of 78 amyloid-positive Baltimore Longitudinal Study of Aging (BLSA) participants (n = 45 male; follow-up time: 6.8 years [SD 3.31]). Amyloid status was ascertained with 11C-Pittsburgh compound B (PiB) PET imaging. RESULTS After amyloid onset, men displayed steeper increases in pTau181, pTau231, and neurofilament light (NfL) compared to women. In this same period, men demonstrated steeper declines in brain volume and cognitive performance. DISCUSSION These findings suggest that sex influences the trajectory of AD pathology, neuronal injury, and symptom progression after individuals become amyloid-positive. HIGHLIGHTS Steeper rates of increase in pTau and GFAP among amyloid-positive individuals. After amyloid onset, steeper increases in pTau and NfL concentrations in men than in women. Steeper declines in brain volume and cognition in men corroborate biomarker results.
Collapse
Affiliation(s)
- Cassandra M. Joynes
- Laboratory of Behavioral NeuroscienceNational Institute on AgingBaltimoreMarylandUSA
| | - Murat Bilgel
- Laboratory of Behavioral NeuroscienceNational Institute on AgingBaltimoreMarylandUSA
| | - Yang An
- Laboratory of Behavioral NeuroscienceNational Institute on AgingBaltimoreMarylandUSA
| | - Abhay R. Moghekar
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- King's College LondonInstitute of PsychiatryPsychology and NeuroscienceMaurice Wohl Clinical Neuroscience InstituteLondonUK
- NIHR Biomedical Research Centre for Mental Health and Biomedical ResearchUnit for Dementia at South London and MaudsleyNHS FoundationLondonUK
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
| | - Przemysław R. Kac
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
| | - Thomas K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesShatinHong Kong
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Madhav Thambisetty
- Laboratory of Behavioral NeuroscienceNational Institute on AgingBaltimoreMarylandUSA
| | - Luigi Ferrucci
- Translational Gerontology BranchNational Institute on AgingBaltimoreMarylandUSA
| | - Susan M. Resnick
- Laboratory of Behavioral NeuroscienceNational Institute on AgingBaltimoreMarylandUSA
| | - Keenan A. Walker
- Laboratory of Behavioral NeuroscienceNational Institute on AgingBaltimoreMarylandUSA
| |
Collapse
|
9
|
Xu Y, Zeger S, Wang Z. Probabilistic clustering using shared latent variable model for assessing Alzheimer's disease biomarkers. Biostatistics 2024; 26:kxaf010. [PMID: 40327339 PMCID: PMC12054513 DOI: 10.1093/biostatistics/kxaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 05/07/2025] Open
Abstract
The preclinical stage of many neurodegenerative diseases can span decades before symptoms become apparent. Understanding the sequence of preclinical biomarker changes provides a critical opportunity for early diagnosis and effective intervention prior to significant loss of patients' brain functions. The main challenge to early detection lies in the absence of direct observation of the disease state and the considerable variability in both biomarkers and disease dynamics among individuals. Recent research hypothesized the existence of subgroups with distinct biomarker patterns due to co-morbidities and degrees of brain resilience. Our ability to diagnose early and intervene during the preclinical stage of neurodegenerative diseases will be enhanced by further insights into heterogeneity in the biomarker-disease relationship. In this article, we focus on Alzheimer's disease (AD) and attempt to identify the systematic patterns within the heterogeneous AD biomarker-disease cascade. Specifically, we quantify the disease progression using a dynamic latent variable whose mixture distribution represents patient subgroups. Model estimation uses Hamiltonian Monte Carlo with the number of clusters determined by the Bayesian Information Criterion. We report simulation studies that investigate the performance of the proposed model in finite sample settings that are similar to our motivating application. We apply the proposed model to the Biomarkers of Cognitive Decline Among Normal Individuals data, a longitudinal study that was conducted over 2 decades among individuals who were initially cognitively normal. Our application yields evidence consistent with the hypothetical model of biomarker dynamics presented in Jack Jr et al. In addition, our analysis identified 2 subgroups with distinct disease-onset patterns. Finally, we develop a dynamic prediction approach to improve the precision of prognoses.
Collapse
Affiliation(s)
- Yizhen Xu
- Division of Biostatistics, Department of Population Health Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Scott Zeger
- Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Zheyu Wang
- Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Division of Quantitative Sciences, Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| |
Collapse
|
10
|
Antoniades M, Srinivasan D, Wen J, Erus G, Abdulkadir A, Mamourian E, Melhem R, Hwang G, Cui Y, Govindarajan ST, Chen AA, Zhou Z, Yang Z, Chen J, Pomponio R, Sotardi S, An Y, Bilgel M, LaMontagne P, Singh A, Benzinger T, Beason-Held L, Marcus DS, Yaffe K, Launer L, Morris JC, Tosun D, Ferrucci L, Bryan RN, Resnick SM, Habes M, Wolk D, Fan Y, Nasrallah IM, Shou H, Davatzikos C. Relationship between MRI brain-age heterogeneity, cognition, genetics and Alzheimer's disease neuropathology. EBioMedicine 2024; 109:105399. [PMID: 39437659 PMCID: PMC11536027 DOI: 10.1016/j.ebiom.2024.105399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Brain ageing is highly heterogeneous, as it is driven by a variety of normal and neuropathological processes. These processes may differentially affect structural and functional brain ageing across individuals, with more pronounced ageing (older brain age) during midlife being indicative of later development of dementia. Here, we examined whether brain-ageing heterogeneity in unimpaired older adults related to neurodegeneration, different cognitive trajectories, genetic and amyloid-beta (Aβ) profiles, and to predicted progression to Alzheimer's disease (AD). METHODS Functional and structural brain age measures were obtained for resting-state functional MRI and structural MRI, respectively, in 3460 cognitively normal individuals across an age range spanning 42-85 years. Participants were categorised into four groups based on the difference between their chronological and predicted age in each modality: advanced age in both (n = 291), resilient in both (n = 260) or advanced in one/resilient in the other (n = 163/153). With the resilient group as the reference, brain-age groups were compared across neuroimaging features of neuropathology (white matter hyperintensity volume, neuronal loss measured with Neurite Orientation Dispersion and Density Imaging, AD-specific atrophy patterns measured with the Spatial Patterns of Abnormality for Recognition of Early Alzheimer's Disease index, amyloid burden using amyloid positron emission tomography (PET), progression to mild cognitive impairment and baseline and longitudinal cognitive measures (trail making task, mini mental state examination, digit symbol substitution task). FINDINGS Individuals with advanced structural and functional brain-ages had more features indicative of neurodegeneration and they had poor cognition. Individuals with a resilient brain-age in both modalities had a genetic variant that has been shown to be associated with age of onset of AD. Mixed brain-age was associated with selective cognitive deficits. INTERPRETATION The advanced group displayed evidence of increased atrophy across all neuroimaging features that was not found in either of the mixed groups. This is in line with biomarkers of preclinical AD and cerebrovascular disease. These findings suggest that the variation in structural and functional brain ageing across individuals reflects the degree of underlying neuropathological processes and may indicate the propensity to develop dementia in later life. FUNDING The National Institute on Aging, the National Institutes of Health, the Swiss National Science Foundation, the Kaiser Foundation Research Institute and the National Heart, Lung, and Blood Institute.
Collapse
Affiliation(s)
- Mathilde Antoniades
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Dhivya Srinivasan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA
| | - Guray Erus
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Clinical Neuroscience, Center for Research in Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - Elizabeth Mamourian
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yuhan Cui
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja Tirumalai Govindarajan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew A Chen
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zhen Zhou
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiong Chen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymond Pomponio
- Department of Biostatistics, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Susan Sotardi
- Department of Radiology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ashish Singh
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lori Beason-Held
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Lenore Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institute of Health, Baltimore, MD 21224, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mohamad Habes
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - David Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Christos Davatzikos
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Wen J, Yang Z, Nasrallah IM, Cui Y, Erus G, Srinivasan D, Abdulkadir A, Mamourian E, Hwang G, Singh A, Bergman M, Bao J, Varol E, Zhou Z, Boquet-Pujadas A, Chen J, Toga AW, Saykin AJ, Hohman TJ, Thompson PM, Villeneuve S, Gollub R, Sotiras A, Wittfeld K, Grabe HJ, Tosun D, Bilgel M, An Y, Marcus DS, LaMontagne P, Benzinger TL, Heckbert SR, Austin TR, Launer LJ, Espeland M, Masters CL, Maruff P, Fripp J, Johnson SC, Morris JC, Albert MS, Bryan RN, Resnick SM, Ferrucci L, Fan Y, Habes M, Wolk D, Shen L, Shou H, Davatzikos C. Genetic and clinical correlates of two neuroanatomical AI dimensions in the Alzheimer's disease continuum. Transl Psychiatry 2024; 14:420. [PMID: 39368996 PMCID: PMC11455841 DOI: 10.1038/s41398-024-03121-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024] Open
Abstract
Alzheimer's disease (AD) is associated with heterogeneous atrophy patterns. We employed a semi-supervised representation learning technique known as Surreal-GAN, through which we identified two latent dimensional representations of brain atrophy in symptomatic mild cognitive impairment (MCI) and AD patients: the "diffuse-AD" (R1) dimension shows widespread brain atrophy, and the "MTL-AD" (R2) dimension displays focal medial temporal lobe (MTL) atrophy. Critically, only R2 was associated with widely known sporadic AD genetic risk factors (e.g., APOE ε4) in MCI and AD patients at baseline. We then independently detected the presence of the two dimensions in the early stages by deploying the trained model in the general population and two cognitively unimpaired cohorts of asymptomatic participants. In the general population, genome-wide association studies found 77 genes unrelated to APOE differentially associated with R1 and R2. Functional analyses revealed that these genes were overrepresented in differentially expressed gene sets in organs beyond the brain (R1 and R2), including the heart (R1) and the pituitary gland, muscle, and kidney (R2). These genes were enriched in biological pathways implicated in dendritic cells (R2), macrophage functions (R1), and cancer (R1 and R2). Several of them were "druggable genes" for cancer (R1), inflammation (R1), cardiovascular diseases (R1), and diseases of the nervous system (R2). The longitudinal progression showed that APOE ε4, amyloid, and tau were associated with R2 at early asymptomatic stages, but this longitudinal association occurs only at late symptomatic stages in R1. Our findings deepen our understanding of the multifaceted pathogenesis of AD beyond the brain. In early asymptomatic stages, the two dimensions are associated with diverse pathological mechanisms, including cardiovascular diseases, inflammation, and hormonal dysfunction-driven by genes different from APOE-which may collectively contribute to the early pathogenesis of AD. All results are publicly available at https://labs-laboratory.com/medicine/ .
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA.
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Research Lab in Neuroimaging of the Department of Clinical Neurosciences at Lausanne University Hospital, Lausanne, Switzerland
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashish Singh
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Bergman
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erdem Varol
- Department of Statistics, Center for Theoretical Neuroscience, Zuckerman Institute, Columbia University, New York, NY, USA
| | - Zhen Zhou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aleix Boquet-Pujadas
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA
| | - Jiong Chen
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of NeuroImaging, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Andrew J Saykin
- Radiology and Imaging Sciences, Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research Center and the Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt Genetics Institute, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, McGill University, Montréal, QC, Canada
| | - Randy Gollub
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute for Informatics, Washington University School of Medicine, St. Louis, MO, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R Austin
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Lenore J Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Mark Espeland
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, QLD, Australia
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, 21225, USA
| | - Yong Fan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - David Wolk
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology and Penn Memory Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Kawano H, Yamada S, Watanabe Y, Ii S, Otani T, Ito H, Okada K, Iseki C, Tanikawa M, Wada S, Oshima M, Mase M, Yoshida K. Aging and Sex Differences in Brain Volume and Cerebral Blood Flow. Aging Dis 2024; 15:2216-2229. [PMID: 38029394 PMCID: PMC11346398 DOI: 10.14336/ad.2023.1122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023] Open
Abstract
How do regional brain volume ratios and cerebral blood flow (CBF, mL/min) change with aging, and are there sex differences? This study aimed to comprehensively evaluate the relationships between regional brain volume ratios and CBF in healthy brains. The study participants were healthy volunteers who underwent three-dimensional T1-weighted MRI, time-of-flight MR angiography, and four-dimensional (4D) flow MRI between 2020 and 2022. The brain was automatically segmented into 21 brain subregions from 3D T1-weighted MRI, and CBF in 16 major intracranial arteries were measured by 4D flow MRI. The relationships between segmented brain volume ratios and CBFs around the circle of Willis were comprehensively investigated in each decade and sex. This study included 129 healthy volunteers (mean age ± SD, 48.2 ± 16.8; range, 22-92 years; 43 males and 86 females). The association was strongest between the cortical gray matter volume ratio and total outflow of the intracranial major arteries distal to the circle of Willis (Pearson's correlation coefficient, r: 0.425). In addition, the mean flow of the total inflow and outflow around the circle of Willis were significantly greater in women than men, and significant left-right differences were observed in CBFs even on the peripheral side of the circle of Willis. Moreover, the correlation was strongest between the left cortical gray matter volume ratio and the combined flows of the left anterior and posterior cerebral arteries distal to the circle of Willis (r: 0.486). There was a trend toward greater total intracranial CBF, especially among women in their 40s and younger, who had a larger cortical gray matter volume. This finding may be one of the reasons for the approximately twofold higher incidence of cerebral aneurysms and subarachnoid hemorrhage, and a threefold higher incidence of migraine headaches.
Collapse
Affiliation(s)
- Hiroto Kawano
- Department of Neurosurgery, Shiga University of Medical Science, Shiga, Japan.
| | - Shigeki Yamada
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, Japan.
- Interfaculty Initiative in Information Studies / Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.
| | - Yoshiyuki Watanabe
- Department of Radiology, Shiga University of Medical Science, Shiga, Japan.
| | - Satoshi Ii
- Faculty of System Design, Tokyo Metropolitan University, Tokyo, Japan.
| | - Tomohiro Otani
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan.
| | - Hirotaka Ito
- Medical System Research & Development Center, FUJIFILM Corporation, Tokyo, Japan.
| | - Ko Okada
- Medical System Research & Development Center, FUJIFILM Corporation, Tokyo, Japan.
| | - Chifumi Iseki
- Department of Behavioral Neurology and Cognitive Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
- Division of Neurology and Clinical Neuroscience, Department of Internal Medicine III, Yamagata University School of Medicine, Yamagata, Japan.
| | - Motoki Tanikawa
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, Japan.
| | - Shigeo Wada
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan.
| | - Marie Oshima
- Interfaculty Initiative in Information Studies / Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.
| | - Mitsuhito Mase
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, Japan.
| | - Kazumichi Yoshida
- Department of Neurosurgery, Shiga University of Medical Science, Shiga, Japan.
| |
Collapse
|
13
|
Yang Z, Wen J, Erus G, Govindarajan ST, Melhem R, Mamourian E, Cui Y, Srinivasan D, Abdulkadir A, Parmpi P, Wittfeld K, Grabe HJ, Bülow R, Frenzel S, Tosun D, Bilgel M, An Y, Yi D, Marcus DS, LaMontagne P, Benzinger TLS, Heckbert SR, Austin TR, Waldstein SR, Evans MK, Zonderman AB, Launer LJ, Sotiras A, Espeland MA, Masters CL, Maruff P, Fripp J, Toga AW, O'Bryant S, Chakravarty MM, Villeneuve S, Johnson SC, Morris JC, Albert MS, Yaffe K, Völzke H, Ferrucci L, Nick Bryan R, Shinohara RT, Fan Y, Habes M, Lalousis PA, Koutsouleris N, Wolk DA, Resnick SM, Shou H, Nasrallah IM, Davatzikos C. Brain aging patterns in a large and diverse cohort of 49,482 individuals. Nat Med 2024; 30:3015-3026. [PMID: 39147830 PMCID: PMC11483219 DOI: 10.1038/s41591-024-03144-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 06/20/2024] [Indexed: 08/17/2024]
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.
Collapse
Affiliation(s)
- Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
- GE Healthcare, Bellevue, WA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja T Govindarajan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Paraskevi Parmpi
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- Site Rostock/Greifswald, German Center for Neurodegenerative Diseases (DZNE), Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University of Greifswald, Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Daniel S Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R Austin
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Shari R Waldstein
- Department of Psychology, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Alan B Zonderman
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Lenore J Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute for Informatics, Data Science & Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Mark A Espeland
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Colin L Masters
- Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Maruff
- Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Sid O'Bryant
- Institute for Translational Research University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Mallar M Chakravarty
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Dept of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Yaffe
- Departments of Neurology, Psychiatry and Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, Baltimore, MD, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Russell T Shinohara
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Nikolaos Koutsouleris
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Section for Precision Psychiatry, Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Wen J, Antoniades M, Yang Z, Hwang G, Skampardoni I, Wang R, Davatzikos C. Dimensional Neuroimaging Endophenotypes: Neurobiological Representations of Disease Heterogeneity Through Machine Learning. Biol Psychiatry 2024; 96:564-584. [PMID: 38718880 PMCID: PMC11374488 DOI: 10.1016/j.biopsych.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/29/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024]
Abstract
Machine learning has been increasingly used to obtain individualized neuroimaging signatures for disease diagnosis, prognosis, and response to treatment in neuropsychiatric and neurodegenerative disorders. Therefore, it has contributed to a better understanding of disease heterogeneity by identifying disease subtypes with different brain phenotypic measures. In this review, we first present a systematic literature overview of studies using machine learning and multimodal magnetic resonance imaging to unravel disease heterogeneity in various neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease, schizophrenia, major depressive disorder, autism spectrum disorder, and multiple sclerosis, as well as their potential in a transdiagnostic framework, where neuroanatomical and neurobiological commonalities were assessed across diagnostic boundaries. Subsequently, we summarize relevant machine learning methodologies and their clinical interpretability. We discuss the potential clinical implications of the current findings and envision future research avenues. Finally, we discuss an emerging paradigm called dimensional neuroimaging endophenotypes. Dimensional neuroimaging endophenotypes dissects the neurobiological heterogeneity of neuropsychiatric and neurodegenerative disorders into low-dimensional yet informative, quantitative brain phenotypic representations, serving as robust intermediate phenotypes (i.e., endophenotypes), presumably reflecting the interplay of underlying genetic, lifestyle, and environmental processes associated with disease etiology.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science, University of Southern California, Los Angeles, California.
| | - Mathilde Antoniades
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gyujoon Hwang
- Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rongguang Wang
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
15
|
Karlsson L, Vogel J, Arvidsson I, Åström K, Strandberg O, Seidlitz J, Bethlehem RAI, Stomrud E, Ossenkoppele R, Ashton NJ, Zetterberg H, Blennow K, Palmqvist S, Smith R, Janelidze S, Joie RL, Rabinovici GD, Binette AP, Mattsson-Carlgren N, Hansson O. A machine learning-based prediction of tau load and distribution in Alzheimer's disease using plasma, MRI and clinical variables. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.31.24308264. [PMID: 38853877 PMCID: PMC11160861 DOI: 10.1101/2024.05.31.24308264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Tau positron emission tomography (PET) is a reliable neuroimaging technique for assessing regional load of tau pathology in the brain, commonly used in Alzheimer's disease (AD) research and clinical trials. However, its routine clinical use is limited by cost and accessibility barriers. Here we explore using machine learning (ML) models to predict clinically useful tau-PET composites from low-cost and non-invasive features, e.g., basic clinical variables, plasma biomarkers, and structural magnetic resonance imaging (MRI). Results demonstrated that models including plasma biomarkers yielded the most accurate predictions of tau-PET burden (best model: R-squared=0.66-0.68), with especially high contribution from plasma P-tau217. In contrast, MRI variables stood out as best predictors (best model: R-squared=0.28-0.42) of asymmetric tau load between the two hemispheres (an example of clinically relevant spatial information). The models showed high generalizability to external test cohorts with data collected at multiple sites. Based on these results, we also propose a proof-of-concept two-step classification workflow, demonstrating how the ML models can be translated to a clinical setting. This study uncovers current potential in predicting tau-PET information from scalable cost-effective variables, which could improve diagnosis and prognosis of AD.
Collapse
Affiliation(s)
- Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Jacob Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, SciLifeLab, Lund University, Lund, Sweden
| | - Ida Arvidsson
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Kalle Åström
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Jakob Seidlitz
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104 USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104 USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Richard A. I. Bethlehem
- University of Cambridge, Department of Psychology, Cambridge Biomedical Campus, Cambridge, CB2 3EB, UK
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience, King’s College London, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Gil D. Rabinovici
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
16
|
Wang R, Erus G, Chaudhari P, Davatzikos C. Adapting Machine Learning Diagnostic Models to New Populations Using a Small Amount of Data: Results from Clinical Neuroscience. ARXIV 2024:arXiv:2308.03175v2. [PMID: 39314511 PMCID: PMC11419182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Machine learning (ML) is revolutionizing many areas of engineering and science, including healthcare. However, it is also facing a reproducibility crisis, especially in healthcare. ML models that are carefully constructed from and evaluated on data from one part of the population may not generalize well on data from a different population group, or acquisition instrument settings and acquisition protocols. We tackle this problem in the context of neuroimaging of Alzheimer's disease (AD), schizophrenia (SZ) and brain aging. We develop a weighted empirical risk minimization approach that optimally combines data from a source group, e.g., subjects are stratified by attributes such as sex, age group, race and clinical cohort to make predictions on a target group, e.g., other sex, age group, etc. using a small fraction (10%) of data from the target group. We apply this method to multi-source data of 15,363 individuals from 20 neuroimaging studies to build ML models for diagnosis of AD and SZ, and estimation of brain age. We found that this approach achieves substantially better accuracy than existing domain adaptation techniques: it obtains area under curve greater than 0.95 for AD classification, area under curve greater than 0.7 for SZ classification and mean absolute error less than 5 years for brain age prediction on all target groups, achieving robustness to variations of scanners, protocols, and demographic or clinical characteristics. In some cases, it is even better than training on all data from the target group, because it leverages the diversity and size of a larger training set. We also demonstrate the utility of our models for prognostic tasks such as predicting disease progression in individuals with mild cognitive impairment. Critically, our brain age prediction models lead to new clinical insights regarding correlations with neurophysiological tests. In summary, we present a relatively simple methodology, along with ample experimental evidence, supporting the good generalization of ML models to new datasets and patient cohorts.
Collapse
Affiliation(s)
- Rongguang Wang
- Department of Electrical and Systems Engineering, University of Pennsylvania
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania
| | - Guray Erus
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania
| | - Pratik Chaudhari
- Department of Electrical and Systems Engineering, University of Pennsylvania
- Department of Computer and Information Science, University of Pennsylvania
| | - Christos Davatzikos
- Department of Electrical and Systems Engineering, University of Pennsylvania
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania
- Department of Radiology, Perelman School of Messdicine, University of Pennsylvania
| |
Collapse
|
17
|
Duggan MR, Yang Z, Cui Y, Dark HE, Wen J, Erus G, Hohman TJ, Chen J, Lewis A, Moghekar A, Coresh J, Resnick SM, Davatzikos C, Walker KA. Proteomic analyses reveal plasma EFEMP1 and CXCL12 as biomarkers and determinants of neurodegeneration. Alzheimers Dement 2024; 20:6486-6505. [PMID: 39129354 PMCID: PMC11497673 DOI: 10.1002/alz.14142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Plasma proteomic analyses of unique brain atrophy patterns may illuminate peripheral drivers of neurodegeneration and identify novel biomarkers for predicting clinically relevant outcomes. METHODS We identified proteomic signatures associated with machine learning-derived aging- and Alzheimer's disease (AD) -related brain atrophy patterns in the Baltimore Longitudinal Study of Aging (n = 815). Using data from five cohorts, we examined whether candidate proteins were associated with AD endophenotypes and long-term dementia risk. RESULTS Plasma proteins associated with distinct patterns of age- and AD-related atrophy were also associated with plasma/cerebrospinal fluid (CSF) AD biomarkers, cognition, AD risk, as well as mid-life (20-year) and late-life (8-year) dementia risk. EFEMP1 and CXCL12 showed the most consistent associations across cohorts and were mechanistically implicated as determinants of brain structure using genetic methods, including Mendelian randomization. DISCUSSION Our findings reveal plasma proteomic signatures of unique aging- and AD-related brain atrophy patterns and implicate EFEMP1 and CXCL12 as important molecular drivers of neurodegeneration. HIGHLIGHTS Plasma proteomic signatures are associated with unique patterns of brain atrophy. Brain atrophy-related proteins predict clinically relevant outcomes across cohorts. Genetic variation underlying plasma EFEMP1 and CXCL12 influences brain structure. EFEMP1 and CXCL12 may be important molecular drivers of neurodegeneration.
Collapse
Affiliation(s)
- Michael R. Duggan
- Laboratory of Behavioral NeuroscienceNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Heather E. Dark
- Laboratory of Behavioral NeuroscienceNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Junhao Wen
- Laboratory of Artificial Intelligence and Biomedical ScienceKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jingsha Chen
- Department of EpidemiologyJohns Hopkins University Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Alexandria Lewis
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Abhay Moghekar
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Josef Coresh
- Departments of Population Health and MedicineNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Susan M. Resnick
- Laboratory of Behavioral NeuroscienceNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Keenan A. Walker
- Laboratory of Behavioral NeuroscienceNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
18
|
Kumar S, Earnest T, Yang B, Kothapalli D, Aschenbrenner AJ, Hassenstab J, Xiong C, Ances B, Morris J, Benzinger TLS, Gordon BA, Payne P, Sotiras A. Analyzing heterogeneity in Alzheimer Disease using multimodal normative modeling on imaging-based ATN biomarkers. ARXIV 2024:arXiv:2404.05748v2. [PMID: 39010871 PMCID: PMC11247918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
INTRODUCTION Previous studies have applied normative modeling on a single neuroimaging modality to investigate Alzheimer Disease (AD) heterogeneity. We employed a deep learning-based multimodal normative framework to analyze individual-level variation across ATN (amyloid-tau-neurodegeneration) imaging biomarkers. METHODS We selected cross-sectional discovery (n = 665) and replication cohorts (n = 430) with available T1-weighted MRI, amyloid and tau PET. Normative modeling estimated individual-level abnormal deviations in amyloid-positive individuals compared to amyloid-negative controls. Regional abnormality patterns were mapped at different clinical group levels to assess intra-group heterogeneity. An individual-level disease severity index (DSI) was calculated using both the spatial extent and magnitude of abnormal deviations across ATN. RESULTS Greater intra-group heterogeneity in ATN abnormality patterns was observed in more severe clinical stages of AD. Higher DSI was associated with worse cognitive function and increased risk of disease progression. DISCUSSION Subject-specific abnormality maps across ATN reveal the heterogeneous impact of AD on the brain.
Collapse
Affiliation(s)
- Sayantan Kumar
- Department of Computer Science and Engineering, Washington University in St Louis; 1 Brookings Drive, Saint Louis, MO 63130
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
| | - Tom Earnest
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Braden Yang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Deydeep Kothapalli
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Andrew J. Aschenbrenner
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - Chengie Xiong
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
| | - Beau Ances
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - John Morris
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Brian A. Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Philip Payne
- Department of Computer Science and Engineering, Washington University in St Louis; 1 Brookings Drive, Saint Louis, MO 63130
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
| | - Aristeidis Sotiras
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| |
Collapse
|
19
|
Wittens MMJ, Denissen S, Sima DM, Fransen E, Niemantsverdriet E, Bastin C, Benoit F, Bergmans B, Bier JC, de Deyn PP, Deryck O, Hanseeuw B, Ivanoiu A, Picard G, Ribbens A, Salmon E, Segers K, Sieben A, Struyfs H, Thiery E, Tournoy J, van Binst AM, Versijpt J, Smeets D, Bjerke M, Nagels G, Engelborghs S. Brain age as a biomarker for pathological versus healthy ageing - a REMEMBER study. Alzheimers Res Ther 2024; 16:128. [PMID: 38877568 PMCID: PMC11179390 DOI: 10.1186/s13195-024-01491-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
OBJECTIVES This study aimed to evaluate the potential clinical value of a new brain age prediction model as a single interpretable variable representing the condition of our brain. Among many clinical use cases, brain age could be a novel outcome measure to assess the preventive effect of life-style interventions. METHODS The REMEMBER study population (N = 742) consisted of cognitively healthy (HC,N = 91), subjective cognitive decline (SCD,N = 65), mild cognitive impairment (MCI,N = 319) and AD dementia (ADD,N = 267) subjects. Automated brain volumetry of global, cortical, and subcortical brain structures computed by the CE-labeled and FDA-cleared software icobrain dm (dementia) was retrospectively extracted from T1-weighted MRI sequences that were acquired during clinical routine at participating memory clinics from the Belgian Dementia Council. The volumetric features, along with sex, were combined into a weighted sum using a linear model, and were used to predict 'brain age' and 'brain predicted age difference' (BPAD = brain age-chronological age) for every subject. RESULTS MCI and ADD patients showed an increased brain age compared to their chronological age. Overall, brain age outperformed BPAD and chronological age in terms of classification accuracy across the AD spectrum. There was a weak-to-moderate correlation between total MMSE score and both brain age (r = -0.38,p < .001) and BPAD (r = -0.26,p < .001). Noticeable trends, but no significant correlations, were found between BPAD and incidence of conversion from MCI to ADD, nor between BPAD and conversion time from MCI to ADD. BPAD was increased in heavy alcohol drinkers compared to non-/sporadic (p = .014) and moderate (p = .040) drinkers. CONCLUSIONS Brain age and associated BPAD have the potential to serve as indicators for, and to evaluate the impact of lifestyle modifications or interventions on, brain health.
Collapse
Affiliation(s)
- Mandy M J Wittens
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- Neuroprotection and Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije, Universiteit Brussel (VUB), Brussels, Belgium
| | - Stijn Denissen
- icometrix, Leuven, Belgium
- AIMS lab, Center for Neurosciences (C4N), Vrije Universiteit Brussel, UZ Brussel, Brussels, Belgium
| | - Diana M Sima
- Neuroprotection and Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije, Universiteit Brussel (VUB), Brussels, Belgium
- icometrix, Leuven, Belgium
| | - Erik Fransen
- Centre of Medical Genetics, University of Antwerp, and Antwerp University Hospital - UZA, Edegem, Belgium
| | | | - Christine Bastin
- GIGA-CRC-IVI, Liège University, Allée du Six Août, 8, Liège, 4000, Belgium
| | - Florence Benoit
- Geriatrics Department, Brugmann University Hospital, Universite Libre de Bruxelles, Brussels, Belgium
| | - Bruno Bergmans
- Neurology Department, AZ St-Jan Brugge, Brugge, Belgium
- Ghent University Hospital, Ghent, Belgium
| | - Jean-Christophe Bier
- Neurological department H. U. B. - Erasme Hospital - Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Peter Paul de Deyn
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, Antwerp, 2610, Belgium
- Memory Clinic, Ziekenhuisnetwerk, Antwerp, Belgium
| | - Olivier Deryck
- Neurology Department, AZ St-Jan Brugge, Brugge, Belgium
- Ghent University Hospital, Ghent, Belgium
| | - Bernard Hanseeuw
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, 1200, Belgium
- Department of Neurology, Clinique Universitaires Saint-Luc, Brussels, 1200, Belgium
- WELBIO Department, WEL Research Institute, Wavre, 1300, Belgium
| | - Adrian Ivanoiu
- Department of Neurology, Cliniques Universitaires St Luc, and Institute of Neuroscience, Université Catholique de Louvain, Woluwe-Saint-Lambert (Brussels), Belgium
| | - Gaëtane Picard
- Department of Neurology, Clinique Saint-Pierre, Ottignies, Belgium
| | | | - Eric Salmon
- GIGA-CRC-IVI, Liège University, Allée du Six Août, 8, Liège, 4000, Belgium
- Department of Neurology, Memory Clinic, Centre Hospitalier Universitaire (CHU) Liège, Liège, Belgium
| | - Kurt Segers
- Memory Clinic - Neurology and Geriatrics Department, CHU Brugmann, Van Gehuchtenplein 4, Brussels, 1020, Belgium
| | - Anne Sieben
- Neuropathology Lab, IBB-NeuroBiobank BB190113, Born Bunge Institute, Antwerp, Belgium
- Department of Pathology, Antwerp University Hospital - UZA, Antwerp, Belgium
- Laboratory of Neurology, Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Hanne Struyfs
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- Johnson and Johnson Innovative Medicine, Beerse, Belgium
| | - Evert Thiery
- Department of Neurology, University Hospital Ghent, Ghent University, Ghent, Belgium
| | - Jos Tournoy
- Department of Chronic Diseases, Metabolism and Ageing, Geriatric Medicine and Memory Clinic, University Hospitals Leuven and KU Leuven, Louvain, Belgium
| | - Anne-Marie van Binst
- Radiology Department, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Jan Versijpt
- Department of Neurology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- Neuroprotection and Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije, Universiteit Brussel (VUB), Brussels, Belgium
| | - Dirk Smeets
- Neuroprotection and Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije, Universiteit Brussel (VUB), Brussels, Belgium
- icometrix, Leuven, Belgium
| | - Maria Bjerke
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Neuroprotection and Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije, Universiteit Brussel (VUB), Brussels, Belgium
- Department of Clinical Chemistry, Laboratory of Neurochemistry, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Guy Nagels
- Department of Neurology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- St. Edmund Hall, University of Oxford, Oxford, UK
- AIMS lab, Center for Neurosciences (C4N), Vrije Universiteit Brussel, UZ Brussel, Brussels, Belgium
| | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- Department of Neurology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.
- Neuroprotection and Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije, Universiteit Brussel (VUB), Brussels, Belgium.
| |
Collapse
|
20
|
Skampardoni I, Nasrallah IM, Abdulkadir A, Wen J, Melhem R, Mamourian E, Erus G, Doshi J, Singh A, Yang Z, Cui Y, Hwang G, Ren Z, Pomponio R, Srinivasan D, Govindarajan ST, Parmpi P, Wittfeld K, Grabe HJ, Bülow R, Frenzel S, Tosun D, Bilgel M, An Y, Marcus DS, LaMontagne P, Heckbert SR, Austin TR, Launer LJ, Sotiras A, Espeland MA, Masters CL, Maruff P, Fripp J, Johnson SC, Morris JC, Albert MS, Bryan RN, Yaffe K, Völzke H, Ferrucci L, Benzinger TLS, Ezzati A, Shinohara RT, Fan Y, Resnick SM, Habes M, Wolk D, Shou H, Nikita K, Davatzikos C. Genetic and Clinical Correlates of AI-Based Brain Aging Patterns in Cognitively Unimpaired Individuals. JAMA Psychiatry 2024; 81:456-467. [PMID: 38353984 PMCID: PMC10867779 DOI: 10.1001/jamapsychiatry.2023.5599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 11/29/2023] [Indexed: 02/17/2024]
Abstract
Importance Brain aging elicits complex neuroanatomical changes influenced by multiple age-related pathologies. Understanding the heterogeneity of structural brain changes in aging may provide insights into preclinical stages of neurodegenerative diseases. Objective To derive subgroups with common patterns of variation in participants without diagnosed cognitive impairment (WODCI) in a data-driven manner and relate them to genetics, biomedical measures, and cognitive decline trajectories. Design, Setting, and Participants Data acquisition for this cohort study was performed from 1999 to 2020. Data consolidation and harmonization were conducted from July 2017 to July 2021. Age-specific subgroups of structural brain measures were modeled in 4 decade-long intervals spanning ages 45 to 85 years using a deep learning, semisupervised clustering method leveraging generative adversarial networks. Data were analyzed from July 2021 to February 2023 and were drawn from the Imaging-Based Coordinate System for Aging and Neurodegenerative Diseases (iSTAGING) international consortium. Individuals WODCI at baseline spanning ages 45 to 85 years were included, with greater than 50 000 data time points. Exposures Individuals WODCI at baseline scan. Main Outcomes and Measures Three subgroups, consistent across decades, were identified within the WODCI population. Associations with genetics, cardiovascular risk factors (CVRFs), amyloid β (Aβ), and future cognitive decline were assessed. Results In a sample of 27 402 individuals (mean [SD] age, 63.0 [8.3] years; 15 146 female [55%]) WODCI, 3 subgroups were identified in contrast with the reference group: a typical aging subgroup, A1, with a specific pattern of modest atrophy and white matter hyperintensity (WMH) load, and 2 accelerated aging subgroups, A2 and A3, with characteristics that were more distinct at age 65 years and older. A2 was associated with hypertension, WMH, and vascular disease-related genetic variants and was enriched for Aβ positivity (ages ≥65 years) and apolipoprotein E (APOE) ε4 carriers. A3 showed severe, widespread atrophy, moderate presence of CVRFs, and greater cognitive decline. Genetic variants associated with A1 were protective for WMH (rs7209235: mean [SD] B = -0.07 [0.01]; P value = 2.31 × 10-9) and Alzheimer disease (rs72932727: mean [SD] B = 0.1 [0.02]; P value = 6.49 × 10-9), whereas the converse was observed for A2 (rs7209235: mean [SD] B = 0.1 [0.01]; P value = 1.73 × 10-15 and rs72932727: mean [SD] B = -0.09 [0.02]; P value = 4.05 × 10-7, respectively); variants in A3 were associated with regional atrophy (rs167684: mean [SD] B = 0.08 [0.01]; P value = 7.22 × 10-12) and white matter integrity measures (rs1636250: mean [SD] B = 0.06 [0.01]; P value = 4.90 × 10-7). Conclusions and Relevance The 3 subgroups showed distinct associations with CVRFs, genetics, and subsequent cognitive decline. These subgroups likely reflect multiple underlying neuropathologic processes and affect susceptibility to Alzheimer disease, paving pathways toward patient stratification at early asymptomatic stages and promoting precision medicine in clinical trials and health care.
Collapse
Affiliation(s)
- Ioanna Skampardoni
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- School of Electrical and Computer Engineering, National Technical University of Athens, Greece
| | - Ilya M Nasrallah
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Department of Radiology, University of Pennsylvania, Philadelphia
| | - Ahmed Abdulkadir
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Junhao Wen
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Laboratory of AI and Biomedical Science, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles
| | - Randa Melhem
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Elizabeth Mamourian
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Guray Erus
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Jimit Doshi
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Ashish Singh
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Zhijian Yang
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Yuhan Cui
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Gyujoon Hwang
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Zheng Ren
- Laboratory of AI and Biomedical Science, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles
| | - Raymond Pomponio
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Dhivya Srinivasan
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | | | - Paraskevi Parmpi
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Centre for Neurodegenerative Diseases, Site Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Centre for Neurodegenerative Diseases, Site Greifswald, Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, University of Washington, Seattle
- Department of Epidemiology, University of Washington, Seattle
| | - Thomas R Austin
- Cardiovascular Health Research Unit, University of Washington, Seattle
- Department of Epidemiology, University of Washington, Seattle
| | - Lenore J Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, Maryland
| | - Aristeidis Sotiras
- Department of Radiology and Institute of Informatics, Washington University in St Louis, St Louis, Missouri
| | - Mark A Espeland
- Sticht Centre for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison
| | - John C Morris
- Knight Alzheimer Disease Research Centre, Washington University in St Louis, St Louis, Missouri
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia
| | - Kristine Yaffe
- Departments of Neurology, Psychiatry and Epidemiology and Biostatistics, University of California San Francisco, San Francisco
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Ali Ezzati
- Department of Neurology, University of California, Irvine
| | - Russell T Shinohara
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia
| | - Yong Fan
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Mohamad Habes
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Neuroimage Analytics Laboratory and the Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio
| | - David Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia
| | - Haochang Shou
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia
| | - Konstantina Nikita
- School of Electrical and Computer Engineering, National Technical University of Athens, Greece
| | - Christos Davatzikos
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| |
Collapse
|
21
|
Dark HE, Paterson C, Daya GN, Peng Z, Duggan MR, Bilgel M, An Y, Moghekar A, Davatzikos C, Resnick SM, Loupy K, Simpson M, Candia J, Mosley T, Coresh J, Palta P, Ferrucci L, Shapiro A, Williams SA, Walker KA. Proteomic Indicators of Health Predict Alzheimer's Disease Biomarker Levels and Dementia Risk. Ann Neurol 2024; 95:260-273. [PMID: 37801487 PMCID: PMC10842994 DOI: 10.1002/ana.26817] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/06/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023]
Abstract
OBJECTIVE Few studies have comprehensively examined how health and disease risk influence Alzheimer's disease (AD) biomarkers. The present study examined the association of 14 protein-based health indicators with plasma and neuroimaging biomarkers of AD and neurodegeneration. METHODS In 706 cognitively normal adults, we examined whether 14 protein-based health indices (ie, SomaSignal® tests) were associated with concurrently measured plasma-based biomarkers of AD pathology (amyloid-β [Aβ]42/40 , tau phosphorylated at threonine-181 [pTau-181]), neuronal injury (neurofilament light chain [NfL]), and reactive astrogliosis (glial fibrillary acidic protein [GFAP]), brain volume, and cortical Aβ and tau. In a separate cohort (n = 11,285), we examined whether protein-based health indicators associated with neurodegeneration also predict 25-year dementia risk. RESULTS Greater protein-based risk for cardiovascular disease, heart failure mortality, and kidney disease was associated with lower Aβ42/40 and higher pTau-181, NfL, and GFAP levels, even in individuals without cardiovascular or kidney disease. Proteomic indicators of body fat percentage, lean body mass, and visceral fat were associated with pTau-181, NfL, and GFAP, whereas resting energy rate was negatively associated with NfL and GFAP. Together, these health indicators predicted 12, 31, 50, and 33% of plasma Aβ42/40 , pTau-181, NfL, and GFAP levels, respectively. Only protein-based measures of cardiovascular risk were associated with reduced regional brain volumes; these measures predicted 25-year dementia risk, even among those without clinically defined cardiovascular disease. INTERPRETATION Subclinical peripheral health may influence AD and neurodegenerative disease processes and relevant biomarker levels, particularly NfL. Cardiovascular health, even in the absence of clinically defined disease, plays a central role in brain aging and dementia. ANN NEUROL 2024;95:260-273.
Collapse
Affiliation(s)
- Heather E. Dark
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | | | - Gulzar N. Daya
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Michael R. Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christos Davatzikos
- Section of Biomedical Image Analysis, Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | | | | | - Julián Candia
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - Thomas Mosley
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Priya Palta
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Department of Epidemiology, Columbia Mailman School of Public Health, New York, New York, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - Allison Shapiro
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus
| | | | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
22
|
Wen J, Antoniades M, Yang Z, Hwang G, Skampardoni I, Wang R, Davatzikos C. Dimensional Neuroimaging Endophenotypes: Neurobiological Representations of Disease Heterogeneity Through Machine Learning. ARXIV 2024:arXiv:2401.09517v1. [PMID: 38313197 PMCID: PMC10836087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Machine learning has been increasingly used to obtain individualized neuroimaging signatures for disease diagnosis, prognosis, and response to treatment in neuropsychiatric and neurodegenerative disorders. Therefore, it has contributed to a better understanding of disease heterogeneity by identifying disease subtypes that present significant differences in various brain phenotypic measures. In this review, we first present a systematic literature overview of studies using machine learning and multimodal MRI to unravel disease heterogeneity in various neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease, schizophrenia, major depressive disorder, autism spectrum disorder, multiple sclerosis, as well as their potential in transdiagnostic settings. Subsequently, we summarize relevant machine learning methodologies and discuss an emerging paradigm which we call dimensional neuroimaging endophenotype (DNE). DNE dissects the neurobiological heterogeneity of neuropsychiatric and neurodegenerative disorders into a low-dimensional yet informative, quantitative brain phenotypic representation, serving as a robust intermediate phenotype (i.e., endophenotype) largely reflecting underlying genetics and etiology. Finally, we discuss the potential clinical implications of the current findings and envision future research avenues.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Mathilde Antoniades
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Watertown Plank Rd, Milwaukee, WI, USA
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rongguang Wang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
23
|
Yang Z, Wen J, Abdulkadir A, Cui Y, Erus G, Mamourian E, Melhem R, Srinivasan D, Govindarajan ST, Chen J, Habes M, Masters CL, Maruff P, Fripp J, Ferrucci L, Albert MS, Johnson SC, Morris JC, LaMontagne P, Marcus DS, Benzinger TLS, Wolk DA, Shen L, Bao J, Resnick SM, Shou H, Nasrallah IM, Davatzikos C. Gene-SGAN: discovering disease subtypes with imaging and genetic signatures via multi-view weakly-supervised deep clustering. Nat Commun 2024; 15:354. [PMID: 38191573 PMCID: PMC10774282 DOI: 10.1038/s41467-023-44271-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Disease heterogeneity has been a critical challenge for precision diagnosis and treatment, especially in neurologic and neuropsychiatric diseases. Many diseases can display multiple distinct brain phenotypes across individuals, potentially reflecting disease subtypes that can be captured using MRI and machine learning methods. However, biological interpretability and treatment relevance are limited if the derived subtypes are not associated with genetic drivers or susceptibility factors. Herein, we describe Gene-SGAN - a multi-view, weakly-supervised deep clustering method - which dissects disease heterogeneity by jointly considering phenotypic and genetic data, thereby conferring genetic correlations to the disease subtypes and associated endophenotypic signatures. We first validate the generalizability, interpretability, and robustness of Gene-SGAN in semi-synthetic experiments. We then demonstrate its application to real multi-site datasets from 28,858 individuals, deriving subtypes of Alzheimer's disease and brain endophenotypes associated with hypertension, from MRI and single nucleotide polymorphism data. Derived brain phenotypes displayed significant differences in neuroanatomical patterns, genetic determinants, biological and clinical biomarkers, indicating potentially distinct underlying neuropathologic processes, genetic drivers, and susceptibility factors. Overall, Gene-SGAN is broadly applicable to disease subtyping and endophenotype discovery, and is herein tested on disease-related, genetically-associated neuroimaging phenotypes.
Collapse
Affiliation(s)
- Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Ahmed Abdulkadir
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja T Govindarajan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiong Chen
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Biggs Alzheimer's Institute, University of Texas San Antonio Health Science Center, San Antonio, TX, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, QLD, Australia
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel S Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Oxenford S, Ríos AS, Hollunder B, Neudorfer C, Boutet A, Elias GJB, Germann J, Loh A, Deeb W, Salvato B, Almeida L, Foote KD, Amaral R, Rosenberg PB, Tang-Wai DF, Wolk DA, Burke AD, Sabbagh MN, Salloway S, Chakravarty MM, Smith GS, Lyketsos CG, Okun MS, Anderson WS, Mari Z, Ponce FA, Lozano A, Neumann WJ, Al-Fatly B, Horn A. WarpDrive: Improving spatial normalization using manual refinements. Med Image Anal 2024; 91:103041. [PMID: 38007978 PMCID: PMC10842752 DOI: 10.1016/j.media.2023.103041] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023]
Abstract
Spatial normalization-the process of mapping subject brain images to an average template brain-has evolved over the last 20+ years into a reliable method that facilitates the comparison of brain imaging results across patients, centers & modalities. While overall successful, sometimes, this automatic process yields suboptimal results, especially when dealing with brains with extensive neurodegeneration and atrophy patterns, or when high accuracy in specific regions is needed. Here we introduce WarpDrive, a novel tool for manual refinements of image alignment after automated registration. We show that the tool applied in a cohort of patients with Alzheimer's disease who underwent deep brain stimulation surgery helps create more accurate representations of the data as well as meaningful models to explain patient outcomes. The tool is built to handle any type of 3D imaging data, also allowing refinements in high-resolution imaging, including histology and multiple modalities to precisely aggregate multiple data sources together.
Collapse
Affiliation(s)
- Simón Oxenford
- Movement Disorders and Neuromodulation Unit, Department of Neurology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Ana Sofía Ríos
- Movement Disorders and Neuromodulation Unit, Department of Neurology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Barbara Hollunder
- Movement Disorders and Neuromodulation Unit, Department of Neurology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Einstein Center for Neurosciences Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Clemens Neudorfer
- Movement Disorders and Neuromodulation Unit, Department of Neurology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Brain Modulation Lab, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, United States; Center for Brain Circuit Therapeutics Department of Neurology Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Alexandre Boutet
- Division of Neurosurgery, Department of Surgery, University Health Network and University of Toronto, Toronto, ON M5T2S8, Canada; Krembil Research Institute, University of Toronto, Toronto, ON M5T2S8, Canada; Joint Department of Medical Imaging, University of Toronto, Toronto, ON M5T1W7, Canada
| | - Gavin J B Elias
- Division of Neurosurgery, Department of Surgery, University Health Network and University of Toronto, Toronto, ON M5T2S8, Canada; Krembil Research Institute, University of Toronto, Toronto, ON M5T2S8, Canada
| | - Jurgen Germann
- Division of Neurosurgery, Department of Surgery, University Health Network and University of Toronto, Toronto, ON M5T2S8, Canada; Krembil Research Institute, University of Toronto, Toronto, ON M5T2S8, Canada
| | - Aaron Loh
- Division of Neurosurgery, Department of Surgery, University Health Network and University of Toronto, Toronto, ON M5T2S8, Canada; Krembil Research Institute, University of Toronto, Toronto, ON M5T2S8, Canada
| | - Wissam Deeb
- UMass Chan Medical School, Department of Neurology, Worcester, MA 01655, United States; UMass Memorial Health, Department of Neurology, Worcester, MA 01655, United States
| | - Bryan Salvato
- University of Florida Health Jacksonville, Jacksonville, FL, United States
| | - Leonardo Almeida
- Department of Neurology, University of Minnesota, Twin Cities Campus, Minneapolis, MN, United States
| | - Kelly D Foote
- Norman Fixel Institute for Neurological Diseases, Departments of Neurology and Neurosurgery, University of Florida, Gainesville, FL, United States
| | - Robert Amaral
- Cerebral Imaging Centre, Douglas Research Centre, Montreal, QC, Canada
| | - Paul B Rosenberg
- Department of Psychiatry and Behavioral Sciences and Richman Family Precision Medicine Center of Excellence, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - David F Tang-Wai
- Krembil Research Institute, University of Toronto, Toronto, ON M5T2S8, Canada; Department of Medicine, Division of Neurology, University Health Network and University of Toronto, Toronto, ON M5T2S8, Canada
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
| | - Anna D Burke
- Barrow Neurological Institute, Phoenix, AZ, United States
| | | | - Stephen Salloway
- Department of Psychiatry and Human Behavior and Neurology, Alpert Medical School of Brown University, Providence, RI, United States; Memory & Aging Program, Butler Hospital, Providence, United States
| | - M Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Research Centre, Montreal, QC, Canada; Department of Psychiatry, McGill University, Montreal, QC, Canada; Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Gwenn S Smith
- Cerebral Imaging Centre, Douglas Research Centre, Montreal, QC, Canada
| | | | - Michael S Okun
- Norman Fixel Institute for Neurological Diseases, Departments of Neurology and Neurosurgery, University of Florida, Gainesville, FL, United States
| | | | - Zoltan Mari
- Johns Hopkins School of Medicine, Baltimore, MD, United States; Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
| | | | - Andres Lozano
- Division of Neurosurgery, Department of Surgery, University Health Network and University of Toronto, Toronto, ON M5T2S8, Canada; Krembil Research Institute, University of Toronto, Toronto, ON M5T2S8, Canada
| | - Wolf-Julian Neumann
- Movement Disorders and Neuromodulation Unit, Department of Neurology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bassam Al-Fatly
- Movement Disorders and Neuromodulation Unit, Department of Neurology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Horn
- Movement Disorders and Neuromodulation Unit, Department of Neurology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Brain Modulation Lab, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, United States; Center for Brain Circuit Therapeutics Department of Neurology Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
25
|
Zhu Y, Wu Y, Lv X, Wu J, Shen C, Tang Q, Wang G. The relationship between APOE genotype, CSF Tau and cognition across the Alzheimer's disease spectrum, moderation and mediation role of insula network connectivity. CNS Neurosci Ther 2024; 30:e14401. [PMID: 37577852 PMCID: PMC10805399 DOI: 10.1111/cns.14401] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/07/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023] Open
Abstract
AIMS To investigate whether insula network connectivity modulates the relationship between apolipoprotein E (APOE) ε4 genotype, cerebrospinal fluid (CSF) biomarkers (Aβ, Tau, and pTau) and cognition across Alzheimer's disease (AD) spectrum. METHODS Forty-six cognitive normal (CN), 35 subjective memory complaint (SMC), 41 mild cognitive impairment (MCI), and 32 AD subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were obtained. Multivariable linear regression analyses were conducted to investigate the main effects and interaction of the APOE genotype and disease status on the insula functional connectivity (IFC) network. Mediation and moderation analysis were performed to investigate whether IFC strengths regulate the association between APOE genotype, CSF biomarkers and cognition. Additionally, the support vector machine (SVM) model integrating APOE genotype, CSF biomarkers, and neuroimaging biomarkers (insula volumes and altered regional IFCs) was used to classify the AD spectrum. RESULTS The interactive effect of the APOE genotype and disease on the insula network was found in the left medial superior frontal gyrus (SFGmed.L), right anterior medial prefrontal cortex (aMPFC.R), and bilateral thalamus (THA.B). The functional connectivities (FCs) in the left insula (LIns) connecting with the left posterior middle temporal gyrus (pMTG.L), SFGmed.L, and right lingual gyrus (LING.R) were correlated with cognition. LIns-SFGmed.L and LIns-pMTG.L FCs could moderate the effects of Tau on cognition. Furthermore, LIns-SFGmed.L FC may suppress the association between APOE genotype and cognition. More importantly, the integrated biomarkers from the SVM model yielded strong powers for classifying the AD spectrum. CONCLUSIONS Insula functional connectivity regulated the association between APOE genotype, CSF Tau and cognition and provided stage-dependent biomarkers for early differentiation of the AD spectrum. The present study used a cross-sectional design. Follow-up studies are needed to validate the relationship.
Collapse
Affiliation(s)
- Yao Zhu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Yan Wu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Xinyi Lv
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Jiaonan Wu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Chunzi Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Qiqiang Tang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Guoping Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | | |
Collapse
|
26
|
Yang Z, Wen J, Erus G, Govindarajan ST, Melhem R, Mamourian E, Cui Y, Srinivasan D, Abdulkadir A, Parmpi P, Wittfeld K, Grabe HJ, Bülow R, Frenzel S, Tosun D, Bilgel M, An Y, Yi D, Marcus DS, LaMontagne P, Benzinger TL, Heckbert SR, Austin TR, Waldstein SR, Evans MK, Zonderman AB, Launer LJ, Sotiras A, Espeland MA, Masters CL, Maruff P, Fripp J, Toga A, O’Bryant S, Chakravarty MM, Villeneuve S, Johnson SC, Morris JC, Albert MS, Yaffe K, Völzke H, Ferrucci L, Bryan NR, Shinohara RT, Fan Y, Habes M, Lalousis PA, Koutsouleris N, Wolk DA, Resnick SM, Shou H, Nasrallah IM, Davatzikos C. Five dominant dimensions of brain aging are identified via deep learning: associations with clinical, lifestyle, and genetic measures. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.29.23300642. [PMID: 38234857 PMCID: PMC10793523 DOI: 10.1101/2023.12.29.23300642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Brain aging is a complex process influenced by various lifestyle, environmental, and genetic factors, as well as by age-related and often co-existing pathologies. MRI and, more recently, AI methods have been instrumental in understanding the neuroanatomical changes that occur during aging in large and diverse populations. However, the multiplicity and mutual overlap of both pathologic processes and affected brain regions make it difficult to precisely characterize the underlying neurodegenerative profile of an individual from an MRI scan. Herein, we leverage a state-of-the art deep representation learning method, Surreal-GAN, and present both methodological advances and extensive experimental results that allow us to elucidate the heterogeneity of brain aging in a large and diverse cohort of 49,482 individuals from 11 studies. Five dominant patterns of neurodegeneration were identified and quantified for each individual by their respective (herein referred to as) R-indices. Significant associations between R-indices and distinct biomedical, lifestyle, and genetic factors provide insights into the etiology of observed variances. Furthermore, baseline R-indices showed predictive value for disease progression and mortality. These five R-indices contribute to MRI-based precision diagnostics, prognostication, and may inform stratification into clinical trials.
Collapse
Affiliation(s)
- Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja T. Govindarajan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Paraskevi Parmpi
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University of Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Daniel S. Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R. Austin
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Shari R. Waldstein
- Department of Psychology, University of Maryland, Baltimore County, Catonsville, MD, USA
| | - Michele K. Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Alan B. Zonderman
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Lenore J. Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute of Informatics, Washington University in St. Luis, St. Luis, MO63110, USA
| | - Mark A. Espeland
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Colin L. Masters
- Florey Institute, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Paul Maruff
- Florey Institute, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Arthur Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Sid O’Bryant
- Institute for Translational Research University of North Texas Health Science Center Fort Worth Texas USA
| | - Mallar M. Chakravarty
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Yaffe
- Departments of Neurology, Psychiatry and Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, USA
| | - Nick R. Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Russell T. Shinohara
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Biggs Alzheimer’s Institute, University of Texas San Antonio Health Science Center, USA
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Nikolaos Koutsouleris
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- Section for Precision Psychiatry, Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - David A. Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M. Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
27
|
Wen J, Nasrallah IM, Abdulkadir A, Satterthwaite TD, Yang Z, Erus G, Robert-Fitzgerald T, Singh A, Sotiras A, Boquet-Pujadas A, Mamourian E, Doshi J, Cui Y, Srinivasan D, Skampardoni I, Chen J, Hwang G, Bergman M, Bao J, Veturi Y, Zhou Z, Yang S, Dazzan P, Kahn RS, Schnack HG, Zanetti MV, Meisenzahl E, Busatto GF, Crespo-Facorro B, Pantelis C, Wood SJ, Zhuo C, Shinohara RT, Gur RC, Gur RE, Koutsouleris N, Wolf DH, Saykin AJ, Ritchie MD, Shen L, Thompson PM, Colliot O, Wittfeld K, Grabe HJ, Tosun D, Bilgel M, An Y, Marcus DS, LaMontagne P, Heckbert SR, Austin TR, Launer LJ, Espeland M, Masters CL, Maruff P, Fripp J, Johnson SC, Morris JC, Albert MS, Bryan RN, Resnick SM, Fan Y, Habes M, Wolk D, Shou H, Davatzikos C. Genomic loci influence patterns of structural covariance in the human brain. Proc Natl Acad Sci U S A 2023; 120:e2300842120. [PMID: 38127979 PMCID: PMC10756284 DOI: 10.1073/pnas.2300842120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023] Open
Abstract
Normal and pathologic neurobiological processes influence brain morphology in coordinated ways that give rise to patterns of structural covariance (PSC) across brain regions and individuals during brain aging and diseases. The genetic underpinnings of these patterns remain largely unknown. We apply a stochastic multivariate factorization method to a diverse population of 50,699 individuals (12 studies and 130 sites) and derive data-driven, multi-scale PSCs of regional brain size. PSCs were significantly correlated with 915 genomic loci in the discovery set, 617 of which are newly identified, and 72% were independently replicated. Key pathways influencing PSCs involve reelin signaling, apoptosis, neurogenesis, and appendage development, while pathways of breast cancer indicate potential interplays between brain metastasis and PSCs associated with neurodegeneration and dementia. Using support vector machines, multi-scale PSCs effectively derive imaging signatures of several brain diseases. Our results elucidate genetic and biological underpinnings that influence structural covariance patterns in the human brain.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science, Department of Neurology, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Ilya M. Nasrallah
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA19104
| | - Ahmed Abdulkadir
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Theodore D. Satterthwaite
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Zhijian Yang
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Guray Erus
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Timothy Robert-Fitzgerald
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Ashish Singh
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Aristeidis Sotiras
- Department of Radiology, Washington University School of Medicine, St. Louis, MO63110
| | - Aleix Boquet-Pujadas
- Biomedical Imaging Group, Department of Biomedical Engineering, École Polytechnique Fédérale de Lausanne, Lausanne1015, Switzerland
| | - Elizabeth Mamourian
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Jimit Doshi
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Yuhan Cui
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Dhivya Srinivasan
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Ioanna Skampardoni
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Jiong Chen
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Gyujoon Hwang
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Mark Bergman
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| | - Yogasudha Veturi
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Zhen Zhou
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Shu Yang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| | - Paola Dazzan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, LondonWC2R 2LS, United Kingdom
| | - Rene S. Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Hugo G. Schnack
- Department of Psychiatry, University Medical Center Utrecht, Utrecht 3584 CX Ut, Netherlands
| | - Marcus V. Zanetti
- Institute of Psychiatry, Department of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo05508-070, Brazil
| | - Eva Meisenzahl
- Department of Psychiatry and Psychotherapy, Heinrich Heine University, Düsseldorf40204, Germany
| | - Geraldo F. Busatto
- Institute of Psychiatry, Department of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo05508-070, Brazil
| | - Benedicto Crespo-Facorro
- Hospital Universitario Virgen del Rocio, School of Medicine, University of Sevilla,Sevilla41004, Spain
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Stephen J. Wood
- Orygen and the Centre for Youth Mental Health, Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Chuanjun Zhuo
- Key Laboratory of Real Tine Tracing of Brain Circuits in Psychiatry and Neurology, Department of Psychiatry, Tianjin Medical University, Tianjin300070, China
| | - Russell T. Shinohara
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Ruben C. Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Raquel E. Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Nikolaos Koutsouleris
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilian University, Munich 80539, Germany
| | - Daniel H. Wolf
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Andrew J. Saykin
- Indiana Alzheimer’s Disease Research Center, Department of Radiology, Indiana University School of Medicine, Indianapolis, IN46202-3082
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| | - Paul M. Thompson
- Imaging Genetics Center, Department of Neurology, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Olivier Colliot
- Institut du Cerveau, Sorbonne Université, Paris75013, France
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases, University Medicine Greifswald, Greifswald17475, Germany
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases, University Medicine Greifswald, Greifswald17475, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94143
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore21224, MD
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore21224, MD
| | - Daniel S. Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO63110
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO63110
| | - Susan R. Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA98195
| | - Thomas R. Austin
- Department of Epidemiology, University of Washington, Seattle, WA98195
| | - Lenore J. Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Washington, MD20817
| | - Mark Espeland
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Divisions of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC27101
| | - Colin L. Masters
- Florey Institute of Neuroscience and Mental Health, Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC3010, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC3010, Australia
| | - Jurgen Fripp
- Health and Biosecurity, Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD4029, Australia
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Institute, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI53792
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Department of Neurology, Washington University in St. Louis, St. Louis, MO63110
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - R. Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA19104
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore21224, MD
| | - Yong Fan
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Mohamad Habes
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX78229
| | - David Wolk
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA19104
| | - Haochang Shou
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Christos Davatzikos
- AI in Biomedical Imaging Laboratory, Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
28
|
Dempsey DA, Deardorff R, Wu YC, Yu M, Apostolova LG, Brosch J, Clark DG, Farlow MR, Gao S, Wang S, Saykin AJ, Risacher SL. BrainAGE Estimation: Influence of Field Strength, Voxel Size, Race, and Ethnicity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.05.23299222. [PMID: 38106123 PMCID: PMC10723496 DOI: 10.1101/2023.12.05.23299222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The BrainAGE method is used to estimate biological brain age using structural neuroimaging. However, the stability of the model across different scan parameters and races/ethnicities has not been thoroughly investigated. Estimated brain age was compared within- and across- MRI field strength and across voxel sizes. Estimated brain age gap (BAG) was compared across demographically matched groups of different self-reported races and ethnicities in ADNI and IMAS cohorts. Longitudinal ComBat was used to correct for potential scanner effects. The brain age method was stable within field strength, but less stable across different field strengths. The method was stable across voxel sizes. There was a significant difference in BAG between races, but not ethnicities. Correction procedures are suggested to eliminate variation across scanner field strength while maintaining accurate brain age estimation. Further studies are warranted to determine the factors contributing to racial differences in BAG.
Collapse
Affiliation(s)
- Desarae A. Dempsey
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Rachael Deardorff
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yu-Chien Wu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Meichen Yu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Liana G. Apostolova
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jared Brosch
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - David G. Clark
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Martin R. Farlow
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sujuan Gao
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sophia Wang
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | | |
Collapse
|
29
|
Borchert RJ, Azevedo T, Badhwar A, Bernal J, Betts M, Bruffaerts R, Burkhart MC, Dewachter I, Gellersen HM, Low A, Lourida I, Machado L, Madan CR, Malpetti M, Mejia J, Michopoulou S, Muñoz-Neira C, Pepys J, Peres M, Phillips V, Ramanan S, Tamburin S, Tantiangco HM, Thakur L, Tomassini A, Vipin A, Tang E, Newby D, Ranson JM, Llewellyn DJ, Veldsman M, Rittman T. Artificial intelligence for diagnostic and prognostic neuroimaging in dementia: A systematic review. Alzheimers Dement 2023; 19:5885-5904. [PMID: 37563912 DOI: 10.1002/alz.13412] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/18/2023] [Accepted: 06/02/2023] [Indexed: 08/12/2023]
Abstract
INTRODUCTION Artificial intelligence (AI) and neuroimaging offer new opportunities for diagnosis and prognosis of dementia. METHODS We systematically reviewed studies reporting AI for neuroimaging in diagnosis and/or prognosis of cognitive neurodegenerative diseases. RESULTS A total of 255 studies were identified. Most studies relied on the Alzheimer's Disease Neuroimaging Initiative dataset. Algorithmic classifiers were the most commonly used AI method (48%) and discriminative models performed best for differentiating Alzheimer's disease from controls. The accuracy of algorithms varied with the patient cohort, imaging modalities, and stratifiers used. Few studies performed validation in an independent cohort. DISCUSSION The literature has several methodological limitations including lack of sufficient algorithm development descriptions and standard definitions. We make recommendations to improve model validation including addressing key clinical questions, providing sufficient description of AI methods and validating findings in independent datasets. Collaborative approaches between experts in AI and medicine will help achieve the promising potential of AI tools in practice. HIGHLIGHTS There has been a rapid expansion in the use of machine learning for diagnosis and prognosis in neurodegenerative disease Most studies (71%) relied on the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset with no other individual dataset used more than five times There has been a recent rise in the use of more complex discriminative models (e.g., neural networks) that performed better than other classifiers for classification of AD vs healthy controls We make recommendations to address methodological considerations, addressing key clinical questions, and validation We also make recommendations for the field more broadly to standardize outcome measures, address gaps in the literature, and monitor sources of bias.
Collapse
Affiliation(s)
- Robin J Borchert
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Tiago Azevedo
- Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | - AmanPreet Badhwar
- Department of Pharmacology and Physiology, University of Montreal, Montreal, Canada
- Centre de recherche de l'Institut Universitaire de Gériatrie (CRIUGM), Montreal, Canada
| | - Jose Bernal
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Matthew Betts
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - Rose Bruffaerts
- Computational Neurology, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | | | - Ilse Dewachter
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Helena M Gellersen
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Audrey Low
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | | | - Luiza Machado
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Jhony Mejia
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, Colombia
| | - Sofia Michopoulou
- Imaging Physics, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Carlos Muñoz-Neira
- Research into Memory, Brain sciences and dementia Group (ReMemBr Group), Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Jack Pepys
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Marion Peres
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Siddharth Ramanan
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Lokendra Thakur
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, UK
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alessandro Tomassini
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | | | - Eugene Tang
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Danielle Newby
- Department of Psychiatry, University of Oxford, Oxford, UK
| | | | - David J Llewellyn
- University of Exeter Medical School, Exeter, UK
- Alan Turing Institute, London, UK
| | - Michele Veldsman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
Choi US, Park JY, Lee JJ, Choi KY, Won S, Lee KH. Predicting mild cognitive impairments from cognitively normal brains using a novel brain age estimation model based on structural magnetic resonance imaging. Cereb Cortex 2023; 33:10858-10866. [PMID: 37718166 DOI: 10.1093/cercor/bhad331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/19/2023] Open
Abstract
Brain age prediction is a practical method used to quantify brain aging and detect neurodegenerative diseases such as Alzheimer's disease (AD). However, very few studies have considered brain age prediction as a biomarker for the conversion of cognitively normal (CN) to mild cognitive impairment (MCI). In this study, we developed a novel brain age prediction model using brain volume and cortical thickness features. We calculated an acceleration of brain age (ABA) derived from the suggested model to estimate different diagnostic groups (CN, MCI, and AD) and to classify CN to MCI and MCI to AD conversion groups. We observed a strong association between ABA and the 3 diagnostic groups. Additionally, the classification models for CN to MCI conversion and MCI to AD conversion exhibited acceptable and robust performances, with area under the curve values of 0.66 and 0.76, respectively. We believe that our proposed model provides a reliable estimate of brain age for elderly individuals and can identify those at risk of progressing from CN to MCI. This model has great potential to reveal a diagnosis associated with a change in cognitive decline.
Collapse
Affiliation(s)
- Uk-Su Choi
- Gwangju Alzheimer's and Related Dementia Cohort Research Center, Chosun University, Gwangju 61452, Republic of Korea
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jun Young Park
- Gwangju Alzheimer's and Related Dementia Cohort Research Center, Chosun University, Gwangju 61452, Republic of Korea
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- Neurozen Inc., Seoul 06168, Republic of Korea
| | - Jang Jae Lee
- Gwangju Alzheimer's and Related Dementia Cohort Research Center, Chosun University, Gwangju 61452, Republic of Korea
| | - Kyu Yeong Choi
- Gwangju Alzheimer's and Related Dementia Cohort Research Center, Chosun University, Gwangju 61452, Republic of Korea
| | - Sungho Won
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
| | - Kun Ho Lee
- Gwangju Alzheimer's and Related Dementia Cohort Research Center, Chosun University, Gwangju 61452, Republic of Korea
- Department of Biomedical Sciences, Chosun University, Gwangju 61452, Republic of Korea
- Korea Brain Research Institute, Daegu 41061, Republic of Korea
| |
Collapse
|
31
|
Yamada S, Ito H, Matsumasa H, Tanikawa M, Ii S, Otani T, Wada S, Oshima M, Watanabe Y, Mase M. Tightened Sulci in the High Convexities as a Noteworthy Feature of Idiopathic Normal Pressure Hydrocephalus. World Neurosurg 2023; 176:e427-e437. [PMID: 37245671 DOI: 10.1016/j.wneu.2023.05.077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 05/30/2023]
Abstract
OBJECTIVE The presence of tightened sulci in the high-convexities (THC) is a key morphological feature for the diagnosis of idiopathic normal pressure hydrocephalus (iNPH), but the exact localization of THC has yet to be defined. The purpose of this study was to define THC and compare its volume, percentage, and index between iNPH patients and healthy controls. METHODS According to the THC definition, the high-convexity part of the subarachnoid space was segmented and measured the volume and percentage from the 3D T1-weighted and T2-weighted magnetic resonance images in 43 patients with iNPH and 138 healthy controls. RESULTS THC was defined as a decrease in the high-convexity part of the subarachnoid space located above the body of the lateral ventricles, with anterior end on the coronal plane perpendicular to the anterior commissure-posterior commissure (AC-PC) line passing through the front edge of the genu of corpus callosum, the posterior end in the bilateral posterior parts of the callosomarginal sulci, and the lateral end at 3 cm from the midline on the coronal plane perpendicular to the AC-PC line passing through the midpoint between AC and PC. Compared to the volume and volume percentage, the high-convexity part of the subarachnoid space volume per ventricular volume ratio < 0.6 was the most detectable index of THC on both 3D T1-weighted and T2-weighted magnetic resonance images. CONCLUSIONS To improve the diagnostic accuracy of iNPH, the definition of THC was clarified, and high-convexity part of the subarachnoid space volume per ventricular volume ratio <0.6 proposed as the best index for THC detection in this study.
Collapse
Affiliation(s)
- Shigeki Yamada
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, Japan; Interfaculty Initiative in Information Studies/Institute of Industrial Science, The University of Tokyo, Tokyo, Japan; Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan.
| | - Hirotaka Ito
- Medical System Research & Development Center, FUJIFILM Corporation, Tokyo, Japan
| | - Hironori Matsumasa
- Medical System Research & Development Center, FUJIFILM Corporation, Tokyo, Japan
| | - Motoki Tanikawa
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| | - Satoshi Ii
- Faculty of System Design, Tokyo Metropolitan University, Tokyo, Japan
| | - Tomohiro Otani
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Shigeo Wada
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Marie Oshima
- Interfaculty Initiative in Information Studies/Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Yoshiyuki Watanabe
- Department of Radiology, Shiga University of Medical Science, Shiga, Japan
| | - Mitsuhito Mase
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| |
Collapse
|
32
|
Kress GT, Popa ES, Thompson PM, Bookheimer SY, Thomopoulos SI, Ching CRK, Zheng H, Hirsh DA, Merrill DA, Panos SE, Raji CA, Siddarth P, Bramen JE. Preliminary validation of a structural magnetic resonance imaging metric for tracking dementia-related neurodegeneration and future decline. Neuroimage Clin 2023; 39:103458. [PMID: 37421927 PMCID: PMC10338152 DOI: 10.1016/j.nicl.2023.103458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 07/10/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline and atrophy in the medial temporal lobe (MTL) and subsequent brain regions. Structural magnetic resonance imaging (sMRI) has been widely used in research and clinical care for diagnosis and monitoring AD progression. However, atrophy patterns are complex and vary by patient. To address this issue, researchers have made efforts to develop more concise metrics that can summarize AD-specific atrophy. Many of these methods can be difficult to interpret clinically, hampering adoption. In this study, we introduce a novel index which we call an "AD-NeuroScore," that uses a modified Euclidean-inspired distance function to calculate differences between regional brain volumes associated with cognitive decline. The index is adjusted for intracranial volume (ICV), age, sex, and scanner model. We validated AD-NeuroScore using 929 older adults from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study, with a mean age of 72.7 years (SD = 6.3; 55.1-91.5) and cognitively normal (CN), mild cognitive impairment (MCI), or AD diagnoses. Our validation results showed that AD-NeuroScore was significantly associated with diagnosis and disease severity scores (measured by MMSE, CDR-SB, and ADAS-11) at baseline. Furthermore, baseline AD-NeuroScore was associated with both changes in diagnosis and disease severity scores at all time points with available data. The performance of AD-NeuroScore was equivalent or superior to adjusted hippocampal volume (AHV), a widely used metric in AD research. Further, AD-NeuroScore typically performed as well as or sometimes better when compared to other existing sMRI-based metrics. In conclusion, we have introduced a new metric, AD-NeuroScore, which shows promising results in detecting AD, benchmarking disease severity, and predicting disease progression. AD-NeuroScore differentiates itself from other metrics by being clinically practical and interpretable.
Collapse
Affiliation(s)
- Gavin T Kress
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA 90404, USA; Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Emily S Popa
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA 90404, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90292, USA
| | - Susan Y Bookheimer
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA 90404, USA; David Geffen School of Medicine, University of California, Los Angeles, Westwood, CA 90095, USA
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90292, USA
| | - Christopher R K Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90292, USA
| | - Hong Zheng
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90292, USA
| | - Daniel A Hirsh
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA 90404, USA.
| | - David A Merrill
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA 90404, USA; Department of Translational Neurosciences and Neurotherapeutics, Providence Saint John's Cancer Institute, Santa Monica, CA 90404, USA; UCLA Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Westwood, CA 90095, USA
| | - Stella E Panos
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA 90404, USA
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Prabha Siddarth
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA 90404, USA; UCLA Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Westwood, CA 90095, USA
| | - Jennifer E Bramen
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA 90404, USA.
| |
Collapse
|
33
|
Habes M, Jacobson AM, Braffett BH, Rashid T, Ryan CM, Shou H, Cui Y, Davatzikos C, Luchsinger JA, Biessels GJ, Bebu I, Gubitosi-Klug RA, Bryan RN, Nasrallah IM. Patterns of Regional Brain Atrophy and Brain Aging in Middle- and Older-Aged Adults With Type 1 Diabetes. JAMA Netw Open 2023; 6:e2316182. [PMID: 37261829 PMCID: PMC10236234 DOI: 10.1001/jamanetworkopen.2023.16182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/09/2023] [Indexed: 06/02/2023] Open
Abstract
Importance Little is known about structural brain changes in type 1 diabetes (T1D) and whether there are early manifestations of a neurodegenerative condition like Alzheimer disease (AD) or evidence of premature brain aging. Objective To evaluate neuroimaging markers of brain age and AD-like atrophy in participants with T1D in the Diabetes Control and Complications Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications (EDIC) study, identify which brain regions are associated with the greatest changes in patients with T1D, and assess the association between cognition and brain aging indices. Design, Setting, and Participants This cohort study leveraged data collected during the combined DCCT (randomized clinical trial, 1983-1993) and EDIC (observational study, 1994 to present) studies at 27 clinical centers in the US and Canada. A total of 416 eligible EDIC participants and 99 demographically similar adults without diabetes were enrolled in the magnetic resonance imaging (MRI) ancillary study, which reports cross-sectional data collected in 2018 to 2019 and relates it to factors measured longitudinally in DCCT/EDIC. Data analyses were performed between July 2020 and April 2022. Exposure T1D diagnosis. Main Outcomes and Measures Psychomotor and mental efficiency were evaluated using verbal fluency, digit symbol substitution test, trail making part B, and the grooved pegboard. Immediate memory scores were derived from the logical memory subtest of the Wechsler memory scale and the Wechsler digit symbol substitution test. MRI and machine learning indices were calculated to predict brain age and quantify AD-like atrophy. Results This study included 416 EDIC participants with a median (range) age of 60 (44-74) years (87 of 416 [21%] were older than 65 years) and a median (range) diabetes duration of 37 (30-51) years. EDIC participants had consistently higher brain age values compared with controls without diabetes, indicative of approximately 6 additional years of brain aging (EDIC participants: β, 6.16; SE, 0.71; control participants: β, 1.04; SE, 0.04; P < .001). In contrast, AD regional atrophy was comparable between the 2 groups. Regions with atrophy in EDIC participants vs controls were observed mainly in the bilateral thalamus and putamen. Greater brain age was associated with lower psychomotor and mental efficiency among EDIC participants (β, -0.04; SE, 0.01; P < .001), but not among controls. Conclusions and Relevance The findings of this study suggest an increase in brain aging among individuals with T1D without any early signs of AD-related neurodegeneration. These increases were associated with reduced cognitive performance, but overall, the abnormal patterns seen in this sample were modest, even after a mean of 38 years with T1D.
Collapse
Affiliation(s)
- Mohamad Habes
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Alan M. Jacobson
- NYU Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, New York
| | | | - Tanweer Rashid
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | | | - Haochang Shou
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia
| | - Yuhan Cui
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | | | - Geert J. Biessels
- Department of Neurology, UMCU Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ionut Bebu
- George Washington University, Biostatistics Center, Rockville, Maryland
| | - Rose A. Gubitosi-Klug
- Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - R. Nick Bryan
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ilya M. Nasrallah
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | | |
Collapse
|
34
|
Wyss P, Ginsbourger D, Shou H, Davatzikos C, Klöppel S, Abdulkadir A. Adaptive data-driven selection of sequences of biological and cognitive markers in pre-clinical diagnosis of dementia. Sci Rep 2023; 13:6406. [PMID: 37076487 PMCID: PMC10115887 DOI: 10.1038/s41598-023-32867-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/04/2023] [Indexed: 04/21/2023] Open
Abstract
Effective clinical decision procedures must balance multiple competing objectives such as time-to-decision, acquisition costs, and accuracy. We describe and evaluate POSEIDON, a data-driven method for PrOspective SEquentIal DiagnOsis with Neutral zones to individualize clinical classifications. We evaluated the framework with an application in which the algorithm sequentially proposes to include cognitive, imaging, or molecular markers if a sufficiently more accurate prognosis of clinical decline to manifest Alzheimer's disease is expected. Over a wide range of cost parameter data-driven tuning lead to quantitatively lower total cost compared to ad hoc fixed sets of measurements. The classification accuracy based on all longitudinal data from participants that was acquired over 4.8 years on average was 0.89. The sequential algorithm selected 14 percent of available measurements and concluded after an average follow-up time of 0.74 years at the expense of 0.05 lower accuracy. Sequential classifiers were competitive from a multi-objective perspective since they could dominate fixed sets of measurements by making fewer errors using less resources. Nevertheless, the trade-off of competing objectives depends on inherently subjective prescribed cost parameters. Thus, despite the effectiveness of the method, the implementation into consequential clinical applications will remain controversial and evolve around the choice of cost parameters.
Collapse
Affiliation(s)
- Patric Wyss
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - David Ginsbourger
- Institute of Mathematical Statistics and Actuarial Science, University of Bern, Bern, Switzerland
| | - Haochang Shou
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
- Center for Biomedical Image Computing and Analytics (CBICA), Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Stefan Klöppel
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Ahmed Abdulkadir
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA.
- Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
35
|
Bidzan L, Grabowski J, Przybylak M, Ali S. Aggressive behavior and prognosis in patients with mild cognitive impairment. Dement Neuropsychol 2023; 17:e20200096. [PMID: 37223838 PMCID: PMC10202333 DOI: 10.1590/1980-5764-dn-2020-0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/08/2021] [Accepted: 05/07/2021] [Indexed: 05/25/2023] Open
Abstract
The diagnosis of mild cognitive impairment (MCI) is associated with an increased risk of developing dementia. When evaluating the further prognosis of MCI, the occurrence of neuropsychiatric symptoms, particularly aggressive and impulsive behavior, may play an important role. Objective The aim of this study was to evaluate the relationship between aggressive behavior and cognitive dysfunction in patients diagnosed with MCI. Methods The results are based on a 7-year prospective study. At the time of inclusion in the study, participants, recruited from an outpatient clinic, were assessed with Mini-Mental State Examination (MMSE) and the Cohen-Mansfield Agitation Inventory (CMAI). A reassessment was performed after 1 year using the MMSE scale in all patients. The time of next MMSE administration was depended on the clinical condition of patients took place at the end of follow-up, that is, at the time of diagnosis of the dementia or after 7 years from inclusion when the criteria for dementia were not met. Results Of the 193 patients enrolled in the study, 75 were included in the final analysis. Patients who converted to dementia during the observation period exhibited a greater severity of symptoms in each of the assessed CMAI categories. Moreover, there was a significant correlation between the global result of CMAI and the results of the physical nonaggressive and verbal aggressive subscales with cognitive decline during the first year of observation. Conclusions Despite several limitations to the study, aggressive and impulsive behaviors seem to be an unfavorable prognostic factor in the course of MCI.
Collapse
Affiliation(s)
- Leszek Bidzan
- Medical University of Gdańsk, Faculty of Medicine, Department of Developmental, Psychotic and Geriatric Psychiatry, Gdańsk, Poland
| | - Jakub Grabowski
- Medical University of Gdańsk, Faculty of Medicine, Department of Developmental, Psychotic and Geriatric Psychiatry, Gdańsk, Poland
| | - Mateusz Przybylak
- Medical University of Gdańsk, Faculty of Medicine, Department of Developmental, Psychotic and Geriatric Psychiatry, Gdańsk, Poland
| | - Shan Ali
- Medical University of Gdańsk, Faculty of Medicine, Department of Developmental, Psychotic and Geriatric Psychiatry, Adult Psychiatry Student’s Scientific Circle, Gdańsk, Poland
| |
Collapse
|
36
|
Duggan MR, Butler L, Peng Z, Daya GN, Moghekar A, An Y, Rapp SR, Hayden KM, Shadyab AH, Natale G, Liu L, Snetselaar L, Moaddel R, Rebholz CM, Sullivan K, Ballantyne CM, Resnick SM, Ferrucci L, Walker KA. Plasma proteins related to inflammatory diet predict future cognitive impairment. Mol Psychiatry 2023; 28:1599-1609. [PMID: 36737481 PMCID: PMC10208977 DOI: 10.1038/s41380-023-01975-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/05/2023]
Abstract
Dysregulation of the immune system and dietary patterns that increase inflammation can increase the risk for cognitive decline, but the mechanisms by which inflammatory nutritional habits may affect the development of cognitive impairment in aging are not well understood. To determine whether plasma proteins linked to inflammatory diet predict future cognitive impairment, we applied high-throughput proteomic assays to plasma samples from a subset (n = 1528) of Women's Health Initiative Memory Study (WHIMS) participants (mean [SD] baseline age, 71.3 [SD 3.8] years). Results provide insights into how inflammatory nutritional patterns are associated with an immune-related proteome and identify a group of proteins (CXCL10, CCL3, HGF, OPG, CDCP1, NFATC3, ITGA11) related to future cognitive impairment over a 14-year follow-up period. Several of these inflammatory diet proteins were also associated with dementia risk across two external cohorts (ARIC, ESTHER), correlated with plasma biomarkers of Alzheimer's disease (AD) pathology (Aβ42/40) and/or neurodegeneration (NfL), and related to an MRI-defined index of neurodegenerative brain atrophy in a separate cohort (BLSA). In addition to evaluating their biological relevance, assessing their potential role in AD, and characterizing their immune-tissue/cell-specific expression, we leveraged published RNA-seq results to examine how the in vitro regulation of genes encoding these candidate proteins might be altered in response to an immune challenge. Our findings indicate how dietary patterns with higher inflammatory potential relate to plasma levels of immunologically relevant proteins and highlight the molecular mediators which predict subsequent risk for age-related cognitive impairment.
Collapse
Affiliation(s)
- Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Lauren Butler
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Gulzar N Daya
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Stephen R Rapp
- Department of Social Sciences and Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Psychiatry & Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kathleen M Hayden
- Department of Psychiatry & Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Aladdin H Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, USA
| | - Ginny Natale
- Program in Public Health, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Longjian Liu
- Department of Epidemiology and Biostatistics, Drexel University Dornsife School of Public Health, Philadelphia, PA, USA
| | - Linda Snetselaar
- Department of Epidemiology, University of Iowa, Iowa City, IA, USA
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kevin Sullivan
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
37
|
Miller B, Kim SJ, Mehta HH, Cao K, Kumagai H, Thumaty N, Leelaprachakul N, Braniff RG, Jiao H, Vaughan J, Diedrich J, Saghatelian A, Arpawong TE, Crimmins EM, Ertekin-Taner N, Tubi MA, Hare ET, Braskie MN, Décarie-Spain L, Kanoski SE, Grodstein F, Bennett DA, Zhao L, Toga AW, Wan J, Yen K, Cohen P. Mitochondrial DNA variation in Alzheimer's disease reveals a unique microprotein called SHMOOSE. Mol Psychiatry 2023; 28:1813-1826. [PMID: 36127429 PMCID: PMC10027624 DOI: 10.1038/s41380-022-01769-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 08/15/2022] [Accepted: 08/26/2022] [Indexed: 01/22/2023]
Abstract
Mitochondrial DNA variants have previously associated with disease, but the underlying mechanisms have been largely elusive. Here, we report that mitochondrial SNP rs2853499 associated with Alzheimer's disease (AD), neuroimaging, and transcriptomics. We mapped rs2853499 to a novel mitochondrial small open reading frame called SHMOOSE with microprotein encoding potential. Indeed, we detected two unique SHMOOSE-derived peptide fragments in mitochondria by using mass spectrometry-the first unique mass spectrometry-based detection of a mitochondrial-encoded microprotein to date. Furthermore, cerebrospinal fluid (CSF) SHMOOSE levels in humans correlated with age, CSF tau, and brain white matter volume. We followed up on these genetic and biochemical findings by carrying out a series of functional experiments. SHMOOSE acted on the brain following intracerebroventricular administration, differentiated mitochondrial gene expression in multiple models, localized to mitochondria, bound the inner mitochondrial membrane protein mitofilin, and boosted mitochondrial oxygen consumption. Altogether, SHMOOSE has vast implications for the fields of neurobiology, Alzheimer's disease, and microproteins.
Collapse
Affiliation(s)
- Brendan Miller
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Su-Jeong Kim
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Hemal H Mehta
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kevin Cao
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Hiroshi Kumagai
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Neehar Thumaty
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Naphada Leelaprachakul
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Regina Gonzalez Braniff
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Henry Jiao
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Joan Vaughan
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jolene Diedrich
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Thalida E Arpawong
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Eileen M Crimmins
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | | | - Meral A Tubi
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Evan T Hare
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Meredith N Braskie
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Léa Décarie-Spain
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Francine Grodstein
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lu Zhao
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Junxiang Wan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kelvin Yen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
38
|
N-acetyl-aspartate and Myo-inositol as Markers of White Matter Microstructural Organization in Mild Cognitive Impairment: Evidence from a DTI- 1H-MRS Pilot Study. Diagnostics (Basel) 2023; 13:diagnostics13040654. [PMID: 36832141 PMCID: PMC9955118 DOI: 10.3390/diagnostics13040654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
We implemented a multimodal approach to examine associations between structural and neurochemical changes that could signify neurodegenerative processes related to mild cognitive impairment (MCI). Fifty-nine older adults (60-85 years; 22 MCI) underwent whole-brain structural 3T MRI (T1W, T2W, DTI) and proton magnetic resonance spectroscopy (1H-MRS). The regions of interest (ROIs) for 1H-MRS measurements were the dorsal posterior cingulate cortex, left hippocampal cortex, left medial temporal cortex, left primary sensorimotor cortex, and right dorsolateral prefrontal cortex. The findings revealed that subjects in the MCI group showed moderate to strong positive associations between the total N-acetylaspartate to total creatine and the total N-acetylaspartate to myo-inositol ratios in the hippocampus and dorsal posterior cingulate cortex and fractional anisotropy (FA) of WM tracts crossing these regions-specifically, the left temporal tapetum, right corona radiata, and right posterior cingulate gyri. In addition, negative associations between the myo-inositol to total creatine ratio and FA of the left temporal tapetum and right posterior cingulate gyri were observed. These observations suggest that the biochemical integrity of the hippocampus and cingulate cortex is associated with a microstructural organization of ipsilateral WM tracts originating in the hippocampus. Specifically, elevated myo-inositol might be an underlying mechanism for decreased connectivity between the hippocampus and the prefrontal/cingulate cortex in MCI.
Collapse
|
39
|
Huang P, Zhang M. Magnetic Resonance Imaging Studies of Neurodegenerative Disease: From Methods to Translational Research. Neurosci Bull 2023; 39:99-112. [PMID: 35771383 PMCID: PMC9849544 DOI: 10.1007/s12264-022-00905-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/07/2022] [Indexed: 01/22/2023] Open
Abstract
Neurodegenerative diseases (NDs) have become a significant threat to an aging human society. Numerous studies have been conducted in the past decades to clarify their pathologic mechanisms and search for reliable biomarkers. Magnetic resonance imaging (MRI) is a powerful tool for investigating structural and functional brain alterations in NDs. With the advantages of being non-invasive and non-radioactive, it has been frequently used in both animal research and large-scale clinical investigations. MRI may serve as a bridge connecting micro- and macro-level analysis and promoting bench-to-bed translational research. Nevertheless, due to the abundance and complexity of MRI techniques, exploiting their potential is not always straightforward. This review aims to briefly introduce research progress in clinical imaging studies and discuss possible strategies for applying MRI in translational ND research.
Collapse
Affiliation(s)
- Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| |
Collapse
|
40
|
Krämer C, Stumme J, da Costa Campos L, Rubbert C, Caspers J, Caspers S, Jockwitz C. Classification and prediction of cognitive performance differences in older age based on brain network patterns using a machine learning approach. Netw Neurosci 2023; 7:122-147. [PMID: 37339286 PMCID: PMC10270720 DOI: 10.1162/netn_a_00275] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/22/2022] [Indexed: 09/22/2023] Open
Abstract
Age-related cognitive decline varies greatly in healthy older adults, which may partly be explained by differences in the functional architecture of brain networks. Resting-state functional connectivity (RSFC) derived network parameters as widely used markers describing this architecture have even been successfully used to support diagnosis of neurodegenerative diseases. The current study aimed at examining whether these parameters may also be useful in classifying and predicting cognitive performance differences in the normally aging brain by using machine learning (ML). Classifiability and predictability of global and domain-specific cognitive performance differences from nodal and network-level RSFC strength measures were examined in healthy older adults from the 1000BRAINS study (age range: 55-85 years). ML performance was systematically evaluated across different analytic choices in a robust cross-validation scheme. Across these analyses, classification performance did not exceed 60% accuracy for global and domain-specific cognition. Prediction performance was equally low with high mean absolute errors (MAEs ≥ 0.75) and low to none explained variance (R2 ≤ 0.07) for different cognitive targets, feature sets, and pipeline configurations. Current results highlight limited potential of functional network parameters to serve as sole biomarker for cognitive aging and emphasize that predicting cognition from functional network patterns may be challenging.
Collapse
Affiliation(s)
- Camilla Krämer
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Johanna Stumme
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lucas da Costa Campos
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian Rubbert
- Department of Diagnostic and Interventional Radiology, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julian Caspers
- Department of Diagnostic and Interventional Radiology, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Svenja Caspers
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christiane Jockwitz
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
41
|
Toledo JB, Rashid T, Liu H, Launer L, Shaw LM, Heckbert SR, Weiner M, Seshadri S, Habes M. SPARE-Tau: A flortaucipir machine-learning derived early predictor of cognitive decline. PLoS One 2022; 17:e0276392. [PMID: 36327215 PMCID: PMC9632811 DOI: 10.1371/journal.pone.0276392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Recently, tau PET tracers have shown strong associations with clinical outcomes in individuals with cognitive impairment and cognitively unremarkable elderly individuals. flortaucipir PET scans to measure tau deposition in multiple brain areas as the disease progresses. This information needs to be summarized to evaluate disease severity and predict disease progression. We, therefore, sought to develop a machine learning-derived index, SPARE-Tau, which successfully detects pathology in the earliest disease stages and accurately predicts progression compared to a priori-based region of interest approaches (ROI). METHODS 587 participants of the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort had flortaucipir scans, structural MRI scans, and an Aβ biomarker test (CSF or florbetapir PET) performed on the same visit. We derived the SPARE-Tau index in a subset of 367 participants. We evaluated associations with clinical measures for CSF p-tau, SPARE-MRI, and flortaucipir PET indices (SPARE-Tau, meta-temporal, and average Braak ROIs). Bootstrapped multivariate adaptive regression splines linear regression analyzed the association between the biomarkers and baseline ADAS-Cog13 scores. Bootstrapped multivariate linear regression models evaluated associations with clinical diagnosis. Cox-hazards and mixed-effects models investigated clinical progression and longitudinal ADAS-Cog13 changes. The Aβ positive cognitively unremarkable participants, not included in the SPARE-Tau training, served as an independent validation group. RESULTS Compared to CSF p-tau, meta-temporal, and averaged Braak tau PET ROIs, SPARE-Tau showed the strongest association with baseline ADAS-cog13 scores and diagnosis. SPARE-Tau also presented the strongest association with clinical progression in cognitively unremarkable participants and longitudinal ADAS-Cog13 changes. Results were confirmed in the Aβ+ cognitively unremarkable hold-out sample participants. CSF p-tau showed the weakest cross-sectional associations and longitudinal prediction. DISCUSSION Flortaucipir indices showed the strongest clinical association among the studied biomarkers (flortaucipir, florbetapir, structural MRI, and CSF p-tau) and were predictive in the preclinical disease stages. Among the flortaucipir indices, the machine-learning derived SPARE-Tau index was the most sensitive clinical progression biomarker. The combination of different biomarker modalities better predicted cognitive performance.
Collapse
Affiliation(s)
- Jon B. Toledo
- Department of Neurology, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Neurology Houston Methodist Hospital, Houston, Texas, United States of America
| | - Tanweer Rashid
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), San Antonio, Texas, United States of America
| | - Hangfan Liu
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), San Antonio, Texas, United States of America
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lenore Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Susan R. Heckbert
- Department of Epidemiology and Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America
| | - Michael Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, California, United States of America
- Department of Radiology, University of California, San Francisco, California, United States of America
- Department of Medicine, University of California, San Francisco, California, United States of America
- Department of Psychiatry, University of California, San Francisco, California, United States of America
- Department of Neurology, University of California, San Francisco, California, United States of America
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, Texas, United States of America
| | - Mohamad Habes
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), San Antonio, Texas, United States of America
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, Texas, United States of America
| | | |
Collapse
|
42
|
Taylor A, Zhang F, Niu X, Heywood A, Stocks J, Feng G, Popuri K, Beg MF, Wang L. Investigating the temporal pattern of neuroimaging-based brain age estimation as a biomarker for Alzheimer's Disease related neurodegeneration. Neuroimage 2022; 263:119621. [PMID: 36089183 PMCID: PMC9995621 DOI: 10.1016/j.neuroimage.2022.119621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
Neuroimaging-based brain-age estimation via machine learning has emerged as an important new approach for studying brain aging. The difference between one's estimated brain age and chronological age, the brain age gap (BAG), has been proposed as an Alzheimer's Disease (AD) biomarker. However, most past studies on the BAG have been cross-sectional. Quantifying longitudinal changes in an individual's BAG temporal pattern would likely improve prediction of AD progression and clinical outcome based on neurophysiological changes. To fill this gap, our study conducted predictive modeling using a large neuroimaging dataset with up to 8 years of follow-up to examine the temporal patterns of the BAG's trajectory and how it varies by subject-level characteristics (sex, APOEɛ4 carriership) and disease status. Specifically, we explored the pattern and rate of change in BAG over time in individuals who remain stable with normal cognition or mild cognitive impairment (MCI), as well as individuals who progress to clinical AD. Combining multimodal imaging data in a support vector regression model to estimate brain age yielded improved performance over single modality. Multilevel modeling results showed the BAG followed a linear increasing trajectory with a significantly faster rate in individuals with MCI who progressed to AD compared to cognitively normal or MCI individuals who did not progress. The dynamic changes in the BAG during AD progression were further moderated by sex and APOEɛ4 carriership. Our findings demonstrate the BAG as a potential biomarker for understanding individual specific temporal patterns related to AD progression.
Collapse
Affiliation(s)
- Alexei Taylor
- Department of Psychological and Brain Sciences, Drexel University, Philadelphia, PA 19104, USA
| | - Fengqing Zhang
- Department of Psychological and Brain Sciences, Drexel University, Philadelphia, PA 19104, USA.
| | - Xin Niu
- Department of Psychological and Brain Sciences, Drexel University, Philadelphia, PA 19104, USA
| | - Ashley Heywood
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jane Stocks
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gangyi Feng
- Department of Linguistics and Modern Languages, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China; Brain and Mind Institute, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, V6A1S6 BCE, Canada
| | - Lei Wang
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Psychiatry and Behavioral Health, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
43
|
Baloni P, Arnold M, Buitrago L, Nho K, Moreno H, Huynh K, Brauner B, Louie G, Kueider-Paisley A, Suhre K, Saykin AJ, Ekroos K, Meikle PJ, Hood L, Price ND, Doraiswamy PM, Funk CC, Hernández AI, Kastenmüller G, Baillie R, Han X, Kaddurah-Daouk R. Multi-Omic analyses characterize the ceramide/sphingomyelin pathway as a therapeutic target in Alzheimer's disease. Commun Biol 2022; 5:1074. [PMID: 36209301 PMCID: PMC9547905 DOI: 10.1038/s42003-022-04011-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/20/2022] [Indexed: 11/09/2022] Open
Abstract
Dysregulation of sphingomyelin and ceramide metabolism have been implicated in Alzheimer's disease. Genome-wide and transcriptome-wide association studies have identified various genes and genetic variants in lipid metabolism that are associated with Alzheimer's disease. However, the molecular mechanisms of sphingomyelin and ceramide disruption remain to be determined. We focus on the sphingolipid pathway and carry out multi-omics analyses to identify central and peripheral metabolic changes in Alzheimer's patients, correlating them to imaging features. Our multi-omics approach is based on (a) 2114 human post-mortem brain transcriptomics to identify differentially expressed genes; (b) in silico metabolic flux analysis on context-specific metabolic networks identified differential reaction fluxes; (c) multimodal neuroimaging analysis on 1576 participants to associate genetic variants in sphingomyelin pathway with Alzheimer's disease pathogenesis; (d) plasma metabolomic and lipidomic analysis to identify associations of lipid species with dysregulation in Alzheimer's; and (e) metabolite genome-wide association studies to define receptors within the pathway as a potential drug target. We validate our hypothesis in amyloidogenic APP/PS1 mice and show prolonged exposure to fingolimod alleviated synaptic plasticity and cognitive impairment in mice. Our integrative multi-omics approach identifies potential targets in the sphingomyelin pathway and suggests modulators of S1P metabolism as possible candidates for Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Priyanka Baloni
- Institute for Systems Biology, Seattle, WA, USA
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, Durham, NC, USA
| | - Luna Buitrago
- Department of Neurology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Kwangsik Nho
- Indiana Alzheimer's Disease Research Center and Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Herman Moreno
- Department of Neurology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Barbara Brauner
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Gregory Louie
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, Durham, NC, USA
| | - Alexandra Kueider-Paisley
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, Durham, NC, USA
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, PO 24144, Doha, Qatar
| | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center and Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kim Ekroos
- Lipidomics Consulting Ltd., Esbo, Finland
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Leroy Hood
- Institute for Systems Biology, Seattle, WA, USA
| | | | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, Durham, NC, USA
| | - Cory C Funk
- Institute for Systems Biology, Seattle, WA, USA
| | - A Iván Hernández
- Department of Pathology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Xianlin Han
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA.
| |
Collapse
|
44
|
He Q, Shi L, Luo Y, Wan C, Malone IB, Mok VCT, Cole JH, Anatürk M. Validation of the Alzheimer's disease-resemblance atrophy index in classifying and predicting progression in Alzheimer's disease. Front Aging Neurosci 2022; 14:932125. [PMID: 36062150 PMCID: PMC9435378 DOI: 10.3389/fnagi.2022.932125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/14/2022] [Indexed: 12/01/2022] Open
Abstract
Background Automated tools for characterising dementia risk have the potential to aid in the diagnosis, prognosis, and treatment of Alzheimer's disease (AD). Here, we examined a novel machine learning-based brain atrophy marker, the AD-resemblance atrophy index (AD-RAI), to assess its test-retest reliability and further validate its use in disease classification and prediction. Methods Age- and sex-matched 44 probable AD (Age: 69.13 ± 7.13; MMSE: 27-30) and 22 non-demented control (Age: 69.38 ± 7.21; MMSE: 27-30) participants were obtained from the Minimal Interval Resonance Imaging in Alzheimer's Disease (MIRIAD) dataset. Serial T1-weighted images (n = 678) from up to nine time points over a 2-year period, including 179 pairs of back-to-back scans acquired on same participants on the same day and 40 pairs of scans acquired at 2-week intervals were included. All images were automatically processed with AccuBrain® to calculate the AD-RAI. Its same-day repeatability and 2-week reproducibility were first assessed. The discriminative performance of AD-RAI was evaluated using the receiver operating characteristic curve, where DeLong's test was used to evaluate its performance against quantitative medial temporal lobe atrophy (QMTA) and hippocampal volume adjusted by intracranial volume (ICV)-proportions and ICV-residuals methods, respectively (HVR and HRV). Linear mixed-effects modelling was used to investigate longitudinal trajectories of AD-RAI and baseline AD-RAI prediction of cognitive decline. Finally, the longitudinal associations between AD-RAI and MMSE scores were assessed. Results AD-RAI had excellent same-day repeatability and excellent 2-week reproducibility. AD-RAI's AUC (99.8%; 95%CI = [99.3%, 100%]) was equivalent to that of QMTA (96.8%; 95%CI = [92.9%, 100%]), and better than that of HVR (86.8%; 95%CI = [78.2%, 95.4%]) or HRV (90.3%; 95%CI = [83.0%, 97.6%]). While baseline AD-RAI was significantly higher in the AD group, it did not show detectable changes over 2 years. Baseline AD-RAI was negatively associated with MMSE scores and the rate of the change in MMSE scores over time. A negative longitudinal association was also found between AD-RAI values and the MMSE scores among AD patients. Conclusions The AD-RAI represents a potential biomarker that may support AD diagnosis and be used to predict the rate of future cognitive decline in AD patients.
Collapse
Affiliation(s)
- Qiling He
- UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Lin Shi
- Department of Imaging and Interventional Radiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yishan Luo
- BrainNow Research Institute, Shenzhen, China
| | - Chao Wan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ian B. Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Vincent C. T. Mok
- Gerald Choa Neuroscience Centre, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, Division of Neurology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - James H. Cole
- Department of Computer Science, Faculty of Engineering Science, University College London, London, United Kingdom
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Melis Anatürk
- Department of Computer Science, Faculty of Engineering Science, University College London, London, United Kingdom
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Palmer JR, Wang C, Kong D, Cespedes M, Pye J, Hickie IB, Barnett M, Naismith SL. Rest-activity rhythms and tract specific white matter lesions in older adults at risk for cognitive decline. Mol Psychiatry 2022; 27:3410-3416. [PMID: 35764707 PMCID: PMC9708592 DOI: 10.1038/s41380-022-01641-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 03/04/2022] [Accepted: 05/23/2022] [Indexed: 11/09/2022]
Abstract
White matter lesions (WMLs) are common in older adults and represent an important predictor of negative long-term outcomes. Rest-activity rhythm disturbance is also common, however, few studies have investigated associations between these factors. We employed a novel AI-based automatic WML segmentation tool and diffusion-weighted tractography to investigate associations between tract specific WML volumes and non-parametric actigraphy measures in older adults at risk for cognitive decline. The primary non-parametric measures of interest were inter-daily stability (IS), intra-daily variability and relative amplitude, with the anterior thalamic radiation (ATR), superior longitudinal fasciculus (SLF) and inferior longitudinal fasciculus (ILF) selected as tracts of interest. One hundred and eight participants at risk for cognitive decline (classified as experiencing subjective or objective cognitive decline) were included (mean age = 68.85 years, SD = 8.91). Of the primary non-parametric measures of interest, results showed that lower IS was associated with a greater likelihood of higher WML burden in the ATR (OR = 1.82, 95% CI [1.12,3.15]). Analysis of secondary non-parametric measures revealed later onset of the least active period to be associated with greater likelihood of high WML burden in the SLF (OR = 1.55, 95% CI [1.00,2.53]) and increased activity during the least active 5-h period to be associated with a greater likelihood of high whole-brain WML burden (OR = 1.83, 95% CI [1.06,3.47]). This study shows integrity of the ATR and SLF, and overall WML burden is linked to altered rest-activity rhythms in older adults at risk for cognitive decline, with those demonstrating altered rest-activity rhythms showing 50%-80% higher odds of having high WML burden.
Collapse
Affiliation(s)
- Jake R Palmer
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Department of Psychology, Macquarie University, Sydney, NSW, Australia
| | - Chenyu Wang
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Centre, Sydney, NSW, Australia
| | - Dexiao Kong
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Marcela Cespedes
- Australian e-Health Research Centre, CSIRO Health and Biosecurity, Herston, QLD, Australia
| | - Jonathon Pye
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
- Susan Wakil School of Nursing and Midwifery, The University of Sydney, Sydney, NSW, Australia
| | - Ian B Hickie
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Michael Barnett
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Centre, Sydney, NSW, Australia
| | - Sharon L Naismith
- School of Psychology, The University of Sydney, Sydney, NSW, Australia.
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- NHMRC Centre of Research Excellence to Optimise Sleep in Brain Ageing and Neurodegeneration, Sydney, NSW, Australia.
| |
Collapse
|
46
|
Zuroff L, Wisse LEM, Glenn T, Xie SX, Nasrallah IM, Habes M, Dubroff J, de Flores R, Xie L, Yushkevich P, Doshi J, Davatsikos C, Shaw LM, Tropea TF, Chen-Plotkin AS, Wolk DA, Das S, Mechanic-Hamilton D. Self- and Partner-Reported Subjective Memory Complaints: Association with Objective Cognitive Impairment and Risk of Decline. J Alzheimers Dis Rep 2022; 6:411-430. [PMID: 36072364 PMCID: PMC9397901 DOI: 10.3233/adr-220013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2022] [Indexed: 11/15/2022] Open
Abstract
Background Episodic memory decline is a hallmark of Alzheimer's disease (AD). Subjective memory complaints (SMCs) may represent one of the earliest signs of impending cognitive decline. The degree to which self- or partner-reported SMCs predict cognitive change remains unclear. Objective We aimed to evaluate the relationship between self- and partner-reported SMCs, objective cognitive performance, AD biomarkers, and risk of future decline in a well-characterized longitudinal memory center cohort. We also evaluated whether study partner characteristics influence reports of SMCs. Methods 758 participants and 690 study partners were recruited from the Penn Alzheimer's Disease Research Center Clinical Core. Participants included those with Normal Cognition, Mild Cognitive Impairment, and AD. SMCs were measured using the Prospective and Retrospective Memory Questionnaire (PRMQ), and were evaluated for their association with cognition, genetic, plasma, and neuroimaging biomarkers of AD, cognitive and functional decline, and diagnostic progression over an average of four years. Results We found that partner-reported SMCs were more consistent with cognitive test performance and increasing symptom severity than self-reported SMCs. Partner-reported SMCs showed stronger correlations with AD-associated brain atrophy, plasma biomarkers of neurodegeneration, and longitudinal cognitive and functional decline. A 10-point increase on baseline PRMQ increased the annual risk of diagnostic progression by approximately 70%. Study partner demographics and relationship to participants influenced reports of SMCs in AD participants only. Conclusion Partner-reported SMCs, using the PRMQ, have a stronger relationship with the neuroanatomic and cognitive changes associated with AD than patient-reported SMCs. Further work is needed to evaluate whether SMCs could be used to screen for future decline.
Collapse
Affiliation(s)
- Leah Zuroff
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura EM Wisse
- Department of Diagnostic Radiology, Lund University, Lund, Sweden
| | - Trevor Glenn
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sharon X. Xie
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M. Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), San Antonio, TX, USA
| | - Jacob Dubroff
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Robin de Flores
- Université de Caen Normandie, INSERM UMRS U1237, Caen, France
| | - Long Xie
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Yushkevich
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jimit Doshi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatsikos
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas F. Tropea
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alice S. Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sandhitsu Das
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dawn Mechanic-Hamilton
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
47
|
Lee HA, Kyeong S, Kim DH. Long-term effects of defoliant exposure on brain atrophy progression in humans. Neurotoxicology 2022; 92:25-32. [PMID: 35830900 DOI: 10.1016/j.neuro.2022.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/30/2022] [Accepted: 07/08/2022] [Indexed: 10/17/2022]
Abstract
As the most toxic dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin is classified as a group 1 human carcinogen. We investigated the long-term effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure on the progression of brain atrophy in humans. We retrospectively selected 546 patients exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (exposed group) and 1353 patients not exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (control group). The patients in both groups underwent brain T1-weighted magnetic resonance imaging (MRI) twice. We divided the patients into two propensity score-matched groups, analyzed voxel-wise whole brain atrophy in the MRI images of each patient, and compared the progression of brain atrophy between the two groups. The exposed group showed significant brain atrophy progression in the bilateral frontal and temporal lobes, compared with the control group. The ventrolateral prefrontal area in the frontal lobe and whole temporal lobe were the main atrophic regions in the exposed group, compared with the control group. The neurotoxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin can damage the brain, even in patients exposed to it over 40 years ago. Humans exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin should thus be evaluated for progression of brain atrophy.
Collapse
Affiliation(s)
- Hyun Ah Lee
- Department of Physical Medicine and Rehabilitation, Veterans Health Service Medical Center, Seoul, South Korea
| | - Sohyon Kyeong
- Department of Physical Medicine and Rehabilitation, Veterans Health Service Medical Center, Seoul, South Korea
| | - Dae Hyun Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
48
|
Mori S, Onda K, Fujita S, Suzuki T, Ikeda M, Zay Yar Myint K, Hikage J, Abe O, Tomimoto H, Oishi K, Taguchi J. Brain atrophy in middle age using magnetic resonance imaging scans from Japan’s health screening programme. Brain Commun 2022; 4:fcac211. [PMID: 36043138 PMCID: PMC9416065 DOI: 10.1093/braincomms/fcac211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 06/12/2022] [Accepted: 08/20/2022] [Indexed: 12/21/2022] Open
Abstract
Although health screening plays a key role in the management of chronic diseases associated with lifestyle choices, brain health is not generally monitored, remaining a black box prior to the manifestation of clinical symptoms. Japan is unique in this regard, as brain MRI scans have been widely performed for more than two decades as part of Brain Dock, a comprehensive health screening programme. A vast number of stored images (well over a million) of longitudinal scans and extensive health data are available, offering a valuable resource for investigating the prevalence of various types of brain-related health conditions occurring throughout adulthood. In this paper, we report on the findings of our preliminary quantitative analysis of T1-weighted MRIs of the brain obtained from 13 980 subjects from three participating sites during the period 2015–19. We applied automated segmentation analysis and observed age-dependent volume loss of various brain structures. We subsequently investigated the effects of scan protocols and the feasibility of calibration for pooling the data. Last, the degree of brain atrophy was correlated with four known risk factors of dementia; blood glucose level, hypertension, obesity, and alcohol consumption. In this initial analysis, we identified brain ventricular volume as an effective marker of age-dependent brain atrophy, being highly sensitive to ageing and evidencing strong robustness against protocol variability. We established the normal range of ventricular volumes at each age, which is an essential first step for establishing criteria used to interpret data obtained for individual participants. We identified a subgroup of individuals at midlife with ventricles that substantially exceeded the average size. The correlation studies revealed that all four risk factors were associated with greater ventricular volumes at midlife, some of which reached highly significant sizes. This study demonstrates the feasibility of conducting a large-scale quantitative analysis of existing Brain Dock data in Japan. It will importantly guide future efforts to investigate the prevalence of large ventricles at midlife and the potential reduction of this prevalence, and hence of dementia risk, through lifestyle changes.
Collapse
Affiliation(s)
- Susumu Mori
- Department of Radiology, Johns Hopkins University, School of Medicine , 330 Traylor Bldg, 217 Rutland Ave, Baltimore, MD 21205 , USA
| | - Kengo Onda
- Tokyo Medical and Dental University , 1 Chome-5-45 Yushima, Bunkyo City, Tokyo 113-0034 , Japan
| | - Shohei Fujita
- Department of Radiology, The University of Tokyo, Graduate School of Medicine , 7-3-1 Hongo, Bunkyo City, Tokyo 113-0033 , Japan
| | - Toshiaki Suzuki
- Resorttrust.Inc, Engyou Bldg.8F , Roppongi 7-15-14, Minato-ku, Tokyo 106-0032 , Japan
| | - Mikimasa Ikeda
- Resorttrust.Inc, Engyou Bldg.8F , Roppongi 7-15-14, Minato-ku, Tokyo 106-0032 , Japan
| | - Khin Zay Yar Myint
- Advanced Medical Care Inc. , Midtown Tower 6F, Akasaka 9-7-1, Minato-ku, Tokyo 107-6206 , Japan
| | - Jun Hikage
- Resorttrust.Inc, Engyou Bldg.8F , Roppongi 7-15-14, Minato-ku, Tokyo 106-0032 , Japan
| | - Osamu Abe
- Department of Radiology, The University of Tokyo, Graduate School of Medicine , 7-3-1 Hongo, Bunkyo City, Tokyo 113-0033 , Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Hidekazu Tomimoto, Mie University 2-174 , Edobashi, Tsu, Mie 514-0001 , Japan
| | - Kenichi Oishi
- Department of Radiology, Johns Hopkins University, School of Medicine , 330 Traylor Bldg, 217 Rutland Ave, Baltimore, MD 21205 , USA
| | - Junichi Taguchi
- Tokyo Midtown Clinic , 9-7-1-6F Akasaka, Minato, Tokyo 107-6206 , Japan
| |
Collapse
|
49
|
Toledo JB, Liu H, Grothe MJ, Rashid T, Launer L, Shaw LM, Snoussi H, Heckbert S, Weiner M, Trojanwoski JQ, Seshadri S, Habes M. Disentangling tau and brain atrophy cluster heterogeneity across the Alzheimer's disease continuum. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12305. [PMID: 35619830 PMCID: PMC9127251 DOI: 10.1002/trc2.12305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 12/01/2022]
Abstract
Introduction Neuroimaging heterogeneity in dementia has been examined using single modalities. We evaluated the associations of magnetic resonance imaging (MRI) atrophy and flortaucipir positron emission tomography (PET) clusters across the Alzheimer's disease (AD) spectrum. Methods We included 496 Alzheimer's Disease Neuroimaging Initiative participants with brain MRI, flortaucipir PET scan, and amyloid beta biomarker measures obtained. We applied a novel robust collaborative clustering (RCC) approach on the MRI and flortaucipir PET scans. We derived indices for AD-like (SPARE-AD index) and brain age (SPARE-BA) atrophy. Results We identified four tau (I-IV) and three atrophy clusters. Tau clusters were associated with the apolipoprotein E genotype. Atrophy clusters were associated with white matter hyperintensity volumes. Only the hippocampal sparing atrophy cluster showed a specific association with brain aging imaging index. Tau clusters presented stronger clinical associations than atrophy clusters. Tau and atrophy clusters were partially associated. Conclusions Each neuroimaging modality captured different aspects of brain aging, genetics, vascular changes, and neurodegeneration leading to individual multimodal phenotyping.
Collapse
Affiliation(s)
- Jon B. Toledo
- Department of NeurologyUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Hangfan Liu
- Center for Biomedical Image Computing and Analytics (CBICA)University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Michel J. Grothe
- Unidad de Trastornos del Movimiento, Instituto de Bioedicina de Sevilla (IBiS)Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
| | - Tanweer Rashid
- Center for Biomedical Image Computing and Analytics (CBICA)University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexasUSA
| | - Lenore Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research ProgramNational Institute on AgingBethesdaMarylandUSA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Haykel Snoussi
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexasUSA
| | - Susan Heckbert
- Department of Epidemiology and Cardiovascular Health Research UnitUniversity of WashingtonSeattleWashingtonUSA
- Department of RadiologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of PsychiatryUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Michael Weiner
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Alzheimer's Disease Core Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - John Q. Trojanwoski
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute on AgingPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sudha Seshadri
- Udall Parkinson's Research CenterPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUniversity of Texas Health Sciences CenterSan AntonioTexasUSA
| | - Mohamad Habes
- Center for Biomedical Image Computing and Analytics (CBICA)University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexasUSA
| | | |
Collapse
|
50
|
Hwang G, Abdulkadir A, Erus G, Habes M, Pomponio R, Shou H, Doshi J, Mamourian E, Rashid T, Bilgel M, Fan Y, Sotiras A, Srinivasan D, Morris JC, Albert MS, Bryan NR, Resnick SM, Nasrallah IM, Davatzikos C, Wolk DA. Disentangling Alzheimer's disease neurodegeneration from typical brain ageing using machine learning. Brain Commun 2022; 4:fcac117. [PMID: 35611306 PMCID: PMC9123890 DOI: 10.1093/braincomms/fcac117] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 02/17/2022] [Accepted: 05/04/2022] [Indexed: 11/17/2022] Open
Abstract
Neuroimaging biomarkers that distinguish between changes due to typical brain ageing and Alzheimer's disease are valuable for determining how much each contributes to cognitive decline. Supervised machine learning models can derive multivariate patterns of brain change related to the two processes, including the Spatial Patterns of Atrophy for Recognition of Alzheimer's Disease (SPARE-AD) and of Brain Aging (SPARE-BA) scores investigated herein. However, the substantial overlap between brain regions affected in the two processes confounds measuring them independently. We present a methodology, and associated results, towards disentangling the two. T1-weighted MRI scans of 4054 participants (48-95 years) with Alzheimer's disease, mild cognitive impairment (MCI), or cognitively normal (CN) diagnoses from the Imaging-based coordinate SysTem for AGIng and NeurodeGenerative diseases (iSTAGING) consortium were analysed. Multiple sets of SPARE scores were investigated, in order to probe imaging signatures of certain clinically or molecularly defined sub-cohorts. First, a subset of clinical Alzheimer's disease patients (n = 718) and age- and sex-matched CN adults (n = 718) were selected based purely on clinical diagnoses to train SPARE-BA1 (regression of age using CN individuals) and SPARE-AD1 (classification of CN versus Alzheimer's disease) models. Second, analogous groups were selected based on clinical and molecular markers to train SPARE-BA2 and SPARE-AD2 models: amyloid-positive Alzheimer's disease continuum group (n = 718; consisting of amyloid-positive Alzheimer's disease, amyloid-positive MCI, amyloid- and tau-positive CN individuals) and amyloid-negative CN group (n = 718). Finally, the combined group of the Alzheimer's disease continuum and amyloid-negative CN individuals was used to train SPARE-BA3 model, with the intention to estimate brain age regardless of Alzheimer's disease-related brain changes. The disentangled SPARE models, SPARE-AD2 and SPARE-BA3, derived brain patterns that were more specific to the two types of brain changes. The correlation between the SPARE-BA Gap (SPARE-BA minus chronological age) and SPARE-AD was significantly reduced after the decoupling (r = 0.56-0.06). The correlation of disentangled SPARE-AD was non-inferior to amyloid- and tau-related measurements and to the number of APOE ε4 alleles but was lower to Alzheimer's disease-related psychometric test scores, suggesting the contribution of advanced brain ageing to the latter. The disentangled SPARE-BA was consistently less correlated with Alzheimer's disease-related clinical, molecular and genetic variables. By employing conservative molecular diagnoses and introducing Alzheimer's disease continuum cases to the SPARE-BA model training, we achieved more dissociable neuroanatomical biomarkers of typical brain ageing and Alzheimer's disease.
Collapse
Affiliation(s)
- Gyujoon Hwang
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Raymond Pomponio
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jimit Doshi
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Tanweer Rashid
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Yong Fan
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Aristeidis Sotiras
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Dhivya Srinivasan
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - John C. Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nick R. Bryan
- Department of Diagnostic Medicine, University of Texas, Austin, TX, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Ilya M. Nasrallah
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - David A. Wolk
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology and Penn Memory Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|